1
|
Jiménez-Jiménez FJ, Alonso-Navarro H, Salgado-Cámara P, García-Martín E, Agúndez JAG. Antioxidant Therapies in the Treatment of Multiple Sclerosis. Biomolecules 2024; 14:1266. [PMID: 39456199 PMCID: PMC11506420 DOI: 10.3390/biom14101266] [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: 08/06/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Several studies have proposed a potential role for oxidative stress in the development of multiple sclerosis (MS). For this reason, it seems tentative to think that treatment with antioxidant substances could be useful in the treatment of this disease. In this narrative review, we provide a summary of the current findings on antioxidant treatments, both in experimental models of MS, especially in experimental autoimmune encephalomyelitis (EAE) and in the cuprizone-induced demyelination model, and clinical trials in patients diagnosed with MS. Practically all the antioxidants tested in experimental models of MS have shown improvement in clinical parameters, in delaying the evolution of the disease, and in improving histological and biochemical parameters, including decreased levels of markers of inflammation and oxidative stress in the central nervous system and other tissues. Only a few clinical trials have been carried out to investigate the potential efficacy of antioxidant substances in patients with MS, most of them in the short term and involving a short series of patients, so the results of these should be considered inconclusive. In this regard, it would be desirable to design long-term, randomized, multicenter clinical trials with a long series of patients, assessing several antioxidants that have demonstrated efficacy in experimental models of MS.
Collapse
Grants
- PI18/00540 Fondo de Investigación Sanitaria, Instituto de Salud Carlos, Madrid, Spain
- PI21/01683 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Madrid, Spain
- IB20134 Junta de Extremadura, Mérida, Spain
- GR21073 Junta de Extremadura, Mérida, Spain
Collapse
Affiliation(s)
| | - Hortensia Alonso-Navarro
- Section of Neurology, Hospital Universitario del Sureste, E28500 Arganda del Rey, Spain; (H.A.-N.); (P.S.-C.)
| | - Paula Salgado-Cámara
- Section of Neurology, Hospital Universitario del Sureste, E28500 Arganda del Rey, Spain; (H.A.-N.); (P.S.-C.)
| | - Elena García-Martín
- University Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, E10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| | - José A. G. Agúndez
- University Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, E10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| |
Collapse
|
2
|
Muñoz-Jurado A, Escribano BM, Túnez I. Animal model of multiple sclerosis: Experimental autoimmune encephalomyelitis. Methods Cell Biol 2024; 188:35-60. [PMID: 38880527 DOI: 10.1016/bs.mcb.2024.03.013] [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] [Indexed: 06/18/2024]
Abstract
Multiple sclerosis (MS) is a very complex and heterogeneous disease, with an unknown etiology and which, currently, remains incurable. For this reason, animal models are crucial to investigate this disease, which has increased in prevalence in recent years, affecting 2.8 million people worldwide, and is the leading cause of non-traumatic disability in young adults between the ages of 20-30years. Of all the models developed to replicate MS, experimental autoimmune encephalomyelitis (EAE) best reflects the autoimmune pathogenesis of MS. There are different methods to induce it, which will give rise to different types of EAE, which will vary in clinical presentation and severity. Of the EAE models, the most widespread and used is the one induced in rodents due to its advantages over other species. Likewise, EAE has become a widely used model in the development of therapies for the treatment of MS. Likewise, it is very useful to define the cellular and molecular mechanisms involved in the pathogenesis of MS and to establish therapeutic targets for this disease. For all these reasons, the EAE model plays a key role in improving the understanding of MS.
Collapse
Affiliation(s)
- Ana Muñoz-Jurado
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Cordoba, Spain; Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain.
| | - Begoña M Escribano
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Cordoba, Spain; Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain
| | - Isaac Túnez
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain.
| |
Collapse
|
3
|
Wen J, Sackett S, Tanaka M, Zhang Y. Therapeutic Effects of Combined Treatment with the AEA Hydrolysis Inhibitor PF04457845 and the Substrate Selective COX-2 Inhibitor LM4131 in the Mouse Model of Neuropathic Pain. Cells 2023; 12:cells12091275. [PMID: 37174675 PMCID: PMC10177584 DOI: 10.3390/cells12091275] [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: 03/15/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Chronic neuropathic pain resulting from peripheral nerve damage is a significant clinical problem, which makes it imperative to develop the mechanism-based therapeutic approaches. Enhancement of endogenous cannabinoids by blocking their hydrolysis has been shown to reduce inflammation and neuronal damage in a number of neurological disorders and neurodegenerative diseases. However, recent studies suggest that inhibition of their hydrolysis can shift endocannabinoids 2-arachidonoyl glycerol (2-AG) and anandamide (AEA) toward the oxygenation pathway mediated by cyclooxygenase-2 (COX-2) to produce proinflammatory prostaglandin glycerol esters (PG-Gs) and prostaglandin ethanolamides (PG-EAs). Thus, blocking both endocannabinoid hydrolysis and oxygenation is likely to be more clinically beneficial. In this study, we used the chronic constriction injury (CCI) mouse model to explore the therapeutic effects of simultaneous inhibition of AEA hydrolysis and oxygenation in the treatment of neuropathic pain. We found that the fatty acid amide hydrolase (FAAH) inhibitor PF04457845 and the substrate-selective COX-2 inhibitor LM4131 dose-dependently reduced thermal hyperalgesia and mechanical allodynia in the CCI mice. In addition to ameliorating the pain behaviors, combined treatment with subeffective doses of these inhibitors greatly attenuated the accumulation of inflammatory cells in both sciatic nerve and spinal cord. Consistently, the increased proinflammatory cytokines IL-1β, IL-6, and chemokine MCP-1 in the CCI mouse spinal cord and sciatic nerve were also significantly reduced by combination of low doses of PF04457845 and LM4131 treatment. Therefore, our study suggests that simultaneous blockage of endocannabinoid hydrolysis and oxygenation by using the substrate-selective COX-2 inhibitor, which avoids the cardiovascular and gastrointestinal side effects associated with the use of general COX-2 inhibitors, might be a suitable strategy for the treatment of inflammatory and neuropathic pain.
Collapse
Affiliation(s)
- Jie Wen
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Scott Sackett
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Mikiei Tanaka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
4
|
Xu Z, Lu S, Liu X, Tang L, Liu Z, Cui J, Wang W, Lu W, Huang J. Drug repurposing of ilepcimide that ameliorates experimental autoimmune encephalomyelitis via restricting inflammatory response and oxidative stress. Toxicol Appl Pharmacol 2023; 458:116328. [PMID: 36455640 DOI: 10.1016/j.taap.2022.116328] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS) that remains incurable. Herein, we demonstrated that ilepcimide (Antiepilepsirine), an antiepileptic drug used for decades, protects mice from experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. Our studies found that ilepcimide treatment effectively ameliorates demyelination, blood-brain barrier leakage and infiltration of CD4+ and CD8+ T cells in EAE mice. On the one hand, ilepcimide can inhibit dihydroorotate dehydrogenase (DHODH), an important therapeutic target for MS. Computer molecular docking, thermal shift and fluorescence quenching assay demonstrated the directly interaction between ilepcimide and DHODH. Accordingly, ilepcimide observably repressed T cell proliferation in mixed lymphocyte reaction (MLR) assay and concanavalin A (Con-A) model in a DHODH-dependent manner. On the other hand, ilepcimide exhibited neuroprotective effect possibly through activating NRF2 antioxidant pathway in mouse neural crest-derived Neuro2a cells. Collectively, our findings have revealed the therapeutic potential of ilepcimide in EAE mouse model via restricting inflammatory response and oxidative stress, offering a potential opportunity for repurposing existing drug ilepcimide for MS therapy.
Collapse
Affiliation(s)
- Zhaomin Xu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Sisi Lu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xi Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Lu Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zehui Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiayan Cui
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wanyan Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Jin Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
5
|
Zhang Y, Song S, Li H, Wang X, Song L, Xue J. Polysaccharide from Ganoderma lucidum alleviates cognitive impairment in a mouse model of chronic cerebral hypoperfusion by regulating CD4 +CD25 +Foxp3 + regulatory T cells. Food Funct 2022; 13:1941-1952. [PMID: 35088782 DOI: 10.1039/d1fo03698j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ganoderma lucidum (G. lucidum) is a kind of edible and medicinal mushroom. G. lucidum polysaccharide-1 (GLP-1) is one of the polysaccharides purified from crude GLP. Chronic cerebral hypoperfusion (CCH) as the common pathological basis of various forms of dementia is an important cause of cognitive impairment. In this study, a step-down test was used to evaluate the cognitive ability of CCH mice. Flow cytometry was used to detect the proportion of CD4+CD25+Foxp3+ regulatory T (Foxp3+Treg) cells. ELISA analysis and western blot analysis were used to detect the transforming growth factor-β1 (TGF-β1) and Interleukin-10 (IL-10) levels that Foxp3+Treg cells secreted. Metabolomic analysis based on gas chromatography-mass spectrometry (GC-MS) was used to evaluate the effect of GLP-1 on dysfunctional metabolism caused by inflammation. Results indicate that GLP-1 exhibited an alleviating cognitive impairment effect on CCH mice. The mechanism was related to GLP-1 by increasing Foxp3+Treg cell levels to increase levels of IL-10 and TGF-β1 and regulate abnormal energy metabolism. These findings could provide preliminary results to exploit G. lucidum as a health care product or functional food for the adjuvant therapy of cognitive impairment of CCH.
Collapse
Affiliation(s)
- Yan Zhang
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, PR China.
| | - Shuang Song
- Graduate School, Jilin Institute of Chemical Technology, Jilin 132022, PR China
| | - Haitao Li
- Department of Pathology, Traditional Chinese Medicine Academy of Sciences of Jilin Province, Changchun 130021, PR China
| | - Xinyan Wang
- Graduate School, Jilin Institute of Chemical Technology, Jilin 132022, PR China
| | - Lianlian Song
- Department of Pathology, Traditional Chinese Medicine Academy of Sciences of Jilin Province, Changchun 130021, PR China
| | - Jianfei Xue
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, PR China.
| |
Collapse
|
6
|
Wu Z, Ma H, Liu Z, Zheng L, Yu Z, Cao S, Fang W, Wu L, Li W, Liu G, Huang J, Tang Y. wSDTNBI: a novel network-based inference method for virtual screening. Chem Sci 2022; 13:1060-1079. [PMID: 35211272 PMCID: PMC8790893 DOI: 10.1039/d1sc05613a] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/15/2021] [Indexed: 12/21/2022] Open
Abstract
In recent years, the rapid development of network-based methods for the prediction of drug-target interactions (DTIs) provides an opportunity for the emergence of a new type of virtual screening (VS), namely, network-based VS. Herein, we reported a novel network-based inference method named wSDTNBI. Compared with previous network-based methods that use unweighted DTI networks, wSDTNBI uses weighted DTI networks whose edge weights are correlated with binding affinities. A two-pronged approach based on weighted DTI and drug-substructure association networks was employed to calculate prediction scores. To show the practical value of wSDTNBI, we performed network-based VS on retinoid-related orphan receptor γt (RORγt), and purchased 72 compounds for experimental validation. Seven of the purchased compounds were confirmed to be novel RORγt inverse agonists by in vitro experiments, including ursonic acid and oleanonic acid with IC50 values of 10 nM and 0.28 μM, respectively. Moreover, the direct contact between ursonic acid and RORγt was confirmed using the X-ray crystal structure, and in vivo experiments demonstrated that ursonic acid and oleanonic acid have therapeutic effects on multiple sclerosis. These results indicate that wSDTNBI might be a powerful tool for network-based VS in drug discovery.
Collapse
Affiliation(s)
- Zengrui Wu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Hui Ma
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Zehui Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Lulu Zheng
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Zhuohang Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Shuying Cao
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Wenqing Fang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Lili Wu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Jin Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| |
Collapse
|
7
|
Schumann L, Wilken-Schmitz A, Trautmann S, Vogel A, Schreiber Y, Hahnefeld L, Gurke R, Geisslinger G, Tegeder I. Increased Fat Taste Preference in Progranulin-Deficient Mice. Nutrients 2021; 13:4125. [PMID: 34836380 PMCID: PMC8623710 DOI: 10.3390/nu13114125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/22/2021] [Accepted: 11/12/2021] [Indexed: 11/25/2022] Open
Abstract
Progranulin deficiency in mice is associated with deregulations of the scavenger receptor signaling of CD36/SCARB3 in immune disease models, and CD36 is a dominant receptor in taste bud cells in the tongue and contributes to the sensation of dietary fats. Progranulin-deficient mice (Grn-/-) are moderately overweight during middle age. We therefore asked if there was a connection between progranulin/CD36 in the tongue and fat taste preferences. By using unbiased behavioral analyses in IntelliCages and Phenomaster cages we showed that progranulin-deficient mice (Grn-/-) developed a strong preference of fat taste in the form of 2% milk over 0.3% milk, and for diluted MCTs versus tap water. The fat preference in the 7d-IntelliCage observation period caused an increase of 10% in the body weight of Grn-/- mice, which did not occur in the wildtype controls. CD36 expression in taste buds was reduced in Grn-/- mice at RNA and histology levels. There were no differences in the plasma or tongue lipids of various classes including sphingolipids, ceramides and endocannabinoids. The data suggest that progranulin deficiency leads to a lower expression of CD36 in the tongue resulting in a stronger urge for fatty taste and fatty nutrition.
Collapse
Affiliation(s)
- Lana Schumann
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Annett Wilken-Schmitz
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Alexandra Vogel
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Yannick Schreiber
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Lisa Hahnefeld
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), 60596 Frankfurt, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| |
Collapse
|
8
|
Schmitz K, Trautmann S, Hahnefeld L, Fischer C, Schreiber Y, Wilken-Schmitz A, Gurke R, Brunkhorst R, Werner ER, Watschinger K, Wicker S, Thomas D, Geisslinger G, Tegeder I. Sapropterin (BH4) Aggravates Autoimmune Encephalomyelitis in Mice. Neurotherapeutics 2021; 18:1862-1879. [PMID: 33844153 PMCID: PMC8609075 DOI: 10.1007/s13311-021-01043-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2021] [Indexed: 02/04/2023] Open
Abstract
Depletion of the enzyme cofactor, tetrahydrobiopterin (BH4), in T-cells was shown to prevent their proliferation upon receptor stimulation in models of allergic inflammation in mice, suggesting that BH4 drives autoimmunity. Hence, the clinically available BH4 drug (sapropterin) might increase the risk of autoimmune diseases. The present study assessed the implications for multiple sclerosis (MS) as an exemplary CNS autoimmune disease. Plasma levels of biopterin were persistently low in MS patients and tended to be lower with high Expanded Disability Status Scale (EDSS). Instead, the bypass product, neopterin, was increased. The deregulation suggested that BH4 replenishment might further drive the immune response or beneficially restore the BH4 balances. To answer this question, mice were treated with sapropterin in immunization-evoked autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. Sapropterin-treated mice had higher EAE disease scores associated with higher numbers of T-cells infiltrating the spinal cord, but normal T-cell subpopulations in spleen and blood. Mechanistically, sapropterin treatment was associated with increased plasma levels of long-chain ceramides and low levels of the poly-unsaturated fatty acid, linolenic acid (FA18:3). These lipid changes are known to contribute to disruptions of the blood-brain barrier in EAE mice. Indeed, RNA data analyses revealed upregulations of genes involved in ceramide synthesis in brain endothelial cells of EAE mice (LASS6/CERS6, LASS3/CERS3, UGCG, ELOVL6, and ELOVL4). The results support the view that BH4 fortifies autoimmune CNS disease, mechanistically involving lipid deregulations that are known to contribute to the EAE pathology.
Collapse
Affiliation(s)
- Katja Schmitz
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Lisa Hahnefeld
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Caroline Fischer
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Yannick Schreiber
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
| | - Annett Wilken-Schmitz
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
| | - Robert Brunkhorst
- Department of Clinical Neurology, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Ernst R Werner
- Institute of Biological Chemistry, Medical University of Innsbruck, Biocenter, Austria
| | - Katrin Watschinger
- Institute of Biological Chemistry, Medical University of Innsbruck, Biocenter, Austria
| | - Sabine Wicker
- Occupational Health Services, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases, Frankfurt, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, Frankfurt, Germany.
| |
Collapse
|
9
|
Li P, Li H, Shu X, Wu M, Liu J, Hao T, Cui H, Zheng L. Intra-articular delivery of flurbiprofen sustained release thermogel: improved therapeutic outcome of collagenase II-induced rat knee osteoarthritis. Drug Deliv 2021; 27:1034-1043. [PMID: 32627602 PMCID: PMC8216450 DOI: 10.1080/10717544.2020.1787555] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Knee osteoarthritis (OA) is a common degenerative disease. Intra-articular administration of flurbiprofen is frequently employed in clinic to treat OA, while repeated injections are required because of the limited effective duration. To improve therapeutic outcome and prolong the treatment interval, a poly(ε-caprolactone-co-lactide)-b-poly(ethylene glycol)-b-poly(ε-caprolactone-co-lactide) (PCLA-PEG-PCLA) triblock copolymer based flurbiprofen thermosensitive gel for the sustained intra-articular drug delivery was designed in this study. The anti-OA effects of this flurbiprofen thermogel were investigated on collagenase II-induced rat knee OA model by multiple approaches and compared with that of conventional sodium hyaluronate and flurbiprofen injecta. In vitro drug release studies indicated that flurbiprofen was sustained released from the thermosensitive gel for more than three weeks. This sustained drug release system exerted comparable short-term analgesic effects and distinctly improved long-term analgesic efficacy in terms of the increased percentage of the total ipsilateral paw print intensity and the reduced Knee-Bend scores of OA rats. The inflammatory response was attenuated in the samples of flurbiprofen gel treated group by showing decreased IL-1, IL-6, and IL-11 levels in the joint fluid and down-regulated IL-1, IL-6, IL-11, COX-2, TNF-α, and NF-κB/p65 expression in the articular cartilages. The results suggest the suitability of thermosensitive copolymer PCLA-PEG-PCLA for sustained intra-articular effects of flurbiprofen and provide in vivo experimental evidence for potential clinical application of this flurbiprofen delivery system to better management of OA cases.
Collapse
Affiliation(s)
- Peinan Li
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, Dalian, China
| | - Haokun Li
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, Dalian, China
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Moli Wu
- Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jia Liu
- Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Tangna Hao
- Department of Pharmacy, Second Clinical College, Dalian Medical University, Dalian, China
| | - Hongxia Cui
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Lianjie Zheng
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, Dalian, China
| |
Collapse
|
10
|
Hu CF, Wu SP, Lin GJ, Shieh CC, Hsu CS, Chen JW, Chen SH, Hong JS, Chen SJ. Microglial Nox2 Plays a Key Role in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. Front Immunol 2021; 12:638381. [PMID: 33868265 PMCID: PMC8050344 DOI: 10.3389/fimmu.2021.638381] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
While oxidative stress has been linked to multiple sclerosis (MS), the role of superoxide-producing phagocyte NADPH oxidase (Nox2) in central nervous system (CNS) pathogenesis remains unclear. This study investigates the impact of Nox2 gene ablation on pro- and anti-inflammatory cytokine and chemokine production in a mouse experimental autoimmune encephalomyelitis (EAE) model. Nox2 deficiency attenuates EAE-induced neural damage and reduces disease severity, pathogenic immune cells infiltration, demyelination, and oxidative stress in the CNS. The number of autoreactive T cells, myeloid cells, and activated microglia, as well as the production of cytokines and chemokines, including GM-CSF, IFNγ, TNFα, IL-6, IL-10, IL-17A, CCL2, CCL5, and CXCL10, were much lower in the Nox2-/- CNS tissues but remained unaltered in the peripheral lymphoid organs. RNA-seq profiling of microglial transcriptome identified a panel of Nox2 dependent proinflammatory genes: Pf4, Tnfrsf9, Tnfsf12, Tnfsf13, Ccl7, Cxcl3, and Cxcl9. Furthermore, gene ontology and pathway enrichment analyses revealed that microglial Nox2 plays a regulatory role in multiple pathways known to be important for MS/EAE pathogenesis, including STAT3, glutathione, leukotriene biosynthesis, IL-8, HMGB1, NRF2, systemic lupus erythematosus in B cells, and T cell exhaustion signaling. Taken together, our results provide new insights into the critical functions performed by microglial Nox2 during the EAE pathogenesis, suggesting that Nox2 inhibition may represent an important therapeutic target for MS.
Collapse
Affiliation(s)
- Chih-Fen Hu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Gu-Jiun Lin
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Chang Shieh
- Institute of Clinical Medicine, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Chih-Sin Hsu
- Genomics Center for Clinical and Biotechnological Applications of Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jing-Wun Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Heng Chen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Shyi-Jou Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
11
|
Bilirubin nanomedicine ameliorates the progression of experimental autoimmune encephalomyelitis by modulating dendritic cells. J Control Release 2021; 331:74-84. [DOI: 10.1016/j.jconrel.2021.01.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/05/2021] [Accepted: 01/10/2021] [Indexed: 12/14/2022]
|
12
|
Jung F, Thurn M, Krollik K, Li D, Dressman J, Alig E, Fink L, Schmidt MU, Wacker MG. Sustained-release hot melt extrudates of the weak acid TMP-001: A case study using PBB modelling. Eur J Pharm Biopharm 2021; 160:23-34. [PMID: 33484866 DOI: 10.1016/j.ejpb.2021.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 11/27/2022]
Abstract
Over the last 30 years, hot melt extrusion has become a leading technology in the manufacture of amorphous drug delivery systems. Mostly applied as an 'enabling formulation' for poorly soluble compounds, application in the design of sustained-release formulations increasingly attracts the attention of the pharmaceutical industry. The drug candidate TMP-001 is currently under evaluation for the early treatment of Multiple Sclerosis. Although this weak acid falls into class II of the Biopharmaceutics Classification System, the compound exhibits high solubility in the upper intestine resulting in high peroral bioavailability. In the present studies, four different formulation prototypes varying in their sustained-release behavior were developed, using L-arginine as a pore-forming agent in concentrations ranging between 0 and 20%. Initially, biorelevant release testing was applied to assess the dissolution behavior of the prototypes. For these formulations, a total drug release of 44.7%, 64.6%, 75%, and 90.5% was achieved in FaSSIF-v2 after 24 h. Two candidates were selected for further characterization considering the crystal structure and the physical stability of the amorphous state of TMP-001 in the formulations together with the release behavior in Level II biorelevant media. Our findings indicate L-arginine as a valuable excipient in the formulation of hot melt extrudates, as its presence led to a considerable stabilization of the amorphous state and favorably impacted the milling process and release behavior of TMP-001. To properly evaluate the proposed formulations and the importance of colonic dissolution and absorption on the overall bioavailability, a physiologically-based biopharmaceutics model was used.
Collapse
Affiliation(s)
- Fabian Jung
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany; Institute of Pharmaceutical Technology, Goethe University, Max-von-Laue-Straße 9, D-60438 Frankfurt/Main, Germany
| | - Manuela Thurn
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Katharina Krollik
- Institute of Pharmaceutical Technology, Goethe University, Max-von-Laue-Straße 9, D-60438 Frankfurt/Main, Germany
| | - David Li
- Department of Pharmacy, Faculty of Science, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore
| | - Jennifer Dressman
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany; Institute of Pharmaceutical Technology, Goethe University, Max-von-Laue-Straße 9, D-60438 Frankfurt/Main, Germany
| | - Edith Alig
- Institute of Inorganic and Analytical Chemistry, Goethe University, Max-von-Laue-Straße 7, D-60438 Frankfurt/Main, Germany
| | - Lothar Fink
- Institute of Inorganic and Analytical Chemistry, Goethe University, Max-von-Laue-Straße 7, D-60438 Frankfurt/Main, Germany
| | - Martin U Schmidt
- Institute of Inorganic and Analytical Chemistry, Goethe University, Max-von-Laue-Straße 7, D-60438 Frankfurt/Main, Germany
| | - Matthias G Wacker
- Department of Pharmacy, Faculty of Science, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore.
| |
Collapse
|
13
|
Low brain endocannabinoids associated with persistent non-goal directed nighttime hyperactivity after traumatic brain injury in mice. Sci Rep 2020; 10:14929. [PMID: 32913220 PMCID: PMC7483739 DOI: 10.1038/s41598-020-71879-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is a frequent cause of chronic headache, fatigue, insomnia, hyperactivity, memory deficits, irritability and posttraumatic stress disorder. Recent evidence suggests beneficial effects of pro-cannabinoid treatments. We assessed in mice levels of endocannabinoids in association with the occurrence and persistence of comparable sequelae after controlled cortical impact in mice using a set of long-term behavioral observations in IntelliCages, motor and nociception tests in two sequential cohorts of TBI/sham mice. TBI mice maintained lower body weights, and they had persistent low levels of brain ethanolamide endocannabinoids (eCBs: AEA, OEA, PEA) in perilesional and subcortical ipsilateral brain tissue (6 months), but rapidly recovered motor functions (within days), and average nociceptive responses were within normal limits, albeit with high variability, ranging from loss of thermal sensation to hypersensitivity. TBI mice showed persistent non-goal directed nighttime hyperactivity, i.e. they visited rewarding and non-rewarding operant corners with high frequency and random success. On successful visits, they made more licks than sham mice resulting in net over-licking. The lower the eCBs the stronger was the hyperactivity. In reward-based learning and reversal learning tasks, TBI mice were not inferior to sham mice, but avoidance memory was less stable. Hence, the major late behavioral TBI phenotype was non-goal directed nighttime hyperactivity and "over-licking" in association with low ipsilateral brain eCBs. The behavioral phenotype would agree with a "post-TBI hyperactivity disorder". The association with persistently low eCBs in perilesional and subcortical regions suggests that eCB deficiency contribute to the post-TBI psychopathology.
Collapse
|
14
|
Hoxha M, Spahiu E, Prendi E, Zappacosta B. A Systematic Review on the Role of Arachidonic Acid Pathway in Multiple Sclerosis. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 21:160-187. [PMID: 32842948 DOI: 10.2174/1871527319666200825164123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/28/2020] [Accepted: 07/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND & OBJECTIVE Multiple sclerosis (MS) is an inflammatory neurodegenerative disease characterized by destruction of oligodendrocytes, immune cell infiltration and demyelination. Inflammation plays a significant role in MS, and the inflammatory mediators such as eicosanoids, leukotrienes, superoxide radicals are involved in pro-inflammatory responses in MS. In this systematic review we tried to define and discuss all the findings of in vivo animal studies and human clinical trials on the potential association between arachidonic acid (AA) pathway and multiple sclerosis. METHODS A systematic literature search across Pubmed, Scopus, Embase and Cochrane database was conducted. This systematic review was performed according to PRISMA guidelines. RESULTS A total of 146 studies were included, of which 34 were conducted in animals, 58 in humans, and 60 studies reported the role of different compounds that target AA mediators or their corresponding enzymes/ receptors, and can have a therapeutic effect in MS. These results suggest that eicosanoids have significant roles in experimental autoimmune encephalomyelitis (EAE) and MS. The data from animal and human studies elucidated that PGI2, PGF2α, PGD2, isoprostanes, PGE2, PLA2, LTs are increased in MS. PLA2 inhibition modulates the progression of the disease. PGE1 analogues can be a useful option in the treatment of MS. CONCLUSIONS All studies reported the beneficial effects of COX and LOX inhibitors in MS. The hybrid compounds, such as COX-2 inhibitors/TP antagonists and 5-LOX inhibitors can be an innovative approach for multiple sclerosis treatment. Future work in MS should shed light in synthesizing new compounds targeting arachidonic acid pathway.
Collapse
Affiliation(s)
- Malvina Hoxha
- Department of Chemical-Toxicological and Pharmacological Evaluations of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, Rruga Dritan Hoxha, Tirana. Albania
| | | | - Emanuela Prendi
- Catholic University Our Lady of Good Counsel, Department of Biomedical Sciences, Rruga Dritan Hoxha, Tirana. Albania
| | - Bruno Zappacosta
- Department of Chemical-Toxicological and Pharmacological Evaluations of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, Rruga Dritan Hoxha, Tirana. Albania
| |
Collapse
|
15
|
Bailly C, Vergoten G. Flurbiprofen as a biphenyl scaffold for the design of small molecules binding to PD-L1 protein dimer. Biochem Pharmacol 2020; 178:114042. [DOI: 10.1016/j.bcp.2020.114042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 05/15/2020] [Indexed: 12/17/2022]
|
16
|
Liu Z, Hu Q, Wang W, Lu S, Wu D, Ze S, He J, Huang Y, Chen W, Xu Y, Lu W, Huang J. Natural product piperine alleviates experimental allergic encephalomyelitis in mice by targeting dihydroorotate dehydrogenase. Biochem Pharmacol 2020; 177:114000. [PMID: 32353424 DOI: 10.1016/j.bcp.2020.114000] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/24/2020] [Indexed: 01/06/2023]
Abstract
Multiple sclerosis (MS) is the most popular chronic and debilitating inflammatory disease of the central nervous system (CNS) that remains incurable. Dihydroorotate dehydrogenase (DHODH) is critical to the activity of T lymphocytes and represents a potential therapeutic target for MS. Here we identify piperine, a bioactive constituent of black pepper, as a potent inhibitor of DHODH with an IC50 value of 0.88 μM. Isothermal titration calorimetry and thermofluor assay demonstrate the directly interaction between piperine and DHODH. The co-complex crystal structure of DHODH and piperine at 1.98 Å resolution further reveal that Tyr356 residue of DHODH is crucial for piperine binding. Importantly, we show that piperine can inhibit T cell overactivation in a DHODH-dependent manner in concanavalin A-triggered T-cell assay and mixed lymphocyte reaction assay. Finally, piperine exhibits strong preventive and therapeutic effect in the MOG-induced experimental allergic encephalomyelitis (EAE), a useful model for studying potential treatments for MS, by restricting inflammatory cells infiltration into the CNS and preventing myelin destruction and blood-brain barrier (BBB) disruption. Taken together, these findings highlight DHODH as a therapeutic target for autoimmune disease of the nervous system, and demonstrate a novel role for piperine in the treatment of MS.
Collapse
Affiliation(s)
- Zehui Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Qian Hu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wanyan Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Sisi Lu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Dang Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Shuyin Ze
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiacheng He
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ying Huang
- Guangdong Institute for Drug Control, Guangdong, China
| | - Wuyan Chen
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Yechun Xu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
17
|
Celarain N, Tomas-Roig J. Aberrant DNA methylation profile exacerbates inflammation and neurodegeneration in multiple sclerosis patients. J Neuroinflammation 2020; 17:21. [PMID: 31937331 PMCID: PMC6961290 DOI: 10.1186/s12974-019-1667-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune and demyelinating disease of the central nervous system characterised by incoordination, sensory loss, weakness, changes in bladder capacity and bowel function, fatigue and cognitive impairment, creating a significant socioeconomic burden. The pathogenesis of MS involves both genetic susceptibility and exposure to distinct environmental risk factors. The gene x environment interaction is regulated by epigenetic mechanisms. Epigenetics refers to a complex system that modifies gene expression without altering the DNA sequence. The most studied epigenetic mechanism is DNA methylation. This epigenetic mark participates in distinct MS pathophysiological processes, including blood-brain barrier breakdown, inflammatory response, demyelination, remyelination failure and neurodegeneration. In this study, we also accurately summarised a list of environmental factors involved in the MS pathogenesis and its clinical course. A literature search was conducted using MEDLINE through PubMED and Scopus. In conclusion, an exhaustive study of DNA methylation might contribute towards new pharmacological interventions in MS by use of epigenetic drugs.
Collapse
Affiliation(s)
- Naiara Celarain
- Girona Neuroimmunology and Multiple Sclerosis Unit (UNIEM), Dr. Josep Trueta University Hospital and Girona Biomedical Research Institute (IDIBGI), Girona, Spain.
| | - Jordi Tomas-Roig
- Girona Neuroimmunology and Multiple Sclerosis Unit (UNIEM), Dr. Josep Trueta University Hospital and Girona Biomedical Research Institute (IDIBGI), Girona, Spain.
| |
Collapse
|
18
|
Zeng X, Zhu S, Lu W, Liu Z, Huang J, Zhou Y, Fang J, Huang Y, Guo H, Li L, Trapp BD, Nussinov R, Eng C, Loscalzo J, Cheng F. Target identification among known drugs by deep learning from heterogeneous networks. Chem Sci 2020; 11:1775-1797. [PMID: 34123272 PMCID: PMC8150105 DOI: 10.1039/c9sc04336e] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Without foreknowledge of the complete drug target information, development of promising and affordable approaches for effective treatment of human diseases is challenging. Here, we develop deepDTnet, a deep learning methodology for new target identification and drug repurposing in a heterogeneous drug-gene-disease network embedding 15 types of chemical, genomic, phenotypic, and cellular network profiles. Trained on 732 U.S. Food and Drug Administration-approved small molecule drugs, deepDTnet shows high accuracy (the area under the receiver operating characteristic curve = 0.963) in identifying novel molecular targets for known drugs, outperforming previously published state-of-the-art methodologies. We then experimentally validate that deepDTnet-predicted topotecan (an approved topoisomerase inhibitor) is a new, direct inhibitor (IC50 = 0.43 μM) of human retinoic-acid-receptor-related orphan receptor-gamma t (ROR-γt). Furthermore, by specifically targeting ROR-γt, topotecan reveals a potential therapeutic effect in a mouse model of multiple sclerosis. In summary, deepDTnet offers a powerful network-based deep learning methodology for target identification to accelerate drug repurposing and minimize the translational gap in drug development.
Collapse
Affiliation(s)
- Xiangxiang Zeng
- College of Information Science and Engineering, Hunan University Changsha Hunan 410082 China
| | - Siyi Zhu
- Department of Computer Science, Xiamen University Xiamen Fujian 361005 China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University Shanghai 200241 China
| | - Zehui Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237 China
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237 China
| | - Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic 9500 Euclid Avenue Cleveland OH 44106 USA +1-216-6361609 +1-216-4447654
| | - Jiansong Fang
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic 9500 Euclid Avenue Cleveland OH 44106 USA +1-216-6361609 +1-216-4447654
| | - Yin Huang
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic 9500 Euclid Avenue Cleveland OH 44106 USA +1-216-6361609 +1-216-4447654
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University Nanjing 210009 China
| | - Huimin Guo
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University Nanjing 210009 China
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University Columbus OH 43210 USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Cleveland OH 44022 USA
| | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick Frederick MD 21702 USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University Tel Aviv 69978 Israel
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic 9500 Euclid Avenue Cleveland OH 44106 USA +1-216-6361609 +1-216-4447654
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University Cleveland OH 44195 USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
- Taussig Cancer Institute, Cleveland Clinic Cleveland Ohio 44195 USA
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School Boston MA 02115 USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic 9500 Euclid Avenue Cleveland OH 44106 USA +1-216-6361609 +1-216-4447654
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University Cleveland OH 44195 USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| |
Collapse
|
19
|
Botz B, Bátai IZ, Kiss T, Pintér E, Helyes Z, Bölcskei K. The fluorescent dye 3,3'-diethylthiatricarbocyanine iodide is unsuitable for in vivo imaging of myelination in the mouse. Brain Res Bull 2019; 156:10-14. [PMID: 31857136 DOI: 10.1016/j.brainresbull.2019.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 01/27/2023]
Abstract
There is a growing interest to use non-invasive optical imaging methods to study central nervous system diseases. The application of a myelin-binding fluorescent dye, 3,3-diethylthiatricarbocyanine iodide (DBT) was recently described for in vivo optical imaging of demyelination in the mouse. In the present study we aimed at adapting the method to our optical imaging systems, the IVIS Lumina II to measure epifluorescence and the fluorescent molecular tomograph (FMT) for 3-dimensional quantification of the fluorophore. Epifluorescent imaging was performed 5-30 min after DBT injection which was followed by FMT imaging at 40 min. Two mice also underwent micro-CT imaging in the FMT cassette for the purpose of FMT-CT co-registration. Ex vivo imaging of the brain and other tissues of the head and neck was carried out 1 h after injection. Both the FMT-CT co-registration and the ex vivo imaging of organs proved that DBT poorly crossed the blood-brain barrier. The dye did not accumulate in the myelin sheath of the sciatic nerve. In contrast, there was an intense accumulation in the pituitary and salivary glands. The FMT-CT co-registration unequivocally demonstrated that the signal localized to the head did not originate from beyond the blood-brain barrier. No myelin binding was demonstrated by the ex vivo imaging either. In conclusion, DBT is unsuitable for in vivo imaging of myelination due to its poor BBB penetration, accumulation in other structures of the head and neck region and lack of selective binding towards myelin in vivo.
Collapse
Affiliation(s)
- Bálint Botz
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, H-7624 Pécs, Ifjúság út 20, Hungary; Department of Radiology, University of Pécs Medical School, H-7624 Pécs, Ifjúság út 13, Hungary
| | - István Zoárd Bátai
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, H-7624 Pécs, Ifjúság út 20, Hungary; Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Szigeti út 12, Hungary
| | - Tamás Kiss
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, H-7624 Pécs, Ifjúság út 20, Hungary
| | - Erika Pintér
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, H-7624 Pécs, Ifjúság út 20, Hungary; Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Szigeti út 12, Hungary
| | - Zsuzsanna Helyes
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, H-7624 Pécs, Ifjúság út 20, Hungary; Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Szigeti út 12, Hungary
| | - Kata Bölcskei
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, H-7624 Pécs, Ifjúság út 20, Hungary; Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Szigeti út 12, Hungary.
| |
Collapse
|
20
|
Zyla K, Larabee CM, Georgescu C, Berkley C, Reyna T, Plafker SM. Dimethyl fumarate mitigates optic neuritis. Mol Vis 2019; 25:446-461. [PMID: 31523122 PMCID: PMC6707756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 08/20/2019] [Indexed: 11/07/2022] Open
Abstract
Purpose Dimethyl fumarate (DMF) has been approved by the U.S. Food and Drug Administration (FDA) for the treatment of relapsing-remitting multiple sclerosis (RRMS), a demyelinating autoimmune disease characterized by acute episodes of motor, sensory, and cognitive symptoms. Optic neuritis is an episodic sequela experienced by some patients with RRMS that typically presents as acute, monocular vision loss. Episodes of optic neuritis damage and kill retinal ganglion cells (RGCs), and can culminate in permanent vision loss. The purpose of these studies was to evaluate the capacity of DMF to mitigate optic neuritis. The work presented combines studies of a mouse model of MS and a retrospective chart analysis of files of patients with RRMS treated at the MS Center of Excellence within the Oklahoma Medical Research Foundation. Methods Experimental autoimmune encephalomyelitis (EAE) is a well-established mouse model that recapitulates cardinal features of somatic and visual MS pathologies. EAE was induced in female C57BL/6J mice by inoculation with myelin oligodendrocyte glycoprotein peptide (residues 35-55; MOG35-55). DMF or vehicle was administered twice a day by oral gavage. Visual acuity was measured longitudinally with optokinetic tracking. Post-mortem analyses included quantification of RGCs in retinal flatmounts and quantitative PCR (qPCR) of Nrf2 target genes and regulators of myelin. Retrospective chart analyses were performed using data obtained from deidentified files of patients with RRMS. Results In the EAE mouse studies, DMF decreased optic neuritis severity, preserved vision and RGCs, and concomitantly reduced motor deficits when administered by two different treatment regimens (prevention or interventional). DMF was more efficacious when administered as an interventional therapy, and the beneficial effects occurred independently of the induction of Nrf2 target genes. A complementary retrospective chart analysis demonstrated that DMF increased the time to a recurrence of optic neuritis, and protected against subsequent bouts of optic neuritis. Conclusions This work underscores the potential of DMF to mitigate the severity and recurrence of optic neuritis episodes in patients with RRMS.
Collapse
Affiliation(s)
- Katarzyna Zyla
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Chelsea M. Larabee
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Constantin Georgescu
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Chelsea Berkley
- The Oklahoma Medical Research Foundation Multiple Sclerosis Center of Excellence, Oklahoma City, OK
| | - Tania Reyna
- The Oklahoma Medical Research Foundation Multiple Sclerosis Center of Excellence, Oklahoma City, OK
| | - Scott M. Plafker
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
21
|
Sen MK, Mahns DA, Coorssen JR, Shortland PJ. Behavioural phenotypes in the cuprizone model of central nervous system demyelination. Neurosci Biobehav Rev 2019; 107:23-46. [PMID: 31442519 DOI: 10.1016/j.neubiorev.2019.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/01/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022]
Abstract
The feeding of cuprizone (CPZ) to animals has been extensively used to model the processes of demyelination and remyelination, with many papers adopting a narrative linked to demyelinating conditions like multiple sclerosis (MS), the aetiology of which is unknown. However, no current animal model faithfully replicates the myriad of symptoms seen in the clinical condition of MS. CPZ ingestion causes mitochondrial and endoplasmic reticulum stress and subsequent apoptosis of oligodendrocytes leads to central nervous system demyelination and glial cell activation. Although there are a wide variety of behavioural tests available for characterizing the functional deficits in animal models of disease, including that of CPZ-induced deficits, they have focused on a narrow subset of outcomes such as motor performance, cognition, and anxiety. The literature has not been systematically reviewed in relation to these or other symptoms associated with clinical MS. This paper reviews these tests and makes recommendations as to which are the most important in order to better understand the role of this model in examining aspects of demyelinating diseases like MS.
Collapse
Affiliation(s)
- Monokesh K Sen
- School of Medicine, Western Sydney University, New South Wales, Australia
| | - David A Mahns
- School of Medicine, Western Sydney University, New South Wales, Australia
| | - Jens R Coorssen
- Departments of Health Sciences and Biological Sciences, Faculties of Applied Health Sciences and Mathematics & Science, Brock University, Ontario, Canada.
| | - Peter J Shortland
- Science and Health, Western Sydney University, New South Wales, Australia.
| |
Collapse
|
22
|
Dual Actions of Ketorolac in Metastatic Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11081049. [PMID: 31344967 PMCID: PMC6721416 DOI: 10.3390/cancers11081049] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022] Open
Abstract
Cytoreductive surgery and chemotherapy are cornerstones of ovarian cancer treatment, yet disease recurrence remains a significant clinical issue. Surgery can release cancer cells into the circulation, suppress anti-tumor immunity, and induce inflammatory responses that support the growth of residual disease. Intervention within the peri-operative window is an under-explored opportunity to mitigate these consequences of surgery and influence the course of metastatic disease to improve patient outcomes. One drug associated with improved survival in cancer patients is ketorolac. Ketorolac is a chiral molecule administered as a 1:1 racemic mixture of the S- and R-enantiomers. The S-enantiomer is considered the active component for its FDA indication in pain management with selective activity against cyclooxygenase (COX) enzymes. The R-enantiomer has a previously unrecognized activity as an inhibitor of Rac1 (Ras-related C3 botulinum toxin substrate) and Cdc42 (cell division control protein 42) GTPases. Therefore, ketorolac differs from other non-steroidal anti-inflammatory drugs (NSAIDs) by functioning as two distinct pharmacologic entities due to the independent actions of each enantiomer. In this review, we summarize evidence supporting the benefits of ketorolac administration for ovarian cancer patients. We also discuss how simultaneous inhibition of these two distinct classes of targets, COX enzymes and Rac1/Cdc42, by S-ketorolac and R-ketorolac respectively, could each contribute to anti-cancer activity.
Collapse
|
23
|
Davis MP, Behm B, Mehta Z, Fernandez C. The Potential Benefits of Palmitoylethanolamide in Palliation: A Qualitative Systematic Review. Am J Hosp Palliat Care 2019; 36:1134-1154. [PMID: 31113223 DOI: 10.1177/1049909119850807] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Palmitoylethanolamide (PEA) is a nutraceutical endocannabinoid that was retrospectively discovered in egg yolks. Feeding poor children with known streptococcal infections prevented rheumatic fever. Subsequently, it was found to alter the course of influenza. Unfortunately, there is little known about its pharmacokinetics. Palmitoylethanolamide targets nonclassical cannabinoid receptors rather than CB1 and CB2 receptors. Palmitoylethanolamide will only indirectly activate classical cannabinoid receptors by an entourage effect. There are a significant number of prospective and randomized trials demonstrating the pain-relieving effects of PEA. There is lesser evidence of benefit in patients with nonpain symptoms related to depression, Parkinson disease, strokes, and autism. There are no reported drug-drug interactions and very few reported adverse effects from PEA. Further research is needed to define the palliative benefits to PEA.
Collapse
|
24
|
Nicoletti F, Mazzon E, Fagone P, Mangano K, Mammana S, Cavalli E, Basile MS, Bramanti P, Scalabrino G, Lange A, Curtin F. Prevention of clinical and histological signs of MOG-induced experimental allergic encephalomyelitis by prolonged treatment with recombinant human EGF. J Neuroimmunol 2019; 332:224-232. [PMID: 31100693 DOI: 10.1016/j.jneuroim.2019.05.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022]
Abstract
Epidermal growth factor (EGF) represents the prototype of the group I EGF family. The pleiotropic effects of the EGF have attracted attention to the possibility that it could be implicated in autoimmune diseases, such as Multiple Sclerosis (MS). We show here that treatment with EGF, as a late prophylactic regime, improved the clinical and histological features of EAE, a preclinical model of MS. In silico analysis further corroborated these findings by demonstrating that EGF receptors are less expressed in CNS from patients with MS as compared to controls. Taken together these data provide clear-cut in vivo proof of concept for a beneficial role of exogenously administered EGF in MS, that may, therefore, represent a novel therapeutic approach.
Collapse
Affiliation(s)
- Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy.
| | | | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - Santa Mammana
- IRCCS Centro Neurolesi 'Bonino-Pulejo', Messina, Italy
| | | | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | | | - Giuseppe Scalabrino
- Department of Biomedical Sciences, Laboratory of Neuropathology, University of Milan, Italy
| | - Alois Lange
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - Francois Curtin
- Division of Clinical Pharmacology and Toxicology, University of Geneva, Switzerland
| |
Collapse
|
25
|
Schmidleithner L, Thabet Y, Schönfeld E, Köhne M, Sommer D, Abdullah Z, Sadlon T, Osei-Sarpong C, Subbaramaiah K, Copperi F, Haendler K, Varga T, Schanz O, Bourry S, Bassler K, Krebs W, Peters AE, Baumgart AK, Schneeweiss M, Klee K, Schmidt SV, Nüssing S, Sander J, Ohkura N, Waha A, Sparwasser T, Wunderlich FT, Förster I, Ulas T, Weighardt H, Sakaguchi S, Pfeifer A, Blüher M, Dannenberg AJ, Ferreirós N, Muglia LJ, Wickenhauser C, Barry SC, Schultze JL, Beyer M. Enzymatic Activity of HPGD in Treg Cells Suppresses Tconv Cells to Maintain Adipose Tissue Homeostasis and Prevent Metabolic Dysfunction. Immunity 2019; 50:1232-1248.e14. [PMID: 31027998 DOI: 10.1016/j.immuni.2019.03.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 12/20/2018] [Accepted: 03/15/2019] [Indexed: 01/01/2023]
Abstract
Regulatory T cells (Treg cells) are important for preventing autoimmunity and maintaining tissue homeostasis, but whether Treg cells can adopt tissue- or immune-context-specific suppressive mechanisms is unclear. Here, we found that the enzyme hydroxyprostaglandin dehydrogenase (HPGD), which catabolizes prostaglandin E2 (PGE2) into the metabolite 15-keto PGE2, was highly expressed in Treg cells, particularly those in visceral adipose tissue (VAT). Nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ)-induced HPGD expression in VAT Treg cells, and consequential Treg-cell-mediated generation of 15-keto PGE2 suppressed conventional T cell activation and proliferation. Conditional deletion of Hpgd in mouse Treg cells resulted in the accumulation of functionally impaired Treg cells specifically in VAT, causing local inflammation and systemic insulin resistance. Consistent with this mechanism, humans with type 2 diabetes showed decreased HPGD expression in Treg cells. These data indicate that HPGD-mediated suppression is a tissue- and context-dependent suppressive mechanism used by Treg cells to maintain adipose tissue homeostasis.
Collapse
Affiliation(s)
- Lisa Schmidleithner
- Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany; LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Yasser Thabet
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Eva Schönfeld
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Maren Köhne
- Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany; LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Daniel Sommer
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Zeinab Abdullah
- Institute of Experimental Immunology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Timothy Sadlon
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Norwich Centre, 55 King William St, North Adelaide, SA 5006, Australia
| | - Collins Osei-Sarpong
- Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany; LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Kotha Subbaramaiah
- Department of Medicine, Weill Cornell Medical College, 525 E. 68(th) Street, New York, NY 10065, USA
| | - Francesca Copperi
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany
| | - Kristian Haendler
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; PRECISE, Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Tamas Varga
- Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Oliver Schanz
- LIMES-Institute, Immunology & Environment, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Svenja Bourry
- Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Kevin Bassler
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Wolfgang Krebs
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Annika E Peters
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Institute of Experimental Immunology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Ann-Kathrin Baumgart
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Institute of Experimental Immunology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Maria Schneeweiss
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Kathrin Klee
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Susanne V Schmidt
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Simone Nüssing
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Jil Sander
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Naganari Ohkura
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Andreas Waha
- Department of Neuropathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Tim Sparwasser
- Institute for Medical Microbiology and Hygiene (IMMH), Johannes Gutenberg-University Mainz, Obere Zahlbacherstr. 67, 55131 Mainz, Germany
| | - F Thomas Wunderlich
- Max Planck Institute for Metabolism Research, Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), Gleueler Str. 50, 50931 Cologne, Germany
| | - Irmgard Förster
- LIMES-Institute, Immunology & Environment, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Thomas Ulas
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Heike Weighardt
- LIMES-Institute, Immunology & Environment, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Liebigstraße 20, 04103 Leipzig, Germany
| | - Andrew J Dannenberg
- Department of Medicine, Weill Cornell Medical College, 525 E. 68(th) Street, New York, NY 10065, USA
| | - Nerea Ferreirós
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Louis J Muglia
- Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Claudia Wickenhauser
- Institute for Pathology, Martin-Luther University Halle - Wittenberg, Magdeburger Str. 14, 06112 Halle (Saale), Germany
| | - Simon C Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Norwich Centre, 55 King William St, North Adelaide, SA 5006, Australia
| | - Joachim L Schultze
- LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; PRECISE, Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Marc Beyer
- Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany; LIMES-Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; PRECISE, Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, Sigmund-Freud-Str. 27, 53127 Bonn, Germany.
| |
Collapse
|
26
|
Fingolimod reduces neuropathic pain behaviors in a mouse model of multiple sclerosis by a sphingosine-1 phosphate receptor 1-dependent inhibition of central sensitization in the dorsal horn. Pain 2019; 159:224-238. [PMID: 29140922 DOI: 10.1097/j.pain.0000000000001106] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune-inflammatory neurodegenerative disease that is often accompanied by a debilitating neuropathic pain. Disease-modifying agents slow down the progression of multiple sclerosis and prevent relapses, yet it remains unclear if they yield analgesia. We explored the analgesic potential of fingolimod (FTY720), an agonist and/or functional antagonist at the sphingosine-1-phosphate receptor 1 (S1PR1), because it reduces hyperalgesia in models of peripheral inflammatory and neuropathic pain. We used a myelin oligodendrocyte glycoprotein 35 to 55 (MOG35-55) mouse model of experimental autoimmune encephalomyelitis, modified to avoid frank paralysis, and thus, allow for assessment of withdrawal behaviors to somatosensory stimuli. Daily intraperitoneal fingolimod reduced behavioral signs of central neuropathic pain (mechanical and cold hypersensitivity) in a dose-dependent and reversible manner. Both autoimmune encephalomyelitis and fingolimod changed hyperalgesia before modifying motor function, suggesting that pain-related effects and clinical neurological deficits were modulated independently. Fingolimod also reduced cellular markers of central sensitization of neurons in the dorsal horn of the spinal cord: glutamate-evoked Ca signaling and stimulus-evoked phospho-extracellular signal-related kinase ERK (pERK) expression, as well as upregulation of astrocytes (GFAP) and macrophage/microglia (Iba1) immunoreactivity. The antihyperalgesic effects of fingolimod were prevented or reversed by the S1PR1 antagonist W146 (1 mg/kg daily, i.p.) and could be mimicked by either repeated or single injection of the S1PR1-selective agonist SEW2871. Fingolimod did not change spinal membrane S1PR1 content, arguing against a functional antagonist mechanism. We conclude that fingolimod behaves as an S1PR1 agonist to reduce pain in multiple sclerosis by reversing central sensitization of spinal nociceptive neurons.
Collapse
|
27
|
Jones M, Wen J, Selvaraj P, Tanaka M, Moran S, Zhang Y. Therapeutic Effect of the Substrate-Selective COX-2 Inhibitor IMMA in the Animal Model of Chronic Constriction Injury. Front Pharmacol 2018; 9:1481. [PMID: 30618769 PMCID: PMC6305478 DOI: 10.3389/fphar.2018.01481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/03/2018] [Indexed: 01/09/2023] Open
Abstract
Enhancement of endocannabinoid signaling has emerged as an attractive strategy for the treatment of pain. In addition to the well-characterized hydrolytic pathways, cyclooxygenase-2 (COX-2) mediated oxygenation is thought to be an alternative route for endocannabinoid metabolism and therefore provides a new avenue for drug intervention. In this study, we examined the therapeutic effect of indomethacin morpholinamide (IMMA), a novel substrate-selective COX-2 inhibitor, in the chronic constriction injury (CCI) mouse model. Treatment with IMMA significantly alleviated hyperalgesia and mechanical allodynia demonstrated by increased thermal withdrawal latency in Hargreaves test and tactile thresholds in Von Frey test. Accumulation of astrocytes and microglia in spinal cord dorsal horn and infiltration of macrophages into the dorsal root ganglion and sciatic nerve were reduced by drug treatment. Co-administration of the CB2 receptor antagonist, but not the CB1 receptor antagonist partially reversed the inhibitory effect of IMMA on pain sensitivity and inflammatory infiltrates. IMMA downregulated the mRNA expression of TNF-α and IL-1β and the production of IL-6 and MCP-1 proteins in the ipsilateral sciatic nerve. The enhanced NF-κB DNA binding activity in the CCI mouse dorsal spinal cord was also significantly reduced, suggesting that inactivation of NF-κB contributes to the anti-inflammatory property of IMMA. However, different from the previous reports showing that IMMA can increase endocannabinoids without interfering with arachidonic acid metabolism, treatment with IMMA failed to elevate the endogenous levels of AEA and 2-AG, but significantly reduced the production of prostaglandin E2 (PGE2). Furthermore, the mRNA expression of enzymes involved in PGE2 production, COX-2 and prostaglandin E synthase 2 in the ipsilateral sciatic nerve was also suppressed by IMMA treatment. Taken together, these results suggested that IMMA might exert anti-nociceptive effects through multiple mechanisms which include, but are not limited to, CB2 receptor activation and reduced PGE2 production.
Collapse
Affiliation(s)
- Melissa Jones
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Jie Wen
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Prabhuanand Selvaraj
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Mikiei Tanaka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sean Moran
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
28
|
Lötsch J, Schiffmann S, Schmitz K, Brunkhorst R, Lerch F, Ferreiros N, Wicker S, Tegeder I, Geisslinger G, Ultsch A. Machine-learning based lipid mediator serum concentration patterns allow identification of multiple sclerosis patients with high accuracy. Sci Rep 2018; 8:14884. [PMID: 30291263 PMCID: PMC6173715 DOI: 10.1038/s41598-018-33077-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/11/2018] [Indexed: 02/07/2023] Open
Abstract
Based on increasing evidence suggesting that MS pathology involves alterations in bioactive lipid metabolism, the present analysis was aimed at generating a complex serum lipid-biomarker. Using unsupervised machine-learning, implemented as emergent self-organizing maps of neuronal networks, swarm intelligence and Minimum Curvilinear Embedding, a cluster structure was found in the input data space comprising serum concentrations of d = 43 different lipid-markers of various classes. The structure coincided largely with the clinical diagnosis, indicating that the data provide a basis for the creation of a biomarker (classifier). This was subsequently assessed using supervised machine-learning, implemented as random forests and computed ABC analysis-based feature selection. Bayesian statistics-based biomarker creation was used to map the diagnostic classes of either MS patients (n = 102) or healthy subjects (n = 301). Eight lipid-markers passed the feature selection and comprised GluCerC16, LPA20:4, HETE15S, LacCerC24:1, C16Sphinganine, biopterin and the endocannabinoids PEA and OEA. A complex classifier or biomarker was developed that predicted MS at a sensitivity, specificity and accuracy of approximately 95% in training and test data sets, respectively. The present successful application of serum lipid marker concentrations to MS data is encouraging for further efforts to establish an MS biomarker based on serum lipidomics.
Collapse
Affiliation(s)
- Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-University, Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany.
- Fraunhofer Institute of Molecular Biology and Applied Ecology - Project Group Translational Medicine and Pharmacology (IME-TMP), Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany.
| | - Susanne Schiffmann
- Fraunhofer Institute of Molecular Biology and Applied Ecology - Project Group Translational Medicine and Pharmacology (IME-TMP), Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany
| | - Katja Schmitz
- Institute of Clinical Pharmacology, Goethe-University, Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany
| | - Robert Brunkhorst
- Department of Neurology, Goethe-University Hospital, Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany
| | - Florian Lerch
- DataBionics Research Group, University of Marburg, Hans - Meerwein - Straße 22, 35032, Marburg, Germany
| | - Nerea Ferreiros
- Institute of Clinical Pharmacology, Goethe-University, Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany
| | - Sabine Wicker
- Occupational Health Service, University Hospital Frankfurt, Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University, Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe-University, Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute of Molecular Biology and Applied Ecology - Project Group Translational Medicine and Pharmacology (IME-TMP), Theodor - Stern - Kai 7, 60590, Frankfurt am Main, Germany
| | - Alfred Ultsch
- DataBionics Research Group, University of Marburg, Hans - Meerwein - Straße 22, 35032, Marburg, Germany
| |
Collapse
|
29
|
Fagone P, Mazzon E, Cavalli E, Bramanti A, Petralia MC, Mangano K, Al-Abed Y, Bramati P, Nicoletti F. Contribution of the macrophage migration inhibitory factor superfamily of cytokines in the pathogenesis of preclinical and human multiple sclerosis: In silico and in vivo evidences. J Neuroimmunol 2018; 322:46-56. [PMID: 29935880 DOI: 10.1016/j.jneuroim.2018.06.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/30/2018] [Accepted: 06/12/2018] [Indexed: 01/05/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine with pleiotropic actions involved in the pathogenesis of autoimmune disorders, including Multiple Sclerosis (MS). We have first evaluated in silico the involvement of MIF, its homologue D-DT, and the receptors CD74, CD44, CXCR2 and CXCR4 in encephalitogenic T cells from a mouse model of MS, the Experimental Allergic Encephalomyelitis (EAE), as well as in circulating T helper cells from MS patients. We show an upregulation of the receptors involved in MIF signaling both in the animal model and in patients. Also, a significant increase in MIF receptors is found in the CNS lesions associated to MS. Finally, the specific inhibitor of MIF, ISO-1, improved both ex vivo and in vivo the features of EAE. Overall, our data indicate that there is a significant involvement of the MIF pathway in MS ethiopathogenesis and that interventions specifically blocking MIF receptors may represent useful therapeutic approaches in the clinical setting.
Collapse
Affiliation(s)
- Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Maria Cristina Petralia
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Department of Formative Processes, University of Catania, Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, Manhasset, New York, United States
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
30
|
de Bruin N, Ferreirós N, Schmidt M, Hofmann M, Angioni C, Geisslinger G, Parnham MJ. Mutual inversion of flurbiprofen enantiomers in various rat and mouse strains. Chirality 2018; 30:632-641. [DOI: 10.1002/chir.22826] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/10/2018] [Accepted: 01/10/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Natasja de Bruin
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Research Center for Translational Medicine and Pharmacology TMP; Industriepark Höchst; Frankfurt am Main Germany
| | - Nerea Ferreirós
- Pharmazentrum Frankfurt/ZAFES; Institute of Clinical Pharmacology, Goethe-University Frankfurt; Frankfurt am Main Germany
| | - Mike Schmidt
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Research Center for Translational Medicine and Pharmacology TMP; Industriepark Höchst; Frankfurt am Main Germany
| | - Martine Hofmann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Research Center for Translational Medicine and Pharmacology TMP; Industriepark Höchst; Frankfurt am Main Germany
| | - Carlo Angioni
- Pharmazentrum Frankfurt/ZAFES; Institute of Clinical Pharmacology, Goethe-University Frankfurt; Frankfurt am Main Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Research Center for Translational Medicine and Pharmacology TMP; Industriepark Höchst; Frankfurt am Main Germany
- Pharmazentrum Frankfurt/ZAFES; Institute of Clinical Pharmacology, Goethe-University Frankfurt; Frankfurt am Main Germany
| | - Michael John Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Research Center for Translational Medicine and Pharmacology TMP; Industriepark Höchst; Frankfurt am Main Germany
| |
Collapse
|
31
|
Stolz L, Derouiche A, Devraj K, Weber F, Brunkhorst R, Foerch C. Anticoagulation with warfarin and rivaroxaban ameliorates experimental autoimmune encephalomyelitis. J Neuroinflammation 2017; 14:152. [PMID: 28754118 PMCID: PMC5534067 DOI: 10.1186/s12974-017-0926-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/21/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In multiple sclerosis, coagulation factors have been shown to modulate inflammation. In this translational study, we investigated whether long-term anticoagulation with warfarin or rivaroxaban has beneficial effects on the course of autoimmune experimental encephalomyelitis (EAE). METHODS Female SJL/J mice treated with anticoagulants namely warfarin or rivaroxaban were immunized with PLP139-151. Stable anticoagulation was maintained throughout the entire experiment. Mice without anticoagulation treated with the vehicle only were used as controls. The neurological deficit was recorded during the course of EAE, and histopathological analyses of inflammatory lesions were performed. RESULTS In preventive settings, both treatment with warfarin and rivaroxaban reduced the maximum EAE score as compared to the control group and led to a reduction of inflammatory lesions in the spinal cord. In contrast, therapeutic treatment with warfarin had no beneficial effects on the clinical course of EAE. Signs of intraparenchymal hemorrhage at the site of the inflammatory lesions were not observed. CONCLUSION We developed long-term anticoagulation models that allowed exploring the course of EAE under warfarin and rivaroxaban treatment. We found a mild preventive effect of both warfarin and rivaroxaban on neurological deficits and local inflammation, indicating a modulation of the disease induction by anticoagulation.
Collapse
Affiliation(s)
- Leonie Stolz
- Department of Neurology, Goethe University, Schleusenweg 2-16, 60528, Frankfurt am Main, Germany.
| | - Amin Derouiche
- Dr. Senckenbergische Anatomie, Institute for Anatomy II, Goethe University, Frankfurt am Main, Germany
| | - Kavi Devraj
- Pharmazentrum Frankfurt, Institute for General Pharmacology and Toxicology, Goethe University, Frankfurt am Main, Germany.,Institute of Neurology (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Frank Weber
- Neurological Clinic Medical Park, Bad Camberg, Germany
| | - Robert Brunkhorst
- Department of Neurology, Goethe University, Schleusenweg 2-16, 60528, Frankfurt am Main, Germany
| | - Christian Foerch
- Department of Neurology, Goethe University, Schleusenweg 2-16, 60528, Frankfurt am Main, Germany
| |
Collapse
|
32
|
Cho JJ, Stewart JM, Drashansky TT, Brusko MA, Zuniga AN, Lorentsen KJ, Keselowsky BG, Avram D. An antigen-specific semi-therapeutic treatment with local delivery of tolerogenic factors through a dual-sized microparticle system blocks experimental autoimmune encephalomyelitis. Biomaterials 2017; 143:79-92. [PMID: 28772190 DOI: 10.1016/j.biomaterials.2017.07.029] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/11/2017] [Accepted: 07/21/2017] [Indexed: 01/07/2023]
Abstract
Antigen-specific treatments are highly desirable for autoimmune diseases in contrast to treatments which induce systemic immunosuppression. A novel antigen-specific therapy has been developed which, when administered semi-therapeutically, is highly efficacious in the treatment of the mouse model for multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). The treatment uses dual-sized, polymeric microparticles (dMPs) loaded with specific antigen and tolerizing factors for intra- and extra-cellular delivery, designed to recruit and modulate dendritic cells toward a tolerogenic phenotype without systemic release. This approach demonstrated robust efficacy and provided complete protection against disease. Therapeutic efficacy required encapsulation of the factors in controlled-release microparticles and was antigen-specific. Disease blocking was associated with a reduction of infiltrating CD4+ T cells, inflammatory cytokine-producing pathogenic CD4+ T cells, and activated macrophages and microglia in the central nervous system. Furthermore, CD4+ T cells isolated from dMP-treated mice were anergic in response to disease-specific, antigen-loaded splenocytes. Additionally, the frequency of CD86hiMHCIIhi dendritic cells in draining lymph nodes of EAE mice treated with Ag-specific dMPs was reduced. Our findings highlight the efficacy of microparticle-based drug delivery platform to mediate antigen-specific tolerance, and suggest that such a multi-factor combinatorial approach can act to block autoimmunity.
Collapse
Affiliation(s)
- Jonathan J Cho
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Joshua M Stewart
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Theodore T Drashansky
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Maigan A Brusko
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ashley N Zuniga
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kyle J Lorentsen
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| | - Dorina Avram
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
33
|
Sanna MD, Quattrone A, Galeotti N. Silencing of the RNA-binding protein HuR attenuates hyperalgesia and motor disability in experimental autoimmune encephalomyelitis. Neuropharmacology 2017; 123:116-125. [PMID: 28599923 DOI: 10.1016/j.neuropharm.2017.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/12/2017] [Accepted: 06/05/2017] [Indexed: 01/23/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system associated with progressive neuronal loss and axonal degeneration. Neuronal lesions and dysfunction lead often to neuropathic pain, the most prevalent and difficult to treat pain syndrome observed in MS patients. Despite its widespread occurrence, the underlying neural mechanisms for MS pain are not fully understood. For a better clarification of the pathophysiology of MS-associated pain, we investigated the role of HuR, an RNA-binding protein that positively regulates the stability of many target mRNAs, including several cytokines. The influence of HuR in the generation of the hypernociceptive response in a mouse model of relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE), an experimental model of MS, was investigated. HuR silencing, obtained through the repeated intrathecal administration of an antisense oligonucleotide (aODN) anti-HuR, completely attenuated hindpaw mechanical allodynia and thermal hyperalgesia developed by RR-EAE mice. Anti-HuR aODN also reduced severity of motor deficits as reflected by a reduction of clinical EAE score and improvement of rotarod performance. RR-EAE mice showed demyelination in spinal cord sections that was significantly reduced by HuR silencing. Double-staining immunofluorescence studies showed a neuronal localization of HuR within dorsal horn spinal cord, consistent with a neuronal mechanism of action. Our findings suggest the involvement of HuR in the hypernociceptive behaviour of RR-EAE mice providing the first pharmacological assessment of an antiallodynic and antihyperalgesic effect of HuR silencing. These data may provide support for HuR modulation as a therapeutic perspective for the management of MS-related neuropathic pain.
Collapse
Affiliation(s)
- Maria Domenica Sanna
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy.
| |
Collapse
|
34
|
Schmitz K, Brunkhorst R, de Bruin N, Mayer CA, Häussler A, Ferreiros N, Schiffmann S, Parnham MJ, Tunaru S, Chun J, Offermanns S, Foerch C, Scholich K, Vogt J, Wicker S, Lötsch J, Geisslinger G, Tegeder I. Dysregulation of lysophosphatidic acids in multiple sclerosis and autoimmune encephalomyelitis. Acta Neuropathol Commun 2017; 5:42. [PMID: 28578681 PMCID: PMC5457661 DOI: 10.1186/s40478-017-0446-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/21/2017] [Indexed: 01/18/2023] Open
Abstract
Abstract Bioactive lipids contribute to the pathophysiology of multiple sclerosis. Here, we show that lysophosphatidic acids (LPAs) are dysregulated in multiple sclerosis (MS) and are functionally relevant in this disease. LPAs and autotaxin, the major enzyme producing extracellular LPAs, were analyzed in serum and cerebrospinal fluid in a cross-sectional population of MS patients and were compared with respective data from mice in the experimental autoimmune encephalomyelitis (EAE) model, spontaneous EAE in TCR1640 mice, and EAE in Lpar2-/- mice. Serum LPAs were reduced in MS and EAE whereas spinal cord LPAs in TCR1640 mice increased during the ‘symptom-free’ intervals, i.e. on resolution of inflammation during recovery hence possibly pointing to positive effects of brain LPAs during remyelination as suggested in previous studies. Peripheral LPAs mildly re-raised during relapses but further dropped in refractory relapses. The peripheral loss led to a redistribution of immune cells from the spleen to the spinal cord, suggesting defects of lymphocyte homing. In support, LPAR2 positive T-cells were reduced in EAE and the disease was intensified in Lpar2 deficient mice. Further, treatment with an LPAR2 agonist reduced clinical signs of relapsing-remitting EAE suggesting that the LPAR2 agonist partially compensated the endogenous loss of LPAs and implicating LPA signaling as a novel treatment approach. Graphical abstract Graphical summary of lysophosphatidic signaling in multiple sclerosis![]() Electronic supplementary material The online version of this article (doi:10.1186/s40478-017-0446-4) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Schmitz K, Tegeder I. Bioluminescence and Near-infrared Imaging of Optic Neuritis and Brain Inflammation in the EAE Model of Multiple Sclerosis in Mice. J Vis Exp 2017. [PMID: 28287595 DOI: 10.3791/55321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) in SJL/J mice is a model for relapsing-remitting multiple sclerosis (RRMS). Clinical EAE scores describing motor function deficits are basic readouts of the immune-mediated inflammation of the spinal cord. However, scores and body weight do not allow for an in vivo assessment of brain inflammation and optic neuritis. The latter is an early and frequent manifestation in about 2/3 of MS patients. Here, we show methods for bioluminescence and near-infrared live imaging to assess EAE evoked optic neuritis, brain inflammation, and blood-brain barrier (BBB) disruption in living mice using an in vivo imaging system. A bioluminescent substrate activated by oxidases primarily showed optic neuritis. The signal was specific and allowed the visualization of medication effects and disease time courses, which paralleled the clinical scores. Pegylated fluorescent nanoparticles that remained within the vasculature for extended periods of time were used to assess the BBB integrity. Near-infrared imaging revealed a BBB leak at the peak of the disease. The signal was the strongest around the eyes. A near-infrared substrate for matrix metalloproteinases was used to assess EAE-evoked inflammation. Auto-fluorescence interfered with the signal, requiring spectral unmixing for quantification. Overall, bioluminescence imaging was a reliable method to assess EAE-associated optic neuritis and medication effects and was superior to the near-infrared techniques in terms of signal specificity, robustness, ease of quantification, and cost.
Collapse
Affiliation(s)
- Katja Schmitz
- Institute of Clinical Pharmacology, University Hospital Frankfurt
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, University Hospital Frankfurt;
| |
Collapse
|
36
|
Grace PM, Loram LC, Christianson JP, Strand KA, Flyer-Adams JG, Penzkover KR, Forsayeth JR, van Dam AM, Mahoney MJ, Maier SF, Chavez RA, Watkins LR. Behavioral assessment of neuropathic pain, fatigue, and anxiety in experimental autoimmune encephalomyelitis (EAE) and attenuation by interleukin-10 gene therapy. Brain Behav Immun 2017; 59:49-54. [PMID: 27189037 PMCID: PMC5108696 DOI: 10.1016/j.bbi.2016.05.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 04/30/2016] [Accepted: 05/14/2016] [Indexed: 02/04/2023] Open
Abstract
Relapsing-remitting multiple sclerosis is commonly associated with motor impairments, neuropathic pain, fatigue, mood disorders, and decreased life expectancy. However, preclinical pharmacological studies predominantly rely on clinical scoring of motor deficit as the sole behavioral endpoint. Thus, the translational potential of these studies is limited. Here, we have assessed the therapeutic potential of a novel anti-inflammatory interleukin-10 (IL-10) non-viral gene therapy formulation (XT-101-R) in a rat relapsing remitting experimental autoimmune encephalomyelitis (EAE) model. EAE induced motor deficits and neuropathic pain as reflected by induction of low-threshold mechanical allodynia, suppressed voluntary wheel running, decreased social exploration, and was associated with markedly enhanced mortality. We also noted that voluntary wheel running was depressed prior to the onset of motor deficit, and may therefore serve as a predictor of clinical symptoms onset. XT-101-R was intrathecally dosed only once at the onset of motor deficits, and attenuated each of the EAE-induced symptoms and improved survival, relative to vehicle control. This is the first pharmacological assessment of such a broad range of EAE symptoms, and provides support for IL-10 gene therapy as a clinical strategy for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Peter M. Grace
- Department of Psychology and The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Lisa C. Loram
- Department of Psychology and The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - John P. Christianson
- Department of Psychology and The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Keith A. Strand
- Department of Psychology and The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Johanna G. Flyer-Adams
- Department of Psychology and The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Kathryn R. Penzkover
- Department of Chemical & Biological Engineering Pharmacology, University of Colorado, Boulder, CO, USA
| | | | - Anne-Marie van Dam
- Department of Anatomy and Neurosciences, VU University Medical Center, Amsterdam, Netherlands
| | - Melissa J. Mahoney
- Department of Chemical & Biological Engineering Pharmacology, University of Colorado, Boulder, CO, USA
| | - Steven F. Maier
- Department of Psychology and The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | | | - Linda R. Watkins
- Department of Psychology and The Center for Neuroscience, University of Colorado, Boulder, CO, USA,Xalud Therapeutics, San Francisco, CA, USA
| |
Collapse
|
37
|
Wobst I, Ebert L, Birod K, Wegner MS, Hoffmann M, Thomas D, Angioni C, Parnham MJ, Steinhilber D, Tegeder I, Geisslinger G, Grösch S. R-Flurbiprofen Traps Prostaglandins within Cells by Inhibition of Multidrug Resistance-Associated Protein-4. Int J Mol Sci 2016; 18:ijms18010068. [PMID: 28042832 PMCID: PMC5297703 DOI: 10.3390/ijms18010068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 02/06/2023] Open
Abstract
R-flurbiprofen is the non-COX-inhibiting enantiomer of flurbiprofen and is not converted to S-flurbiprofen in human cells. Nevertheless, it reduces extracellular prostaglandin E2 (PGE2) in cancer or immune cell cultures and human extracellular fluid. Here, we show that R-flurbiprofen acts through a dual mechanism: (i) it inhibits the translocation of cPLA2α to the plasma membrane and thereby curtails the availability of arachidonic acid and (ii) R-flurbiprofen traps PGE2 inside of the cells by inhibiting multidrug resistance–associated protein 4 (MRP4, ABCC4), which acts as an outward transporter for prostaglandins. Consequently, the effects of R-flurbiprofen were mimicked by RNAi-mediated knockdown of MRP4. Our data show a novel mechanism by which R-flurbiprofen reduces extracellular PGs at physiological concentrations, particularly in cancers with high levels of MRP4, but the mechanism may also contribute to its anti-inflammatory and immune-modulating properties and suggests that it reduces PGs in a site- and context-dependent manner.
Collapse
Affiliation(s)
- Ivonne Wobst
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Lisa Ebert
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (L.E.); (M.J.P.)
| | - Kerstin Birod
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Marthe-Susanna Wegner
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Marika Hoffmann
- Institute of Pharmaceutical Chemistry, ZAFES, Johann Wolfgang Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany;
| | - Dominique Thomas
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Carlo Angioni
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Michael J. Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (L.E.); (M.J.P.)
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, ZAFES, Johann Wolfgang Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany;
| | - Irmgard Tegeder
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Gerd Geisslinger
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (L.E.); (M.J.P.)
| | - Sabine Grösch
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
- Correspondence: ; Tel.: +49/69-6301-7820; Fax: +49/69-6301-7636
| |
Collapse
|
38
|
Progress does not just come in giant leaps: adapting techniques for the study of inflammation to novel applications. Inflamm Res 2016; 66:1-12. [PMID: 27682578 DOI: 10.1007/s00011-016-0988-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/25/2016] [Indexed: 10/20/2022] Open
Abstract
INTRODUCTION Discussion of the relevance of suitable experimental models for the effective translation of drug effects to clinical inflammatory diseases has a long history. Much emphasis is placed these days on genetically transformed mice, which may have developmental drawbacks. But are established models redundant? FINDINGS Drawn from personal experience, examples are provided of the success of tinkering with technology in the context of inflammation. These include the use of specific dietary deficiency conditions, the development of new applications for established drugs and the introduction of a variety of readouts to assess outcome in studies on established disease models. Such approaches have been used to demonstrate inflammation-modulating effects of prostaglandin E, in the development of ebselen, for the introduction of immunomodulatory macrolide drugs and in new approaches to the therapy of multiple sclerosis. CONCLUSION Fine tuning of experimental approaches and evaluation technologies can often still provide innovative, clinically relevant insights into the potential beneficial effects of drugs and pharmacological agents.
Collapse
|
39
|
Zschiebsch K, Fischer C, Pickert G, Häussler A, Radeke H, Grösch S, Ferreirós N, Geisslinger G, Werner ER, Tegeder I. Tetrahydrobiopterin Attenuates DSS-evoked Colitis in Mice by Rebalancing Redox and Lipid Signalling. J Crohns Colitis 2016; 10:965-78. [PMID: 26928964 DOI: 10.1093/ecco-jcc/jjw056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/01/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND AIMS Guanosine triphosphate cyclohydrolase [GCH1] governs the production of the enzyme cofactor tetrahydrobiopterin [BH4] which is essential for biogenic amine synthesis, lipid metabolism via alkylglycerol monooxygenase [AGMO], and redox coupling of nitric oxide synthases [NOSs]. Inflammation-evoked unequal regulation of GCH1 and NOS or AGMO may cause redox stress and lipid imbalances. METHODS The present study assessed potential therapeutic effects of rebalancing these systems with BH4 in experimental colitis in mice. RESULTS Oral treatment with BH4 as a suspension of crushed tablets attenuated colitis, whereas inhibition of its production had opposite effects: aggravated weight loss, epithelial haemorrhages and ulcers, neutrophil infiltrates, production of reactive oxygen species, and unfavourable profile changes of endocannabinoids, ceramides, and lysophosphatidic acids. Conversely, oral BH4 normalised biopterin, reduced in vivo activity of oxidases and peroxidases in the inflamed gut, favoured nitric oxide over hydrogen peroxide, and maintained normal levels of lipid signalling molecules. BH4 favoured thereby resident CD3+CD8+ and regulatory CD3+CD25+ intraepithelial T cells that are important for epithelial integrity. CONCLUSIONS BH4 protected against colitis in mice via two major pathways: [i] by reduction of oxidative stress; and [ii] by re-orchestration of alkyl- and acylglycerolipid signalling via AGMO. Oral treatment with BH4 is a safe approved supplementary therapy for genetic BH4 deficiency and did not excessively increase systemic BH4 levels. Therefore, one may consider repurposing of oral BH4 as an adjunctive treatment for colitis.
Collapse
Affiliation(s)
- Katja Zschiebsch
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Caroline Fischer
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Geethanjali Pickert
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Annett Häussler
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Heinfried Radeke
- Department of Experimental Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Sabine Grösch
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Nerea Ferreirós
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Gerd Geisslinger
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Ernst R Werner
- Division of Biological Chemistry, Medical University of Innsbruck, Austria
| | - Irmgard Tegeder
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| |
Collapse
|
40
|
Barthelmes J, Tafferner N, Kurz J, de Bruin N, Parnham MJ, Geisslinger G, Schiffmann S. Induction of Experimental Autoimmune Encephalomyelitis in Mice and Evaluation of the Disease-dependent Distribution of Immune Cells in Various Tissues. J Vis Exp 2016. [PMID: 27214391 DOI: 10.3791/53933] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Multiple sclerosis is presumed to be an inflammatory autoimmune disease, which is characterized by lesion formation in the central nervous system (CNS) resulting in cognitive and motor impairment. Experimental autoimmune encephalomyelitis (EAE) is a useful animal model of MS, because it is also characterized by lesion formation in the CNS, motor impairment and is also driven by autoimmune and inflammatory reactions. One of the EAE models is induced with a peptide derived from the myelin oligodendrocyte protein (MOG)35-55 in mice. The EAE mice develop a progressive disease course. This course is divided into three phases: the preclinical phase (day 0 - 9), the disease onset (day 10 - 11) and the acute phase (day 12 - 14). MS and EAE are induced by autoreactive T cells that infiltrate the CNS. These T cells secrete chemokines and cytokines which lead to the recruitment of further immune cells. Therefore, the immune cell distribution in the spinal cord during the three disease phases was investigated. To highlight the time point of the disease at which the activation/proliferation/accumulation of T cells, B cells and monocytes starts, the immune cell distribution in lymph nodes, spleen and blood was also assessed. Furthermore, the levels of several cytokines (IL-1β, IL-6, IL-23, TNFα, IFNγ) in the three disease phases were determined, to gain insight into the inflammatory processes of the disease. In conclusion, the data provide an overview of the functional profile of immune cells during EAE pathology.
Collapse
Affiliation(s)
- Julia Barthelmes
- Institute of Clinical Pharmacology, Goethe University Hospital Frankfurt
| | - Nadja Tafferner
- Project Group for Translational Medicine & Pharmacology, Fraunhofer IME
| | - Jennifer Kurz
- Project Group for Translational Medicine & Pharmacology, Fraunhofer IME
| | - Natasja de Bruin
- Project Group for Translational Medicine & Pharmacology, Fraunhofer IME
| | - Michael J Parnham
- Project Group for Translational Medicine & Pharmacology, Fraunhofer IME
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe University Hospital Frankfurt
| | - Susanne Schiffmann
- Project Group for Translational Medicine & Pharmacology, Fraunhofer IME;
| |
Collapse
|
41
|
Endocannabinoids as Guardians of Metastasis. Int J Mol Sci 2016; 17:230. [PMID: 26875980 PMCID: PMC4783962 DOI: 10.3390/ijms17020230] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/24/2015] [Accepted: 02/01/2016] [Indexed: 01/01/2023] Open
Abstract
Endocannabinoids including anandamide and 2-arachidonoylglycerol are involved in cancer pathophysiology in several ways, including tumor growth and progression, peritumoral inflammation, nausea and cancer pain. Recently we showed that the endocannabinoid profiles are deranged during cancer to an extent that this manifests in alterations of plasma endocannabinoids in cancer patients, which was mimicked by similar changes in rodent models of local and metastatic cancer. The present topical review summarizes the complexity of endocannabinoid signaling in the context of tumor growth and metastasis.
Collapse
|
42
|
de Bruin NMWJ, Schmitz K, Schiffmann S, Tafferner N, Schmidt M, Jordan H, Häußler A, Tegeder I, Geisslinger G, Parnham MJ. Multiple rodent models and behavioral measures reveal unexpected responses to FTY720 and DMF in experimental autoimmune encephalomyelitis. Behav Brain Res 2015; 300:160-74. [PMID: 26692368 DOI: 10.1016/j.bbr.2015.12.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 11/30/2015] [Accepted: 12/09/2015] [Indexed: 10/22/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a widely-used rodent model for multiple sclerosis (MS), but a single model can hardly capture all features of MS. We investigated whether behavioral parameters in addition to clinical motor function scores could be used to assess treatment efficacy during score-free intervals in the relapsing-remitting EAE model in SJL/J mice. We studied the effects of the clinical reference compounds FTY720 (fingolimod, 0.5mg/kg/day) and dimethyl fumarate (DMF, 20-30 mg/kg/day) on clinical scores in several rodent EAE models in order to generate efficacy profiles. SJL/J mice with relapsing-remitting EAE were studied using behavioral tests, including rotarod, gait analysis, locomotor activity and grip strength. SJL/J mice were also examined according to Crawley's sociability and preference for social novelty test. Prophylactic treatment with FTY720 prevented clinical scores in three of the four EAE rodent models: Dark Agouti (DA) and Lewis rats and C57BL/6J mice. Neither prophylactic nor late-therapeutic treatment with FTY720 reduced clinical scores or reversed deficits in the rotarod test in SJL/J mice, but we observed effects on motor functions and sociability in the absence of clinical scores. Prophylactic treatment with FTY720 improved the gait of SJL/J mice whereas late-therapeutic treatment improved manifestations of reduced social (re)cognition or preference for social novelty. DMF was tested in three EAE models and did not improve clinical scores at the dose used. These data indicate that improvements in behavioral deficits can occur in absence of clinical scores, which indicate subtle drug effects and may have translational value for human MS.
Collapse
Affiliation(s)
- N M W J de Bruin
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| | - K Schmitz
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - S Schiffmann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - N Tafferner
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - M Schmidt
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - H Jordan
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - A Häußler
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - I Tegeder
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - G Geisslinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - M J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
43
|
Calvo-Rodríguez M, Núñez L, Villalobos C. Non-steroidal anti-inflammatory drugs (NSAIDs) and neuroprotection in the elderly: a view from the mitochondria. Neural Regen Res 2015; 10:1371-2. [PMID: 26604882 PMCID: PMC4625487 DOI: 10.4103/1673-5374.165219] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- María Calvo-Rodríguez
- Instituto de Biología y Genética Molecular (IBGM), Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Valladolid, c/ Sanz y Fores 3, 47003 Valladolid, Spain
| | - Lucía Núñez
- Instituto de Biología y Genética Molecular (IBGM), Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Valladolid, c/ Sanz y Fores 3, 47003 Valladolid, Spain
| | - Carlos Villalobos
- Instituto de Biología y Genética Molecular (IBGM), Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Valladolid, c/ Sanz y Fores 3, 47003 Valladolid, Spain
| |
Collapse
|
44
|
COX-2-Derived Prostaglandin E2 Produced by Pyramidal Neurons Contributes to Neurovascular Coupling in the Rodent Cerebral Cortex. J Neurosci 2015; 35:11791-810. [PMID: 26311764 DOI: 10.1523/jneurosci.0651-15.2015] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Vasodilatory prostaglandins play a key role in neurovascular coupling (NVC), the tight link between neuronal activity and local cerebral blood flow, but their precise identity, cellular origin and the receptors involved remain unclear. Here we show in rats that NMDA-induced vasodilation and hemodynamic responses evoked by whisker stimulation involve cyclooxygenase-2 (COX-2) activity and activation of the prostaglandin E2 (PgE2) receptors EP2 and EP4. Using liquid chromatography-electrospray ionization-tandem mass spectrometry, we demonstrate that PgE2 is released by NMDA in cortical slices. The characterization of PgE2 producing cells by immunohistochemistry and single-cell reverse transcriptase-PCR revealed that pyramidal cells and not astrocytes are the main cell type equipped for PgE2 synthesis, one third expressing COX-2 systematically associated with a PgE2 synthase. Consistent with their central role in NVC, in vivo optogenetic stimulation of pyramidal cells evoked COX-2-dependent hyperemic responses in mice. These observations identify PgE2 as the main prostaglandin mediating sensory-evoked NVC, pyramidal cells as their principal source and vasodilatory EP2 and EP4 receptors as their targets. SIGNIFICANCE STATEMENT Brain function critically depends on a permanent spatiotemporal match between neuronal activity and blood supply, known as NVC. In the cerebral cortex, prostaglandins are major contributors to NVC. However, their biochemical identity remains elusive and their cellular origins are still under debate. Although astrocytes can induce vasodilations through the release of prostaglandins, the recruitment of this pathway during sensory stimulation is questioned. Using multidisciplinary approaches from single-cell reverse transcriptase-PCR, mass spectrometry, to ex vivo and in vivo pharmacology and optogenetics, we provide compelling evidence identifying PgE2 as the main prostaglandin in NVC, pyramidal neurons as their main cellular source and the vasodilatory EP2 and EP4 receptors as their main targets. These original findings will certainly change the current view of NVC.
Collapse
|
45
|
Novel Activities of Select NSAID R-Enantiomers against Rac1 and Cdc42 GTPases. PLoS One 2015; 10:e0142182. [PMID: 26558612 PMCID: PMC4641600 DOI: 10.1371/journal.pone.0142182] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 10/19/2015] [Indexed: 01/02/2023] Open
Abstract
Rho family GTPases (including Rac, Rho and Cdc42) collectively control cell proliferation, adhesion and migration and are of interest as functional therapeutic targets in numerous epithelial cancers. Based on high throughput screening of the Prestwick Chemical Library® and cheminformatics we identified the R-enantiomers of two approved drugs (naproxen and ketorolac) as inhibitors of Rac1 and Cdc42. The corresponding S-enantiomers are considered the active component in racemic drug formulations, acting as non-steroidal anti-inflammatory drugs (NSAIDs) with selective activity against cyclooxygenases. Here, we show that the S-enantiomers of naproxen and ketorolac are inactive against the GTPases. Additionally, more than twenty other NSAIDs lacked inhibitory action against the GTPases, establishing the selectivity of the two identified NSAIDs. R-naproxen was first identified as a lead compound and tested in parallel with its S-enantiomer and the non-chiral 6-methoxy-naphthalene acetic acid (active metabolite of nabumetone, another NSAID) as a structural series. Cheminformatics-based substructure analyses—using the rotationally constrained carboxylate in R-naproxen—led to identification of racemic [R/S] ketorolac as a suitable FDA-approved candidate. Cell based measurement of GTPase activity (in animal and human cell lines) demonstrated that the R-enantiomers specifically inhibit epidermal growth factor stimulated Rac1 and Cdc42 activation. The GTPase inhibitory effects of the R-enantiomers in cells largely mimic those of established Rac1 (NSC23766) and Cdc42 (CID2950007/ML141) specific inhibitors. Docking predicts that rotational constraints position the carboxylate moieties of the R-enantiomers to preferentially coordinate the magnesium ion, thereby destabilizing nucleotide binding to Rac1 and Cdc42. The S-enantiomers can be docked but are less favorably positioned in proximity to the magnesium. R-naproxen and R-ketorolac have potential for rapid translation and efficacy in the treatment of several epithelial cancer types on account of established human toxicity profiles and novel activities against Rho-family GTPases.
Collapse
|
46
|
COX-2-Derived Prostaglandin E2 Produced by Pyramidal Neurons Contributes to Neurovascular Coupling in the Rodent Cerebral Cortex. J Neurosci 2015. [PMID: 26311764 DOI: 10.1523/jneurosci.0651‐15.2015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Vasodilatory prostaglandins play a key role in neurovascular coupling (NVC), the tight link between neuronal activity and local cerebral blood flow, but their precise identity, cellular origin and the receptors involved remain unclear. Here we show in rats that NMDA-induced vasodilation and hemodynamic responses evoked by whisker stimulation involve cyclooxygenase-2 (COX-2) activity and activation of the prostaglandin E2 (PgE2) receptors EP2 and EP4. Using liquid chromatography-electrospray ionization-tandem mass spectrometry, we demonstrate that PgE2 is released by NMDA in cortical slices. The characterization of PgE2 producing cells by immunohistochemistry and single-cell reverse transcriptase-PCR revealed that pyramidal cells and not astrocytes are the main cell type equipped for PgE2 synthesis, one third expressing COX-2 systematically associated with a PgE2 synthase. Consistent with their central role in NVC, in vivo optogenetic stimulation of pyramidal cells evoked COX-2-dependent hyperemic responses in mice. These observations identify PgE2 as the main prostaglandin mediating sensory-evoked NVC, pyramidal cells as their principal source and vasodilatory EP2 and EP4 receptors as their targets. SIGNIFICANCE STATEMENT Brain function critically depends on a permanent spatiotemporal match between neuronal activity and blood supply, known as NVC. In the cerebral cortex, prostaglandins are major contributors to NVC. However, their biochemical identity remains elusive and their cellular origins are still under debate. Although astrocytes can induce vasodilations through the release of prostaglandins, the recruitment of this pathway during sensory stimulation is questioned. Using multidisciplinary approaches from single-cell reverse transcriptase-PCR, mass spectrometry, to ex vivo and in vivo pharmacology and optogenetics, we provide compelling evidence identifying PgE2 as the main prostaglandin in NVC, pyramidal neurons as their main cellular source and the vasodilatory EP2 and EP4 receptors as their main targets. These original findings will certainly change the current view of NVC.
Collapse
|
47
|
Chicca A, Gachet MS, Petrucci V, Schuehly W, Charles RP, Gertsch J. 4'-O-methylhonokiol increases levels of 2-arachidonoyl glycerol in mouse brain via selective inhibition of its COX-2-mediated oxygenation. J Neuroinflammation 2015; 12:89. [PMID: 25962384 PMCID: PMC4490613 DOI: 10.1186/s12974-015-0307-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/24/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE 4'-O-methylhonokiol (MH) is a natural product showing anti-inflammatory, anti-osteoclastogenic, and neuroprotective effects. MH was reported to modulate cannabinoid CB2 receptors as an inverse agonist for cAMP production and an agonist for intracellular [Ca2+]. It was recently shown that MH inhibits cAMP formation via CB2 receptors. In this study, the exact modulation of MH on CB2 receptor activity was elucidated and its endocannabinoid substrate-specific inhibition (SSI) of cyclooxygenase-2 (COX-2) and CNS bioavailability are described for the first time. METHODS CB2 receptor modulation ([35S]GTPγS, cAMP, and β-arrestin) by MH was measured in hCB2-transfected CHO-K1 cells and native conditions (HL60 cells and mouse spleen). The COX-2 SSI was investigated in RAW264.7 cells and in Swiss albino mice by targeted metabolomics using LC-MS/MS. RESULTS MH is a CB2 receptor agonist and a potent COX-2 SSI. It induced partial agonism in both the [35S]GTPγS binding and β-arrestin recruitment assays while being a full agonist in the cAMP pathway. MH selectively inhibited PGE2 glycerol ester formation (over PGE2) in RAW264.7 cells and significantly increased the levels of 2-AG in mouse brain in a dose-dependent manner (3 to 20 mg kg(-1)) without affecting other metabolites. After 7 h from intraperitoneal (i.p.) injection, MH was quantified in significant amounts in the brain (corresponding to 200 to 300 nM). CONCLUSIONS LC-MS/MS quantification shows that MH is bioavailable to the brain and under condition of inflammation exerts significant indirect effects on 2-AG levels. The biphenyl scaffold might serve as valuable source of dual CB2 receptor modulators and COX-2 SSIs as demonstrated by additional MH analogs that show similar effects. The combination of CB2 agonism and COX-2 SSI offers a yet unexplored polypharmacology with expected synergistic effects in neuroinflammatory diseases, thus providing a rationale for the diverse neuroprotective effects reported for MH in animal models.
Collapse
Affiliation(s)
- Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| | - Maria Salomé Gachet
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| | - Vanessa Petrucci
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| | - Wolfgang Schuehly
- Institute of Zoology, Karl-Franzens-University Graz, Universitätsplatz 2, 8010, Graz, Austria.
| | - Roch-Philippe Charles
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| |
Collapse
|