1
|
Mazzantini C, Venturini M, Lana D, Mulas G, Santalmasi C, Magni G, Bruni P, Pugliese AM, Cencetti F, Pellegrini-Giampietro DE, Landucci E. Dual action of sphingosine 1-phosphate pathway in in vitro models of global cerebral ischemia. Neurobiol Dis 2025; 208:106865. [PMID: 40068722 DOI: 10.1016/j.nbd.2025.106865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025] Open
Abstract
It is well accepted that sphingolipids play an important role in the pathological process of cerebral ischemia. In the present study we have investigated the involvement of sphingosine 1-phosphate (S1P) pathway in two different in vitro models of global ischemia. In organotypic hippocampal slices exposed to oxygen and glucose deprivation (OGD) we evaluated the mRNA expression of S1P metabolic enzymes and receptors (S1P1-5) by Real Time-PCR. In the same model we investigated the effect of the inhibitor of S1P lyase (SPL), LX2931, the selective antagonists of S1P2, JTE-013, and S1P3, CAY10444, quantifying the cell death in the CA1 region by propidium iodide fluorescence, and morphological and tissue organization alterations by immunohistochemistry and confocal microscopy. Moreover, we performed extracellular recordings of field excitatory postsynaptic potentials in acute slices exposed to OGD. In organotypic slices OGD induced a significant increase of SPL at mRNA level and of S1P2 and S1P3 at both mRNA and protein level. The incubation with LX2931, JTE-013 or CAY10444 was able to reduce CA1 damage induced by OGD in organotypic slices and provoked a significant delay of the onset of anoxic depolarization on acute slices. Moreover, S1P2 and S1P3 antagonists prevented the increase of TREM2 induced by OGD. Our results reveal a dual role of S1P pathway in brain ischemia: intracellular S1P, degraded via SPL, appears to be beneficial whereas signaling via S1P2 and S1P3 is detrimental to the disease. These findings support the notion that SPL, S1P2 and S1P3 are promising therapeutic targets in brain ischemia.
Collapse
Affiliation(s)
- Costanza Mazzantini
- Dept. of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Martina Venturini
- Dept. of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Daniele Lana
- Dept. of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Gloria Mulas
- Dept. of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Clara Santalmasi
- Dept. of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Giada Magni
- Institute of Applied Physics "Nello Carrara", National Research Council (IFAC-CNR), Sesto Fiorentino, Florence, Italy
| | - Paola Bruni
- Dept. of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Anna Maria Pugliese
- Dept. of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Francesca Cencetti
- Dept. of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy.
| | | | - Elisa Landucci
- Dept. of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy.
| |
Collapse
|
2
|
Chiang PT, Tsai LK, Tsai HH. New targets in spontaneous intracerebral hemorrhage. Curr Opin Neurol 2025; 38:10-17. [PMID: 39325041 PMCID: PMC11706352 DOI: 10.1097/wco.0000000000001325] [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: 09/27/2024]
Abstract
PURPOSE OF REVIEW Intracerebral hemorrhage (ICH) is a devastating stroke with limited medical treatments; thus, timely exploration of emerging therapeutic targets is essential. This review focuses on the latest strategies to mitigate secondary brain injury post-ICH other than targeting surgery or hemostasis, addressing a significant gap in clinical practice and highlighting potential improvements in patient outcomes. RECENT FINDINGS Promising therapeutic targets to reduce secondary brain injury following ICH have recently been identified, including attenuation of iron toxicity and inhibition of ferroptosis, enhancement of endogenous resorption of hematoma, and modulation of perihematomal inflammatory responses and edema. Additionally, novel insights suggest the lymphatic system of the brain may potentially play a role in hematoma clearance and edema management. Various experimental and early-phase clinical trials have demonstrated these approaches may potentially offer clinical benefits, though most research remains in the preliminary stages. SUMMARY Continued research is essential to identify multifaceted treatment strategies for ICH. Clinical translation of these emerging targets could significantly enhance the efficacy of therapeutic interventions and potentially reduce secondary brain damage and improve neurological recovery. Future efforts should focus on large-scale clinical trials to validate these approaches, to pave the way for more effective treatment protocols for spontaneous ICH.
Collapse
Affiliation(s)
- Pu-Tien Chiang
- Department of Neurology, National Taiwan University Hospital
- Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, National Taiwan University Hospital
| | - Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital
| |
Collapse
|
3
|
Mohammed EMA. Understanding Multiple Sclerosis Pathophysiology and Current Disease-Modifying Therapies: A Review of Unaddressed Aspects. FRONT BIOSCI-LANDMRK 2024; 29:386. [PMID: 39614433 DOI: 10.31083/j.fbl2911386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder of the central nervous system (CNS) with an unknown etiology and pathophysiology that is not completely understood. Although great strides have been made in developing disease-modifying therapies (DMTs) that have significantly improved the quality of life for MS patients, these treatments do not entirely prevent disease progression or relapse. Identifying the unaddressed pathophysiological aspects of MS and developing targeted therapies to fill in these gaps are essential in providing long-term relief for patients. Recent research has uncovered some aspects of MS that remain outside the scope of available DMTs, and as such, yield only limited benefits. Despite most MS pathophysiology being targeted by DMTs, many patients still experience disease progression or relapse, indicating that a more detailed understanding is necessary. Thus, this literature review seeks to explore the known aspects of MS pathophysiology, identify the gaps in present DMTs, and explain why current treatments cannot entirely arrest MS progression.
Collapse
Affiliation(s)
- Eiman M A Mohammed
- Kuwait Cancer Control Centre, Department of Medical Laboratory, Molecular Genetics Laboratory, Ministry of Health, 13001 Shuwaikh, Kuwait
| |
Collapse
|
4
|
Bisht M, Kadian JP, Hooda T, Jain N, Lather A, Aggarwal N. Explore the Role of the Sphingosine-1-Phosphate Signalling as a Novel Promising Therapeutic Target for the Management of Parkinson's Disease. Drug Res (Stuttg) 2024; 74:365-378. [PMID: 39353579 DOI: 10.1055/a-2401-4578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Sphingosine-1-phosphate (S1P) is a cellular signalling molecule derived from sphingosine, which is a pro-apoptotic sphingolipid. Sphingolipids control various cellular actions like growth, homeostasis, and stress-related responses. The main sources of S1P in our body are erythrocytes. S1P controls both cellular mediators and other second messengers intracellularly. The S1P receptor also helps in inflammatory and neuroprotective effects (required to manage of Parkinson's). A large number of anti-Parkinson drugs are available, but still, there is a need for more effective and safer drugs. S1P and its receptors could be targeted as novel drugs due to their involvement in neuro-inflammation and Parkinson's. The present review effort to explore the biological role of S1P and related receptors, for their possible involvement in PD; furthermore. Overall, S1P and other related metabolizing enzymes have significant therapeutic opportunities for Parkinson's disease along with other neurological disorders.
Collapse
Affiliation(s)
- Manoj Bisht
- Devasthali Vidyapeeth College of Pharmacy, Rudrapur, Uttarakhand, India
| | - Jai Parkash Kadian
- Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh (Saharanpur)- UP India
| | - Tanuj Hooda
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Neelam Jain
- Department of Pharmaceutical Education & Research, Bhagat Phool Singh Mahila Vishwavidyalaya, Khanpur Kalan, Sonepat, Haryana, India
| | - Amit Lather
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Navidha Aggarwal
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| |
Collapse
|
5
|
Birgbauer E. Lysophospholipid receptors in neurodegeneration and neuroprotection. EXPLORATION OF NEUROPROTECTIVE THERAPY 2024; 4:349-365. [PMID: 39247084 PMCID: PMC11379401 DOI: 10.37349/ent.2024.00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024]
Abstract
The central nervous system (CNS) is one of the most complex physiological systems, and treatment of CNS disorders represents an area of major medical need. One critical aspect of the CNS is its lack of regeneration, such that damage is often permanent. The damage often leads to neurodegeneration, and so strategies for neuroprotection could lead to major medical advances. The G protein-coupled receptor (GPCR) family is one of the major receptor classes, and they have been successfully targeted clinically. One class of GPCRs is those activated by bioactive lysophospholipids as ligands, especially sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA). Research has been increasingly demonstrating the important roles that S1P and LPA, and their receptors, play in physiology and disease. In this review, I describe the role of S1P and LPA receptors in neurodegeneration and potential roles in neuroprotection. Much of our understanding of the role of S1P receptors has been through pharmacological tools. One such tool, fingolimod (also known as FTY720), which is a S1P receptor agonist but a functional antagonist in the immune system, is clinically efficacious in multiple sclerosis by producing a lymphopenia to reduce autoimmune attacks; however, there is evidence that fingolimod is also neuroprotective. Furthermore, fingolimod is neuroprotective in many other neuropathologies, including stroke, Parkinson's disease, Huntington's disease, Rett syndrome, Alzheimer's disease, and others that are discussed here. LPA receptors also appear to be involved, being upregulated in a variety of neuropathologies. Antagonists or mutations of LPA receptors, especially LPA1, are neuroprotective in a variety of conditions, including cortical development, traumatic brain injury, spinal cord injury, stroke and others discussed here. Finally, LPA receptors may interact with other receptors, including a functional interaction with plasticity related genes.
Collapse
Affiliation(s)
- Eric Birgbauer
- Department of Biology, Winthrop University, Rock Hill, SC 29733, USA
| |
Collapse
|
6
|
Magalhães DM, Stewart NA, Mampay M, Rolle SO, Hall CM, Moeendarbary E, Flint MS, Sebastião AM, Valente CA, Dymond MK, Sheridan GK. The sphingosine 1-phosphate analogue, FTY720, modulates the lipidomic signature of the mouse hippocampus. J Neurochem 2024; 168:1113-1142. [PMID: 38339785 DOI: 10.1111/jnc.16073] [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: 08/17/2023] [Revised: 12/27/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024]
Abstract
The small-molecule drug, FTY720 (fingolimod), is a synthetic sphingosine 1-phosphate (S1P) analogue currently used to treat relapsing-remitting multiple sclerosis in both adults and children. FTY720 can cross the blood-brain barrier (BBB) and, over time, accumulate in lipid-rich areas of the central nervous system (CNS) by incorporating into phospholipid membranes. FTY720 has been shown to enhance cell membrane fluidity, which can modulate the functions of glial cells and neuronal populations involved in regulating behaviour. Moreover, direct modulation of S1P receptor-mediated lipid signalling by FTY720 can impact homeostatic CNS physiology, including neurotransmitter release probability, the biophysical properties of synaptic membranes, ion channel and transmembrane receptor kinetics, and synaptic plasticity mechanisms. The aim of this study was to investigate how chronic FTY720 treatment alters the lipid composition of CNS tissue in adolescent mice at a key stage of brain maturation. We focused on the hippocampus, a brain region known to be important for learning, memory, and the processing of sensory and emotional stimuli. Using mass spectrometry-based lipidomics, we discovered that FTY720 increases the fatty acid chain length of hydroxy-phosphatidylcholine (PCOH) lipids in the mouse hippocampus. It also decreases PCOH monounsaturated fatty acids (MUFAs) and increases PCOH polyunsaturated fatty acids (PUFAs). A total of 99 lipid species were up-regulated in the mouse hippocampus following 3 weeks of oral FTY720 exposure, whereas only 3 lipid species were down-regulated. FTY720 also modulated anxiety-like behaviours in young mice but did not affect spatial learning or memory formation. Our study presents a comprehensive overview of the lipid classes and lipid species that are altered in the hippocampus following chronic FTY720 exposure and provides novel insight into cellular and molecular mechanisms that may underlie the therapeutic or adverse effects of FTY720 in the central nervous system.
Collapse
Affiliation(s)
- Daniela M Magalhães
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
- School of Applied Sciences, University of Brighton, Brighton, UK
| | | | - Myrthe Mampay
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Sara O Rolle
- Green Templeton College, University of Oxford, Oxford, UK
| | - Chloe M Hall
- School of Applied Sciences, University of Brighton, Brighton, UK
- Department of Mechanical Engineering, University College London, London, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK
- 199 Biotechnologies Ltd, London, UK
| | - Melanie S Flint
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
| | - Cláudia A Valente
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
| | - Marcus K Dymond
- School of Applied Sciences, University of Brighton, Brighton, UK
| | | |
Collapse
|
7
|
Wang L, Zhang X, Ma C, Wu N. 1-Phosphate receptor agonists: A promising therapeutic avenue for ischemia-reperfusion injury management. Int Immunopharmacol 2024; 131:111835. [PMID: 38508097 DOI: 10.1016/j.intimp.2024.111835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
Ischemia-reperfusion injury (IRI) - a complex pathological condition occurring when blood supply is abruptly restored to ischemic tissues, leading to further tissue damage - poses a significant clinical challenge. Sphingosine-1-phosphate receptors (S1PRs), a specialized set of G-protein-coupled receptors comprising five subtypes (S1PR1 to S1PR5), are prominently present in various cell membranes, including those of lymphocytes, cardiac myocytes, and endothelial cells. Increasing evidence highlights the potential of targeting S1PRs for IRI therapeutic intervention. Notably, preconditioning and postconditioning strategies involving S1PR agonists like FTY720 have demonstrated efficacy in mitigating IRI. As the synthesis of a diverse array of S1PR agonists continues, with FTY720 being a prime example, the body of experimental evidence advocating for their role in IRI treatment is expanding. Despite this progress, comprehensive reviews delineating the therapeutic landscape of S1PR agonists in IRI remain limited. This review aspires to meticulously elucidate the protective roles and mechanisms of S1PR agonists in preventing and managing IRI affecting various organs, including the heart, kidney, liver, lungs, intestines, and brain, to foster novel pharmacological approaches in clinical settings.
Collapse
Affiliation(s)
- Linyuan Wang
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China; The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaowen Zhang
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Nan Wu
- The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
8
|
Gaastra B, Zhang J, Tapper W, Bulters D, Galea I. Sphingosine-1-phosphate Signalling in Aneurysmal Subarachnoid Haemorrhage: Basic Science to Clinical Translation. Transl Stroke Res 2024; 15:352-363. [PMID: 36749550 PMCID: PMC10891271 DOI: 10.1007/s12975-023-01133-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/08/2023]
Abstract
Sphingosine-1-phosphate (S1P) is generated intracellularly and, when transported to the extracellular compartment, predominantly signals through S1P receptors. The S1P signalling pathway has been implicated in the pathophysiology of neurological injury following aneurysmal subarachnoid haemorrhage (aSAH). In this review, we bring together all the available data regarding the role of S1P in neurological injury following aSAH. There is agreement in the literature that S1P increases in the cerebrospinal fluid following aSAH and leads to cerebral artery vasospasm. On the other hand, the role of S1P in the parenchyma is less clear cut, with different studies arguing for beneficial and deleterious effects. A parsimonious interpretation of this apparently conflicting data is presented. We discuss the potential of S1P receptor modulators, in clinical use for multiple sclerosis, to be repurposed for aSAH. Finally, we highlight the gaps in our knowledge of S1P signalling in humans, the clinical challenges of targeting the S1P pathway after aSAH and other research priorities.
Collapse
Affiliation(s)
- Ben Gaastra
- Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK.
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton, Southampton, SO16 6YD, UK.
| | - John Zhang
- Center of Neuroscience Research, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Will Tapper
- Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK
| | - Diederik Bulters
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton, Southampton, SO16 6YD, UK
| | - Ian Galea
- Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
9
|
Skoug C, Erdogan H, Vanherle L, Vieira JPP, Matthes F, Eliasson L, Meissner A, Duarte JMN. Density of Sphingosine-1-Phosphate Receptors Is Altered in Cortical Nerve-Terminals of Insulin-Resistant Goto-Kakizaki Rats and Diet-Induced Obese Mice. Neurochem Res 2024; 49:338-347. [PMID: 37794263 PMCID: PMC10787890 DOI: 10.1007/s11064-023-04033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/21/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a phosphosphingolipid with pleiotropic biological functions. S1P acts as an intracellular second messenger, as well as extracellular ligand to five G-protein coupled receptors (S1PR1-5). In the brain, S1P regulates neuronal proliferation, apoptosis, synaptic activity and neuroglia activation. Moreover, S1P metabolism alterations have been reported in neurodegenerative disorders. We have previously reported that S1PRs are present in nerve terminals, exhibiting distinct sub-synaptic localization and neuromodulation actions. Since type 2 diabetes (T2D) causes synaptic dysfunction, we hypothesized that S1P signaling is modified in nerve terminals. In this study, we determined the density of S1PRs in cortical synaptosomes from insulin-resistant Goto-Kakizaki (GK) rats and Wistar controls, and from mice fed a high-fat diet (HFD) and low-fat-fed controls. Relative to their controls, GK rats showed similar cortical S1P concentration despite higher S1P levels in plasma, yet lower density of S1PR1, S1PR2 and S1PR4 in nerve-terminal-enriched membranes. HFD-fed mice exhibited increased plasma and cortical concentrations of S1P, and decreased density of S1PR1 and S1PR4. These findings point towards altered S1P signaling in synapses of insulin resistance and diet-induced obesity models, suggesting a role of S1P signaling in T2D-associated synaptic dysfunction.
Collapse
Affiliation(s)
- Cecilia Skoug
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, 221 84, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Hüseyin Erdogan
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, 221 84, Lund, Sweden
| | - Lotte Vanherle
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, 221 84, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - João P P Vieira
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, 221 84, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Frank Matthes
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, 221 84, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Lena Eliasson
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden
- Clinical Research Center, Skåne University Hospital, Malmö, Sweden
| | - Anja Meissner
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, 221 84, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Physiology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - João M N Duarte
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, 221 84, Lund, Sweden.
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
10
|
Kar SS, Gharai SR, Sahu SK, Ravichandiran V, Swain SP. The Current Landscape in the Development of Small-molecule Modulators Targeting Sphingosine-1-phosphate Receptors to Treat Neurodegenerative Diseases. Curr Top Med Chem 2024; 24:2431-2446. [PMID: 38676503 DOI: 10.2174/0115680266288509240422112839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/29/2024]
Abstract
Sphingosine 1-phosphate (S1P) is extensively researched as a lysophospholipid and is crucial in various physiological and pathological processes. It achieves this via signalling through five different subtypes of G protein-coupled receptors (GPCRs), namely S1PR1 to S1PR5. S1PR modulators possess the ability to traverse the blood-brain barrier, potentially leading to direct actions within the Central Nervous System (CNS). S1PR modulators specifically bind to receptors located on the surface of naive and central memory lymphocytes, causing these cells to be trapped or confined within the lymph node. The investigation of the S1P pathway has resulted in the approval of three S1PR modulators, namely fingolimod, siponimod, and ozanimod, as medications for the treatment of patients suffering from Multiple Sclerosis (MS). Additionally, new S1PR modulators, such as ponesimod and etrasimod, are currently being developed and tested in clinical trials. Research on the creation of S1P modulators in neurodegenerative illnesses is ongoing as scientists continue to explore novel possibilities for selective S1P modulators. This study provides a concise overview of sphingolipid metabolism, the mechanism by which S1P receptors are affected, and the structural characteristics of several small molecule S1P modulators, with a particular focus on their structure-activity connections.
Collapse
Affiliation(s)
- Sidhartha Sankar Kar
- Faculty of Pharmacy, C. V. Raman Global University, Mahura, Bhubaneswar, 752054, Odisha, India
| | - Soumya Ranjan Gharai
- Department of Pharmaceutical Chemistry, Institute of Pharmacy & Technology, Salipur, Cuttack, 754202, Odisha, India
| | - Sujit Kumar Sahu
- Department of Pharmaceutical Chemistry, Institute of Pharmacy & Technology, Salipur, Cuttack, 754202, Odisha, India
| | - Velayutham Ravichandiran
- Department of Medicinal Chemistry and Centre for Marine Therapeutics (CMT), National Institute of Pharmaceutical Education and Research, Kolkata, 168, Maniktala Main Road, Kolkata, 700054, India
| | - Sharada Prasanna Swain
- Department of Medicinal Chemistry and Centre for Marine Therapeutics (CMT), National Institute of Pharmaceutical Education and Research, Kolkata, 168, Maniktala Main Road, Kolkata, 700054, India
| |
Collapse
|
11
|
Basavarajappa D, Gupta V, Chitranshi N, Viswanathan D, Gupta V, Vander Wall R, Palanivel V, Mirzaei M, You Y, Klistorner A, Graham SL. Anti-inflammatory Effects of Siponimod in a Mouse Model of Excitotoxicity-Induced Retinal Injury. Mol Neurobiol 2023; 60:7222-7237. [PMID: 37542647 PMCID: PMC10657799 DOI: 10.1007/s12035-023-03535-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/22/2023] [Indexed: 08/07/2023]
Abstract
Glaucoma is a leading cause of permanent blindness worldwide and is characterized by neurodegeneration linked to progressive retinal ganglion cell (RGC) death, axonal damage, and neuroinflammation. Glutamate excitotoxicity mediated through N-methyl-D-aspartate (NMDA) receptors plays a crucial role in glaucomatous RGC loss. Sphingosine 1-phosphate receptors (S1PRs) are important mediators of neurodegeneration and neuroinflammation in the brain and the retina. Siponimod is an immunomodulatory drug for multiple sclerosis and is a selective modulator of S1PR subtypes 1 and 5 and has been shown to have beneficial effects on the central nervous system (CNS) in degenerative conditions. Our previous study showed that mice administered orally with siponimod protected inner retinal structure and function against acute NMDA excitotoxicity. To elucidate the molecular mechanisms behind these protective effects, we investigated the inflammatory pathways affected by siponimod treatment in NMDA excitotoxicity model. NMDA excitotoxicity resulted in the activation of glial cells coupled with upregulation of the inflammatory NF-kB pathway and increased expression of TNFα, IL1-β, and IL-6. Siponimod treatment significantly reduced glial activation and suppressed the pro-inflammatory pathways. Furthermore, NMDA-induced activation of NLRP3 inflammasome and upregulation of neurotoxic inducible nitric oxide synthase (iNOS) were significantly diminished with siponimod treatment. Our data demonstrated that siponimod induces anti-inflammatory effects via suppression of glial activation and inflammatory singling pathways that could protect the retina against acute excitotoxicity conditions. These findings provide insights into the anti-inflammatory effects of siponimod in the CNS and suggest a potential therapeutic strategy for neuroinflammatory conditions.
Collapse
Affiliation(s)
- Devaraj Basavarajappa
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia.
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia.
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Deepa Viswanathan
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Roshana Vander Wall
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Viswanthram Palanivel
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Yuyi You
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Alexander Klistorner
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Stuart L Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
- Save Sight Institute, The University of Sydney, Sydney, NSW, 2000, Australia
| |
Collapse
|
12
|
Vališ M, Achiron A, Hartung HP, Mareš J, Tichá V, Štourač P, Halusková S, Angelucci F, Pavelek Z. The Benefits and Risks of Switching from Fingolimod to Siponimod for the Treatment of Relapsing-Remitting and Secondary Progressive Multiple Sclerosis. Drugs R D 2023; 23:331-338. [PMID: 37640862 PMCID: PMC10676342 DOI: 10.1007/s40268-023-00434-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2023] [Indexed: 08/31/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease that affects the central nervous system (CNS). Currently, MS treatment is limited to several Food and Drug Administration (FDA)- and European Medicines Agency (EMA)-approved medications that slow disease progression by immunomodulatory action. Fingolimod and siponimod have similar mechanisms of action, and consequently, their therapeutic effects may be comparable. However, while fingolimod is mainly used for relapsing-remitting MS (RRMS), siponimod, according to EMA label, is recommended for active secondary progressive MS (SPMS). Clinicians and scientists are analysing whether patients can switch from fingolimod to siponimod and identifying the advantages or disadvantages of such a switch from a therapeutic point of view. In this review, we aim to discuss the therapeutic effects of these two drugs and the advantages/disadvantages of switching treatment from fingolimod to siponimod in patients with the most common forms of MS, RRMS and SPMS.
Collapse
Affiliation(s)
- Martin Vališ
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05, Hradec Králové, Czech Republic
| | - Anat Achiron
- Multiple Sclerosis Center, Sheba Medical Center, Tel-Hashomer, Israel
- Neurology Department, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Hans Peter Hartung
- Department of Neurology, Medical School, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
- Department of Neurology, Faculty of Medicine at Palacký University and University Hospital in Olomouc, I. P. Pavlova 6, Olomouc, Czech Republic
- Brain and Mind Center, University of Sydney, Sydney, Australia
| | - Jan Mareš
- Department of Neurology, Faculty of Medicine at Palacký University and University Hospital in Olomouc, I. P. Pavlova 6, Olomouc, Czech Republic
| | - Veronika Tichá
- First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Pavel Štourač
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05, Hradec Králové, Czech Republic
| | - Simona Halusková
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05, Hradec Králové, Czech Republic
| | - Francesco Angelucci
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05, Hradec Králové, Czech Republic
- Memory Clinic, Department of Neurology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Zbyšek Pavelek
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05, Hradec Králové, Czech Republic.
| |
Collapse
|
13
|
Kim E, Fortoul MC, Weimer D, Meggyesy M, Demory Beckler M. Co-occurrence of glioma and multiple sclerosis: Prevailing theories and emerging therapies. Mult Scler Relat Disord 2023; 79:105027. [PMID: 37801959 DOI: 10.1016/j.msard.2023.105027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/01/2023] [Accepted: 09/23/2023] [Indexed: 10/08/2023]
Abstract
Though the concurrence of primary brain tumors and multiple sclerosis (MS) is exceedingly rare, instances have been noted in the literature as early as 1949. Given these observations, researchers have proposed various ideas as to how these malignancies may be linked to MS. Due to insufficient data, none have gained traction or been widely accepted amongst neurologists or neuro-oncologists. What is abundantly clear, however, is the mounting uncertainty faced by clinicians when caring for these individuals. Concerns persist about the potential for disease modifying therapies (DMTs) to initiate or promote tumor growth and progression, and to date, there are no approved treatments capable of mitigating both MS disease activity and tumor growth, let alone established guidelines that clinicians may refer to. Collectively, these gaps in the literature impose limitations to optimizing the care and management of this population. As such, our hope is to stimulate further discussion of this topic and prompt future investigations to explore novel treatment options and advance our understanding of these concurrent disease processes. To this end, the chief objective of this article is to evaluate proposed ideas of how the diseases may be linked, outline emerging therapies for both MS and brain tumors, and describe evidence-based approaches to diagnosing and treating this patient population.
Collapse
Affiliation(s)
- Enoch Kim
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, 3200 S University Drive, Fort Lauderdale, FL 33328, United States
| | - Marla C Fortoul
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, 3200 S University Drive, Fort Lauderdale, FL 33328, United States
| | - Derek Weimer
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, 3200 S University Drive, Fort Lauderdale, FL 33328, United States
| | - Michael Meggyesy
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michelle Demory Beckler
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, 3200 S University Drive, Fort Lauderdale, FL 33328, United States.
| |
Collapse
|
14
|
Roggeri A, Olivero G, Usai C, Vanmierlo T, Pittaluga A. Presynaptic Release-Regulating Sphingosine 1-Phosphate 1/3 Receptors in Cortical Glutamatergic Terminals: Adaptations in EAE Mice and Impact of Therapeutic FTY720. Cells 2023; 12:2343. [PMID: 37830557 PMCID: PMC10571862 DOI: 10.3390/cells12192343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
This study provides evidence of the existence of presynaptic inhibitory sphingosine-1-phosphate receptor 1 (S1P1R) and facilitatory S1P3R in cortical nerve endings (synaptosomes) of healthy mice. The conclusion relies on the findings that (i) the S1P1R agonist CS-2100 (0.1-30 nM) inhibits the 12 mM KCl-evoked glutamate exocytosis (quantified as the release of [3H]D-aspartate) while the S1P3R allosteric agonist CYM-5541 potentiates it and (ii) these effects are inhibited by the S1P1R antagonist Ex 26 (30-300 nM) and the S1P3R antagonist TY-52156 (100-1000 nM), respectively. Confocal microscopy and western blot analysis confirmed the presence of S1P1R and S1P3R proteins in cortical glutamatergic synaptosomes, which were scarcely accessible to biotin in a biotinylation study. Then, we demonstrated that S1P1R and S1P3R densities and their release activity are amplified in cortical synaptosomes of mice suffering from experimental autoimmune encephalomyelitis (EAE), despite receptors maintain their preferential internal distribution. Receptor changes recover following chronic oral therapeutic FTY720 (0.03 mg/Kg/day). These results improve our knowledge of the role of presynaptic release-regulating S1P1Rs and S1P3Rs controlling glutamate transmission in the CNS also unravelling functional adaptations during EAE that recover following chronic FTY720. In a whole, these findings provide new information on the central neuroprotectant activities of FTY720.
Collapse
Affiliation(s)
- Alessandra Roggeri
- Department of Pharmacy (DiFar), University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (A.R.); (G.O.)
| | - Guendalina Olivero
- Department of Pharmacy (DiFar), University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (A.R.); (G.O.)
| | - Cesare Usai
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genoa, Italy;
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, B-3590 Hasselt, Belgium;
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Anna Pittaluga
- Department of Pharmacy (DiFar), Center of Excellence for Biomedical Research, 3Rs Center, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16145 Genoa, Italy
| |
Collapse
|
15
|
Sonsalla MM, Lamming DW. Geroprotective interventions in the 3xTg mouse model of Alzheimer's disease. GeroScience 2023; 45:1343-1381. [PMID: 37022634 PMCID: PMC10400530 DOI: 10.1007/s11357-023-00782-w] [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: 01/10/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative disease. As the population ages, the increasing prevalence of AD threatens massive healthcare costs in the coming decades. Unfortunately, traditional drug development efforts for AD have proven largely unsuccessful. A geroscience approach to AD suggests that since aging is the main driver of AD, targeting aging itself may be an effective way to prevent or treat AD. Here, we discuss the effectiveness of geroprotective interventions on AD pathology and cognition in the widely utilized triple-transgenic mouse model of AD (3xTg-AD) which develops both β-amyloid and tau pathologies characteristic of human AD, as well as cognitive deficits. We discuss the beneficial impacts of calorie restriction (CR), the gold standard for geroprotective interventions, and the effects of other dietary interventions including protein restriction. We also discuss the promising preclinical results of geroprotective pharmaceuticals, including rapamycin and medications for type 2 diabetes. Though these interventions and treatments have beneficial effects in the 3xTg-AD model, there is no guarantee that they will be as effective in humans, and we discuss the need to examine these interventions in additional animal models as well as the urgent need to test if some of these approaches can be translated from the lab to the bedside for the treatment of humans with AD.
Collapse
Affiliation(s)
- Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, 2500 Overlook Terrace, VAH C3127 Research 151, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, 2500 Overlook Terrace, VAH C3127 Research 151, Madison, WI, 53705, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA.
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
16
|
Sharma K, Dev KK. The Effects of Antipsychotics in Experimental Models of Krabbe Disease. Biomedicines 2023; 11:biomedicines11051313. [PMID: 37238985 DOI: 10.3390/biomedicines11051313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
The role of altered myelin in the onset and development of schizophrenia and changes in myelin due to antipsychotics remains unclear. Antipsychotics are D2 receptor antagonists, yet D2 receptor agonists increase oligodendrocyte progenitor numbers and limit oligodendrocyte injury. Conflicting studies suggest these drugs promote the differentiation of neural progenitors to oligodendrocyte lineage, while others report antipsychotics inhibit the proliferation and differentiation of oligodendrocyte precursors. Here, we utilised in-vitro (human astrocytes), ex-vivo (organotypic slice cultures) and in-vivo (twitcher mouse model) experimental study designs of psychosine-induced demyelination, a toxin that accumulates in Krabbe disease (KD), to investigate direct effects of antipsychotics on glial cell dysfunction and demyelination. Typical and atypical antipsychotics, and selective D2 and 5HT2A receptor antagonists, attenuated psychosine-induced cell viability, toxicity, and morphological aberrations in human astrocyte cultures. Haloperidol and clozapine reduced psychosine-induced demyelination in mouse organotypic cerebellar slices. These drugs also attenuated the effects of psychosine on astrocytes and microglia and restored non-phosphorylated neurofilament levels, indicating neuroprotective effects. In the demyelinating twitcher mouse model of KD, haloperidol improved mobility and significantly increased the survival of these animals. Overall, this study suggests that antipsychotics directly regulate glial cell dysfunction and exert a protective effect on myelin loss. This work also points toward the potential use of these pharmacological agents in KD.
Collapse
Affiliation(s)
- Kapil Sharma
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
| |
Collapse
|
17
|
Hashemi E, Yoseph E, Tsai HC, Moreno M, Yeh LH, Mehta SB, Kono M, Proia R, Han MH. Visualizing Sphingosine-1-Phosphate Receptor 1(S1P 1) Signaling During Central Nervous System De- and Remyelination. Cell Mol Neurobiol 2023; 43:1219-1236. [PMID: 35917044 PMCID: PMC10444542 DOI: 10.1007/s10571-022-01245-0] [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: 09/06/2021] [Accepted: 06/14/2022] [Indexed: 11/24/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory-demyelinating disease of the central nervous system (CNS) mediated by aberrant auto-reactive immune responses. The current immune-modulatory therapies are unable to protect and repair immune-mediated neural tissue damage. One of the therapeutic targets in MS is the sphingosine-1-phosphate (S1P) pathway which signals via sphingosine-1-phosphate receptors 1-5 (S1P1-5). S1P receptors are expressed predominantly on immune and CNS cells. Considering the potential neuroprotective properties of S1P signaling, we utilized S1P1-GFP (Green fluorescent protein) reporter mice in the cuprizone-induced demyelination model to investigate in vivo S1P - S1P1 signaling in the CNS. We observed S1P1 signaling in a subset of neural stem cells in the subventricular zone (SVZ) during demyelination. During remyelination, S1P1 signaling is expressed in oligodendrocyte progenitor cells in the SVZ and mature oligodendrocytes in the medial corpus callosum (MCC). In the cuprizone model, we did not observe S1P1 signaling in neurons and astrocytes. We also observed β-arrestin-dependent S1P1 signaling in lymphocytes during demyelination and CNS inflammation. Our findings reveal β-arrestin-dependent S1P1 signaling in oligodendrocyte lineage cells implying a role of S1P1 signaling in remyelination.
Collapse
Affiliation(s)
- Ezzat Hashemi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA
| | - Ezra Yoseph
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA
| | - Hsing-Chuan Tsai
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA
| | - Monica Moreno
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA
| | - Li-Hao Yeh
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Mari Kono
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Richard Proia
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - May H Han
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA.
| |
Collapse
|
18
|
Konen FF, Möhn N, Witte T, Schefzyk M, Wiestler M, Lovric S, Hufendiek K, Schwenkenbecher P, Sühs KW, Friese MA, Klotz L, Pul R, Pawlitzki M, Hagin D, Kleinschnitz C, Meuth SG, Skripuletz T. Treatment of autoimmunity: The impact of disease-modifying therapies in multiple sclerosis and comorbid autoimmune disorders. Autoimmun Rev 2023; 22:103312. [PMID: 36924922 DOI: 10.1016/j.autrev.2023.103312] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
More than 10 disease-modifying therapies (DMT) are approved by the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) for the treatment of multiple sclerosis (MS) and new therapeutic options are on the horizon. Due to different underlying therapeutic mechanisms, a more individualized selection of DMTs in MS is possible, taking into account the patient's current situation. Therefore, concomitant treatment of various comorbid conditions, including autoimmune mediated disorders such as rheumatoid arthritis, should be considered in MS patients. Because the pathomechanisms of autoimmunity partially overlap, DMT could also treat concomitant inflammatory diseases and simplify the patient's treatment. In contrast, the exacerbation and even new occurrence of several autoimmune diseases have been reported as a result of immunomodulatory treatment of MS. To simplify treatment and avoid disease exacerbation, knowledge of the beneficial and adverse effects of DMT in other autoimmune disorders is critical. Therefore, we conducted a literature search and described the beneficial and adverse effects of approved and currently studied DMT in a large number of comorbid autoimmune diseases, including rheumatoid arthritis, ankylosing spondylitis, inflammatory bowel diseases, cutaneous disorders including psoriasis, Sjögren´s syndrome, systemic lupus erythematosus, systemic vasculitis, autoimmune hepatitis, and ocular autoimmune disorders. Our review aims to facilitate the selection of an appropriate DMT in patients with MS and comorbid autoimmune diseases.
Collapse
Affiliation(s)
- Franz Felix Konen
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Torsten Witte
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, 30625 Hannover, Germany..
| | - Matthias Schefzyk
- Department of Dermatology, Allergology and Venerology, Hannover Medical School, 30625 Hannover, Germany..
| | - Miriam Wiestler
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany.
| | - Svjetlana Lovric
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany.
| | - Karsten Hufendiek
- University Eye Hospital, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Kurt-Wolfram Sühs
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany.
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany.
| | - Refik Pul
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen 45147, Germany.
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, University of Tel Aviv, 6 Weizmann St., Tel-Aviv 6423906, Israel.
| | - Christoph Kleinschnitz
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen 45147, Germany.
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| |
Collapse
|
19
|
van Echten-Deckert G. The role of sphingosine 1-phosphate metabolism in brain health and disease. Pharmacol Ther 2023; 244:108381. [PMID: 36907249 DOI: 10.1016/j.pharmthera.2023.108381] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023]
Abstract
Lipids are essential structural and functional components of the central nervous system (CNS). Sphingolipids are ubiquitous membrane components which were discovered in the brain in the late 19th century. In mammals, the brain contains the highest concentration of sphingolipids in the body. Sphingosine 1-phosphate (S1P) derived from membrane sphingolipids evokes multiple cellular responses which, depending on its concentration and localization, make S1P a double-edged sword in the brain. In the present review we highlight the role of S1P in brain development and focus on the often contrasting findings regarding its contributions to the initiation, progression and potential recovery of different brain pathologies, including neurodegeneration, multiple sclerosis (MS), brain cancers, and psychiatric illnesses. A detailed understanding of the critical implications of S1P in brain health and disease may open the door for new therapeutic options. Thus, targeting S1P-metabolizing enzymes and/or signaling pathways might help overcome, or at least ameliorate, several brain illnesses.
Collapse
|
20
|
Singh P, Singh D, Srivastava P, Mishra G, Tiwari AK. Evaluation of advanced, pathophysiologic new targets for imaging of CNS. Drug Dev Res 2023; 84:484-513. [PMID: 36779375 DOI: 10.1002/ddr.22040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/12/2022] [Accepted: 12/31/2022] [Indexed: 02/14/2023]
Abstract
The inadequate information about the in vivo pathological, physiological, and neurological impairments, as well as the absence of in vivo tools for assessing brain penetrance and the efficiency of newly designed drugs, has hampered the development of new techniques for the treatment for variety of new central nervous system (CNS) diseases. The searching sites such as Science Direct and PubMed were used to find out the numerous distinct tracers across 16 CNS targets including tau, synaptic vesicle glycoprotein, the adenosine 2A receptor, the phosphodiesterase enzyme PDE10A, and the purinoceptor, among others. Among the most encouraging are [18 F]FIMX for mGluR imaging, [11 C]Martinostat for Histone deacetylase, [18 F]MNI-444 for adenosine 2A imaging, [11 C]ER176 for translocator protein, and [18 F]MK-6240 for tau imaging. We also reviewed the findings for each tracer's features and potential for application in CNS pathophysiology and therapeutic evaluation investigations, including target specificity, binding efficacy, and pharmacokinetic factors. This review aims to present a current evaluation of modern positron emission tomography tracers for CNS targets, with a focus on recent advances for targets that have newly emerged for imaging in humans.
Collapse
Affiliation(s)
- Priya Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Deepika Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Pooja Srivastava
- Division of Cyclotron and Radiopharmaceuticals Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Gauri Mishra
- Department of Zoology, Swami Shraddhananad College, University of Delhi, Alipur, Delhi, India
| | - Anjani K Tiwari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
21
|
Alessenko AV, Gutner UA, Shupik MA. Involvement of Lipids in the Pathogenesis of Amyotrophic Lateral Sclerosis. Life (Basel) 2023; 13:life13020510. [PMID: 36836867 PMCID: PMC9966871 DOI: 10.3390/life13020510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/26/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive degeneration of upper and lower motor neurons. To study its underlying mechanisms, a variety of models are currently used at the cellular level and in animals with mutations in multiple ALS associated genes, including SOD1, C9ORF72, TDP-43, and FUS. Key mechanisms involved in the disease include excitotoxicity, oxidative stress, mitochondrial dysfunction, neuroinflammatory, and immune reactions. In addition, significant metabolism alterations of various lipids classes, including phospholipids, fatty acids, sphingolipids, and others have been increasingly recognized. Recently, the mechanisms of programmed cell death (apoptosis), which may be responsible for the degeneration of motor neurons observed in the disease, have been intensively studied. In this context, sphingolipids, which are the most important sources of secondary messengers transmitting signals for cell proliferation, differentiation, and apoptosis, are gaining increasing attention in the context of ALS pathogenesis given their role in the development of neuroinflammatory and immune responses. This review describes changes in lipids content and activity of enzymes involved in their metabolism in ALS, both summarizing current evidence from animal models and clinical studies and discussing the potential of new drugs among modulators of lipid metabolism enzymes.
Collapse
|
22
|
Pepe G, Capocci L, Marracino F, Realini N, Lenzi P, Martinello K, Bovier TF, Bichell TJ, Scarselli P, Di Cicco C, Bowman AB, Digilio FA, Fucile S, Fornai F, Armirotti A, Parlato R, Di Pardo A, Maglione V. Treatment with THI, an inhibitor of sphingosine-1-phosphate lyase, modulates glycosphingolipid metabolism and results therapeutically effective in experimental models of Huntington's disease. Mol Ther 2023; 31:282-299. [PMID: 36116006 PMCID: PMC9840122 DOI: 10.1016/j.ymthe.2022.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/05/2022] [Accepted: 09/06/2022] [Indexed: 02/06/2023] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder with no effective cure currently available. Over the past few years our research has shown that alterations in sphingolipid metabolism represent a critical determinant in HD pathogenesis. In particular, aberrant metabolism of sphingosine-1-phosphate (S1P) has been reported in multiple disease settings, including human postmortem brains from HD patients. In this study, we investigate the potential therapeutic effect of the inhibition of S1P degradative enzyme SGPL1, by the chronic administration of the 2-acetyl-5-tetrahydroxybutyl imidazole (THI) inhibitor. We show that THI mitigated motor dysfunctions in both mouse and fly models of HD. The compound evoked the activation of pro-survival pathways, normalized levels of brain-derived neurotrophic factor, preserved white matter integrity, and stimulated synaptic functions in HD mice. Metabolically, THI restored normal levels of hexosylceramides and stimulated the autophagic and lysosomal machinery, facilitating the reduction of nuclear inclusions of both wild-type and mutant huntingtin proteins.
Collapse
Affiliation(s)
| | | | | | - Natalia Realini
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | | | - Tiziana Francesca Bovier
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; Department of Pediatrics Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York 10032, NY, USA
| | - Terry Jo Bichell
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA
| | | | | | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - Filomena A Digilio
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Sergio Fucile
- IRCCS Neuromed, Pozzilli (IS) 86077, Italy; Department of Physiology and Pharmacology, Sapienza Rome University, Rome 00185, Italy
| | - Francesco Fornai
- IRCCS Neuromed, Pozzilli (IS) 86077, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Rosanna Parlato
- Division for Neurodegenerative Diseases, Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim Heidelberg University, Mannheim 68167, Germany
| | | | | |
Collapse
|
23
|
Basavarajappa D, Gupta V, Wall RV, Gupta V, Chitranshi N, Mirshahvaladi SSO, Palanivel V, You Y, Mirzaei M, Klistorner A, Graham SL. S1PR1 signaling attenuates apoptosis of retinal ganglion cells via modulation of cJun/Bim cascade and Bad phosphorylation in a mouse model of glaucoma. FASEB J 2023; 37:e22710. [PMID: 36520045 DOI: 10.1096/fj.202201346r] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/09/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022]
Abstract
Glaucoma is a complex neurodegenerative disease characterized by optic nerve damage and apoptotic retinal ganglion cell (RGC) death, and is the leading cause of irreversible blindness worldwide. Among the sphingosine 1-phosphate receptors (S1PRs) family, S1PR1 is a highly expressed subtype in the central nervous system and has gained rapid attention as an important mediator of pathophysiological processes in the brain and the retina. Our recent study showed that mice treated orally with siponimod drug exerted neuroprotection via modulation of neuronal S1PR1 in experimental glaucoma. This study identified the molecular signaling pathway modulated by S1PR1 activation with siponimod treatment in RGCs in glaucomatous injury. We investigated the critical neuroprotective signaling pathway in vivo using mice deleted for S1PR1 in RGCs. Our results showed marked upregulation of the apoptotic pathway was associated with decreased Akt and Erk1/2 activation levels in the retina in glaucoma conditions. Activation of S1PR1 with siponimod treatment significantly increased neuroprotective Akt and Erk1/2 activation and attenuated the apoptotic signaling via suppression of c-Jun/Bim cascade and by increasing Bad phosphorylation. Conversely, deletion of S1PR1 in RGCs significantly increased the apoptotic cells in the ganglion cell layer in glaucoma and diminished the neuroprotective effects of siponimod treatment on Akt/Erk1/2 activation, c-Jun/Bim cascade, and Bad phosphorylation. Our data demonstrated that activation of S1PR1 in RGCs induces crucial neuroprotective signaling that suppresses the proapoptotic c-Jun/Bim cascade and increases antiapoptotic Bad phosphorylation. Our findings suggest that S1PR1 is a potential therapeutic target for neuroprotection of RGCs in glaucoma.
Collapse
Affiliation(s)
- Devaraj Basavarajappa
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| | - Roshana Vander Wall
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| | - Seyed Shahab Oddin Mirshahvaladi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| | - Viswanthram Palanivel
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| | - Yuyi You
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| | - Alexander Klistorner
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| | - Stuart L Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, New South Wales, Australia
| |
Collapse
|
24
|
Basavarajappa D, Gupta V, Chitranshi N, Wall R, Rajput R, Pushpitha K, Sharma S, Mirzaei M, Klistorner A, Graham S. Siponimod exerts neuroprotective effects on the retina and higher visual pathway through neuronal S1PR1 in experimental glaucoma. Neural Regen Res 2023; 18:840-848. [DOI: 10.4103/1673-5374.344952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
25
|
Wang X, Huang R, Huang B, Li X. S1PR2 Regulates Autophagy Through the AKT/mTOR Pathway to Promote Pathological Damage in Alzheimer's Disease. J Alzheimers Dis 2023; 96:1489-1504. [PMID: 38007654 DOI: 10.3233/jad-230533] [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: 11/27/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a fatal and debilitating neurodegenerative disease. Sphingosine-1-phosphate receptor 2 (S1PR2), one of the receptors of S1P, is a key regulatory factor for various diseases. OBJECTIVE This study aimed to explore the role and possible mechanism of S1PR2 in AD. METHODS S1PR2 expression in the AD mice was detected, and after intervening S1PR2 expression with sh-S1PR2 in AD mice, the behavioral changes, pathological lesions of the hippocampus, autophagy level, and AKT/mTOR pathway activation were analyzed. Furthermore, SH-SY5Y cells were induced by Aβ25-35 to construct an AD cell model, and the effects of sh-S1PR2 on proliferation, apoptosis, autophagy, and AKT/mTOR pathway of AD cells were investigated. In addition, the effects of pathway inhibitor rapamycin on model cells were further analyzed. RESULTS The expression of S1PR2 was significantly increased in AD mice, the sh-S1PR2 significantly improved behavioral dysfunction, alleviated pathological injury of the hippocampus, increased the number of neurons, and inhibited Aβ production and p-tau expression, showing a positive effect on the AD pathology. In addition, silencing of S1PR2 expression significantly promoted the autophagy level and inhibited the activation of the AKT/mTOR pathway in AD model mice. In vitro experiments further confirmed that sh-S1PR2 promoted cell proliferation, inhibited apoptosis, relieved cytopathology, promoted autophagy, and inhibited the activation of the AKT/mTOR pathway in the cell model. The use of rapamycin further confirmed the role of AKT/mTOR pathway-mediated autophagy in the regulation of AD by S1PR2. CONCLUSION S1PR2 promoted AD pathogenesis by inhibiting autophagy through the activation of AKT/mTOR pathway.
Collapse
Affiliation(s)
- Xiaoping Wang
- Department of Neurology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Sichuan, China
| | - Rui Huang
- Department of Neurology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Sichuan, China
| | - Bin Huang
- Department of Neurology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Sichuan, China
| | - Xiaojia Li
- Department of Neurology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Sichuan, China
| |
Collapse
|
26
|
Ozanimod as a novel oral small molecule therapy for the treatment of Crohn's disease: The YELLOWSTONE clinical trial program. Contemp Clin Trials 2022; 122:106958. [PMID: 36208720 PMCID: PMC10008122 DOI: 10.1016/j.cct.2022.106958] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/29/2022] [Accepted: 10/01/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Ozanimod, an oral sphingosine 1-phosphate receptor modulator currently approved for the treatment of moderately to severely active ulcerative colitis and relapsing multiple sclerosis, showed clinical, endoscopic, and histological benefit in the phase 2 STEPSTONE trial for Crohn's disease (CD). We aim to describe the trial design of the YELLOWSTONE phase 3 program evaluating the safety and efficacy of ozanimod in patients with moderately to severely active CD. METHODS The YELLOWSTONE program consists of phase 3, randomized, double-blind, placebo-controlled induction (NCT03440372 and NCT03440385) and maintenance (NCT03464097) trials and an open-label extension (OLE) study (NCT03467958). Patients with inadequate response or intolerance to ≥1 CD treatment are randomized to receive daily ozanimod 0.92 mg (equivalent to ozanimod HCl 1 mg) or placebo for 12 weeks during induction. Those who respond to ozanimod are rerandomized to continue ozanimod or placebo maintenance therapy for 52 weeks. Patients who do not meet criteria for maintenance, experience relapse during maintenance, or complete maintenance or ≥ 1 year of STEPSTONE are eligible for open-label treatment for up to 234 weeks. Efficacy endpoints include clinical, endoscopic, and histologic outcomes. RESULTS Expected 2023 (induction studies), 2024 (maintenance study), and 2026 (OLE). CONCLUSION YELLOWSTONE will provide pivotal phase 3 data on the safety and efficacy of ozanimod in patients with moderately to severely active CD using state-of-the-art methods, including centrally read endoscopic and histologic measurements, along with subjective assessments of symptom control based on the Crohn's Disease Activity Index. These studies could enable approval of ozanimod as a new CD therapy. CLINICAL TRIAL REGISTRATION NUMBERS NCT03440372, NCT03440385, NCT03464097, NCT03467958.
Collapse
|
27
|
Zhuo C, Zhao F, Tian H, Chen J, Li Q, Yang L, Ping J, Li R, Wang L, Xu Y, Cai Z, Song X. Acid sphingomyelinase/ceramide system in schizophrenia: implications for therapeutic intervention as a potential novel target. Transl Psychiatry 2022; 12:260. [PMID: 35739089 PMCID: PMC9226132 DOI: 10.1038/s41398-022-01999-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/09/2022] Open
Abstract
Schizophrenia is a severe mental illness, as the efficacies of current antipsychotic medications are far from satisfactory. An improved understanding of the signaling molecules involved in schizophrenia may provide novel therapeutic targets. Acid sphingomyelinase (ASM) catalyzes cellular membrane sphingomyelin into ceramide, which is further metabolized into sphingosine-1-phophate (S1P). ASM, ceramide, and S1P at the cell surface exert critical roles in the regulation of biophysical processes that include proliferation, apoptosis, and inflammation, and are thereby considered important signaling molecules. Although research on the ASM/ceramide system is still in its infancy, structural and metabolic abnormalities have been demonstrated in schizophrenia. ASM/ceramide system dysfunction is linked to the two important models of schizophrenia, the dopamine (DA) hypothesis through affecting presynaptic DA signaling, and the vulnerability-stress-inflammation model that includes the contribution of stress on the basis of genetic predisposition. In this review, we highlight the current knowledge of ASM/ceramide system dysfunction in schizophrenia gained from human and animal studies, and formulate future directions from the biological landscape for the development of new treatments. Collectively, these discoveries suggest that aberrations in the ASM/ceramide system, especially in ASM activity and levels of ceramide and S1P, may alter cerebral microdomain structure and neuronal metabolism, leading to neurotransmitter (e.g., DA) dysfunction and neuroinflammation. As such, the ASM/ceramide system may offer therapeutic targets for novel medical interventions. Normalization of the aberrant ASM/ceramide system or ceramide reduction by using approved functional inhibitors of ASM, such as fluvoxamine and rosuvastatin, may improve clinical outcomes of patients with schizophrenia. These transformative findings of the ASM/ceramide system in schizophrenia, although intriguing and exciting, may pose scientific questions and challenges that will require further studies for their resolution.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Key Laboratory of Real Time Tracing Brain Circuit, Tianjin Medical Affiliated Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Fourth Hospital, 300140, Tianjin, China. .,The key Laboratory of Psychiatric-Neuroimaging-Genetics and Comorbidity (PNGC_Lab) of Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, 300222, Tianjin, China. .,Brain Micro-imaging Center of Psychiatric Animal Model, Wenzhou Seventh Peoples Hospital, 325000, Wenzhou, China. .,Department of Psychiatry, The Fourth Center Hospital of Tianjin Medical University, 300222, Tianjin, China. .,Key Laboratory of the Macro-Brain Neuroimaging Center of Animal Model, Wenzhou Seventh Peoples Hospital, 325000, Wenzhou, China. .,Department of Psychiatry, The First Hospital of Shanxi Medical University, 03000, Taiyuan, China. .,Department of Psychiatry, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
| | - Feifei Zhao
- Key Laboratory of the Macro-Brain Neuroimaging Center of Animal Model, Wenzhou Seventh Peoples Hospital, 325000 Wenzhou, China
| | - Hongjun Tian
- grid.265021.20000 0000 9792 1228Department of Psychiatry, The Fourth Center Hospital of Tianjin Medical University, 300222 Tianjin, China
| | - Jiayue Chen
- grid.265021.20000 0000 9792 1228Department of Psychiatry, The Fourth Center Hospital of Tianjin Medical University, 300222 Tianjin, China
| | - Qianchen Li
- grid.265021.20000 0000 9792 1228Department of Psychiatry, The Fourth Center Hospital of Tianjin Medical University, 300222 Tianjin, China
| | - Lei Yang
- grid.265021.20000 0000 9792 1228Department of Psychiatry, The Fourth Center Hospital of Tianjin Medical University, 300222 Tianjin, China
| | - Jing Ping
- Key Laboratory of the Macro-Brain Neuroimaging Center of Animal Model, Wenzhou Seventh Peoples Hospital, 325000 Wenzhou, China
| | - Ranli Li
- Key Laboratory of the Macro-Brain Neuroimaging Center of Animal Model, Wenzhou Seventh Peoples Hospital, 325000 Wenzhou, China
| | - Lina Wang
- Key Laboratory of the Macro-Brain Neuroimaging Center of Animal Model, Wenzhou Seventh Peoples Hospital, 325000 Wenzhou, China
| | - Yong Xu
- grid.452461.00000 0004 1762 8478Department of Psychiatry, The First Hospital of Shanxi Medical University, 03000 Taiyuan, China
| | - Ziyao Cai
- Key Laboratory of the Macro-Brain Neuroimaging Center of Animal Model, Wenzhou Seventh Peoples Hospital, 325000 Wenzhou, China
| | - Xueqin Song
- Department of Psychiatry, First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
| |
Collapse
|
28
|
Pournajaf S, Dargahi L, Javan M, Pourgholami MH. Molecular Pharmacology and Novel Potential Therapeutic Applications of Fingolimod. Front Pharmacol 2022; 13:807639. [PMID: 35250559 PMCID: PMC8889014 DOI: 10.3389/fphar.2022.807639] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Fingolimod is a well-tolerated, highly effective disease-modifying therapy successfully utilized in the management of multiple sclerosis. The active metabolite, fingolimod-phosphate, acts on sphingosine-1-phosphate receptors (S1PRs) to bring about an array of pharmacological effects. While being initially recognized as a novel agent that can profoundly reduce T-cell numbers in circulation and the CNS, thereby suppressing inflammation and MS, there is now rapidly increasing knowledge on its previously unrecognized molecular and potential therapeutic effects in diverse pathological conditions. In addition to exerting inhibitory effects on sphingolipid pathway enzymes, fingolimod also inhibits histone deacetylases, transient receptor potential cation channel subfamily M member 7 (TRMP7), cytosolic phospholipase A2α (cPLA2α), reduces lysophosphatidic acid (LPA) plasma levels, and activates protein phosphatase 2A (PP2A). Furthermore, fingolimod induces apoptosis, autophagy, cell cycle arrest, epigenetic regulations, macrophages M1/M2 shift and enhances BDNF expression. According to recent evidence, fingolimod modulates a range of other molecular pathways deeply rooted in disease initiation or progression. Experimental reports have firmly associated the drug with potentially beneficial therapeutic effects in immunomodulatory diseases, CNS injuries, and diseases including Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, and even cancer. Attractive pharmacological effects, relative safety, favorable pharmacokinetics, and positive experimental data have collectively led to its testing in clinical trials. Based on the recent reports, fingolimod may soon find its way as an adjunct therapy in various disparate pathological conditions. This review summarizes the up-to-date knowledge about molecular pharmacology and potential therapeutic uses of fingolimod.
Collapse
Affiliation(s)
- Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
29
|
Fagan SG, Bechet S, Dev KK. Fingolimod Rescues Memory and Improves Pathological Hallmarks in the 3xTg-AD Model of Alzheimer's Disease. Mol Neurobiol 2022; 59:1882-1895. [PMID: 35031916 PMCID: PMC8882098 DOI: 10.1007/s12035-021-02613-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/19/2021] [Indexed: 10/26/2022]
Abstract
Therapeutic strategies for Alzheimer's disease (AD) have largely focused on the regulation of amyloid pathology while those targeting tau pathology, and inflammatory mechanisms are less explored. In this regard, drugs with multimodal and concurrent targeting of Aβ, tau, and inflammatory processes may offer advantages. Here, we investigate one such candidate drug in the triple transgenic 3xTg-AD mouse model of AD, namely the disease-modifying oral neuroimmunomodulatory therapeutic used in patients with multiple sclerosis, called fingolimod. In this study, administration of fingolimod was initiated after behavioral symptoms are known to emerge, at 6 months of age. Treatment continued to 12 months when behavioral tests were performed and thereafter histological and biochemical analysis was conducted on postmortem tissue. The results demonstrate that fingolimod reverses deficits in spatial working memory at 8 and 12 months of age as measured by novel object location and Morris water maze tests. Inflammation in the brain is alleviated as demonstrated by reduced Iba1-positive and CD3-positive cell number, less ramified microglial morphology, and improved cytokine profile. Finally, treatment with fingolimod was shown to reduce phosphorylated tau and APP levels in the hippocampus and cortex. These results highlight the potential of fingolimod as a multimodal therapeutic for the treatment of AD.
Collapse
Affiliation(s)
- Steven G Fagan
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland.
| | - Sibylle Bechet
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
30
|
Gutner UA, Shupik MA. The Role of Sphingosine-1-Phosphate in Neurodegenerative Diseases. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2021. [DOI: 10.1134/s1068162021050277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Nasir G, Chopra R, Elwood F, Ahmed SS. Krabbe Disease: Prospects of Finding a Cure Using AAV Gene Therapy. Front Med (Lausanne) 2021; 8:760236. [PMID: 34869463 PMCID: PMC8633897 DOI: 10.3389/fmed.2021.760236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/15/2021] [Indexed: 11/13/2022] Open
Abstract
Krabbe Disease (KD) is an autosomal metabolic disorder that affects both the central and peripheral nervous systems. It is caused by a functional deficiency of the lysosomal enzyme, galactocerebrosidase (GALC), resulting in an accumulation of the toxic metabolite, psychosine. Psychosine accumulation affects many different cellular pathways, leading to severe demyelination. Although there is currently no effective therapy for Krabbe disease, recent gene therapy-based approaches in animal models have indicated a promising outlook for clinical treatment. This review highlights recent findings in the pathogenesis of Krabbe disease, and evaluates AAV-based gene therapy as a promising strategy for treating this devastating pediatric disease.
Collapse
Affiliation(s)
- Gibran Nasir
- Department of Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA, United States
| | - Rajiv Chopra
- AllianThera Biopharma, Boston, MA, United States
| | - Fiona Elwood
- Department of Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA, United States
| | - Seemin S Ahmed
- Department of Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA, United States
| |
Collapse
|
32
|
Chatzikonstantinou S, Poulidou V, Arnaoutoglou M, Kazis D, Heliopoulos I, Grigoriadis N, Boziki M. Signaling through the S1P-S1PR Axis in the Gut, the Immune and the Central Nervous System in Multiple Sclerosis: Implication for Pathogenesis and Treatment. Cells 2021; 10:cells10113217. [PMID: 34831439 PMCID: PMC8626013 DOI: 10.3390/cells10113217] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 01/14/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is a signaling molecule with complex biological functions that are exerted through the activation of sphingosine 1-phosphate receptors 1–5 (S1PR1–5). S1PR expression is necessary for cell proliferation, angiogenesis, neurogenesis and, importantly, for the egress of lymphocytes from secondary lymphoid organs. Since the inflammatory process is a key element of immune-mediated diseases, including multiple sclerosis (MS), S1PR modulators are currently used to ameliorate systemic immune responses. The ubiquitous expression of S1PRs by immune, intestinal and neural cells has significant implications for the regulation of the gut–brain axis. The dysfunction of this bidirectional communication system may be a significant factor contributing to MS pathogenesis, since an impaired intestinal barrier could lead to interaction between immune cells and microbiota with a potential to initiate abnormal local and systemic immune responses towards the central nervous system (CNS). It appears that the secondary mechanisms of S1PR modulators affecting the gut immune system, the intestinal barrier and directly the CNS, are coordinated to promote therapeutic effects. The scope of this review is to focus on S1P−S1PR functions in the cells of the CNS, the gut and the immune system with particular emphasis on the immunologic effects of S1PR modulation and its implication in MS.
Collapse
Affiliation(s)
- Simela Chatzikonstantinou
- 3rd Department of Neurology, Aristotle University of Thessaloniki, “G.Papanikolaou” Hospital, Leoforos Papanikolaou, Exohi, 57010 Thessaloniki, Greece; (S.C.); (D.K.)
| | - Vasiliki Poulidou
- 1st Department of Neurology, Aristotle University of Thessaloniki, AHEPA Hospital, 1, Stilp Kyriakidi st., 54636 Thessaloniki, Greece; (V.P.); (M.A.)
| | - Marianthi Arnaoutoglou
- 1st Department of Neurology, Aristotle University of Thessaloniki, AHEPA Hospital, 1, Stilp Kyriakidi st., 54636 Thessaloniki, Greece; (V.P.); (M.A.)
| | - Dimitrios Kazis
- 3rd Department of Neurology, Aristotle University of Thessaloniki, “G.Papanikolaou” Hospital, Leoforos Papanikolaou, Exohi, 57010 Thessaloniki, Greece; (S.C.); (D.K.)
| | - Ioannis Heliopoulos
- Department of Neurology, University General Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupoli, Greece;
| | - Nikolaos Grigoriadis
- Multiple Sclerosis Center, Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, Aristotle University of Thessaloniki, AHEPA Hospital, 1, Stilp Kyriakidi st., 54636 Thessaloniki, Greece;
| | - Marina Boziki
- Multiple Sclerosis Center, Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, Aristotle University of Thessaloniki, AHEPA Hospital, 1, Stilp Kyriakidi st., 54636 Thessaloniki, Greece;
- Correspondence:
| |
Collapse
|
33
|
Mechanism-based criteria to improve therapeutic outcomes in progressive multiple sclerosis. Nat Rev Neurol 2021; 18:40-55. [PMID: 34732831 DOI: 10.1038/s41582-021-00581-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 02/07/2023]
Abstract
In contrast to the multiple disease-modifying therapies that are available for relapsing-remitting multiple sclerosis (MS), the therapeutic options for progressive MS (PMS) are limited. Recent advances in our understanding of the neuroimmunology of PMS, including the mechanisms that drive slowly expanding lesions, have fuelled optimism for improved treatment of this condition. In this Review, we highlight the commonly observed neuropathology of PMS and discuss the associated mechanisms of CNS injury. We then apply this knowledge to formulate criteria for therapeutic efficacy in PMS, beginning with the need for early treatment owing to the substantial neuropathology that is already present at the initial clinical presentation. Other requirements include: antagonism of neuroaxonal injury mediators such as pro-inflammatory microglia and lymphocytes; remediation of oxidative stress resulting from iron deposition and mitochondrial dysfunction; and promotion of neuroprotection through remyelination. We consider whether current disease-modifying therapies for relapsing-remitting MS meet the criteria for successful therapeutics in PMS and suggest that the evidence favours the early introduction of sphingosine 1-phosphate receptor modulators. Finally, we weigh up emerging medications, including repurposed generic medications and Bruton's tyrosine kinase inhibitors, against these fundamental criteria. In this new therapeutic era in PMS, success depends collectively on understanding disease mechanisms, drug characteristics (including brain penetration) and rational use.
Collapse
|
34
|
Central Nervous System Tissue Regeneration after Intracerebral Hemorrhage: The Next Frontier. Cells 2021; 10:cells10102513. [PMID: 34685493 PMCID: PMC8534252 DOI: 10.3390/cells10102513] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Despite marked advances in surgical techniques and understanding of secondary brain injury mechanisms, the prognosis of intracerebral hemorrhage (ICH) remains devastating. Harnessing and promoting the regenerative potential of the central nervous system may improve the outcomes of patients with hemorrhagic stroke, but approaches are still in their infancy. In this review, we discuss the regenerative phenomena occurring in animal models and human ICH, provide results related to cellular and molecular mechanisms of the repair process including by microglia, and review potential methods to promote tissue regeneration in ICH. We aim to stimulate research involving tissue restoration after ICH.
Collapse
|
35
|
Alessenko AV, Gutner UA, Nebogatikov VO, Shupik MA, Ustyugov AA. [The role of sphingolipids in pathogenesis of amyotrophic lateral sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:131-140. [PMID: 34481449 DOI: 10.17116/jnevro2021121081131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease characterized by selective degeneration of motor neurons of the spinal cord and motor cortex and brain stem. The key features of the course of this disease are excitotoxicity, oxidative stress, mitochondrial dysfunction, neuro-inflammatory and immune reactions. Recently, the mechanisms of programmed cell death (apoptosis), which may be responsible for the degeneration of motor neurons in this disease, have been intensively studied. In this regard, sphingolipids, which are the most important sources of secondary messengers that transmit cell proliferation, differentiation and apoptosis signals, and are involved in the development of neuroinflammatory and immune responses, are of particular interest in the context of ALS pathogenesis. The review provides information from domestic and foreign authors on the involvement of various sphingolipids (sphingomyelins, ceramides, sphingosine, sphinganin, sphingosine-1-phosphate, galactosylceramides, glucosylceramides, gangliosides) in the development of pro-inflammatory reactions and apoptosis of motor neurons in ALS. The authors discuss the prospects of using new drugs that control the metabolism of sphingolipids for the treatment of ALS.
Collapse
Affiliation(s)
| | - U A Gutner
- Institute of Biochemical Physic, Moscow, Russia
| | - V O Nebogatikov
- Institute of Physiologically Active Compounds, Chernogolovka, Russia
| | - M A Shupik
- Institute of Biochemical Physic, Moscow, Russia
| | - A A Ustyugov
- Institute of Physiologically Active Compounds, Chernogolovka, Russia
| |
Collapse
|
36
|
Brummer T, Ruck T, Meuth SG, Zipp F, Bittner S. Treatment approaches to patients with multiple sclerosis and coexisting autoimmune disorders. Ther Adv Neurol Disord 2021; 14:17562864211035542. [PMID: 34457039 PMCID: PMC8388232 DOI: 10.1177/17562864211035542] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/08/2021] [Indexed: 12/30/2022] Open
Abstract
The past decades have yielded major therapeutic advances in many autoimmune conditions - such as multiple sclerosis (MS) - and thus ushered in a new era of more targeted and increasingly potent immunotherapies. Yet this growing arsenal of therapeutic immune interventions has also rendered therapy much more challenging for the attending physician, especially when treating patients with more than one autoimmune condition. Importantly, some therapeutic strategies are either approved for several autoimmune disorders or may be repurposed for other conditions, therefore opening new curative possibilities in related fields. In this article, we especially focus on frequent and therapeutically relevant concomitant autoimmune conditions faced by neurologists when treating patients with MS, namely psoriasis, rheumatoid arthritis and inflammatory bowel diseases. We provide an overview of the available disease-modifying therapies, highlight possible contraindications, show pathophysiological overlaps and finally present which therapeutics can be utilized as a combinatory treatment, in order to 'kill two birds with one stone'.
Collapse
Affiliation(s)
- Tobias Brummer
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Tobias Ruck
- Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sven G. Meuth
- Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, Rhineland-Palatinate, Mainz 55131, Germany
| |
Collapse
|
37
|
Wang M, Wu H, Wang R, Dai X, Deng R, Wang Y, Bu Y, Sun M, Zhang H. Inhibition of sphingosine 1-phosphate (S1P) receptor 1/2/3 ameliorates biological dysfunction in rheumatoid arthritis fibroblast-like synoviocyte MH7A cells through Gαi/Gαs rebalancing. Clin Exp Pharmacol Physiol 2021; 48:1080-1089. [PMID: 33495999 DOI: 10.1111/1440-1681.13460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/25/2020] [Indexed: 11/29/2022]
Abstract
Sphingosine 1-phosphate (S1P) exerts its various physiological and pathological effects by interacting with G protein-coupled receptors. In addition, S1P can induce biological dysfunction in fibroblast-like synoviocytes (FLSs) in the development of rheumatoid arthritis (RA). However, the mechanism underlying this S1P-induced dysfunction remains unclear. An imbalance between Gαi and Gαs can affect the level of cAMP, an important regulator of numerous cell functions. Therefore, we studied the effects of S1P receptor (S1PR) 1-, 2-, and 3-associated Gαi/Gαs imbalance on the biological function of rheumatoid arthritis fibroblast-like synoviocyte (MH7A cells). The results showed that blocking S1PR1/3 and Gαi, and activating Gαs, inhibited the proliferation, migration, invasion, and proinflammatory cytokine release of MH7A cells in a S1P-induced inflammation model, whereas suppressing S1PR2 only affected the invasion and the release of proinflammatory cytokines of these cells. Analysis of the expression of S1PR1/2/3 and Gαi/Gαs further showed that S1PR1/2/3 could regulate the Gαi/Gαs balance. Furthermore, our data suggested that the level of cAMP was also affected. Combined, our results showed that impaired S1PR1/2/3 signalling can affect MH7A cells biological function via Gαi/Gαs-cAMP signalling, which can provide a new idea for the treatment of RA.
Collapse
Affiliation(s)
- Mengdie Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R & D of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R & D of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Ronghui Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R & D of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Xuejing Dai
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R & D of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R & D of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R & D of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Yanhong Bu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R & D of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Minghui Sun
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R & D of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Heng Zhang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R & D of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| |
Collapse
|
38
|
Lu S, She M, Zeng Q, Yi G, Zhang J. Sphingosine 1-phosphate and its receptors in ischemia. Clin Chim Acta 2021; 521:25-33. [PMID: 34153277 DOI: 10.1016/j.cca.2021.06.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 10/21/2022]
Abstract
Sphingosine 1-phosphate (S1P), a metabolite of sphingolipids, is mainly derived from red blood cells (RBCs), platelets and endothelial cells (ECs). It plays important roles in regulating cell survival, vascular integrity and inflammatory responses through its receptors. S1P receptors (S1PRs), including 5 subtypes (S1PR1-5), are G protein-coupled receptors and have been proved to mediate various and complex roles of S1P in atherosclerosis, myocardial infarction (MI) and ischemic stroke by regulating endothelial function and inflammatory response as well as immune cell behavior. This review emphasizes the functions of S1PRs in atherosclerosis and ischemic diseases such as MI and ischemic stroke, enabling mechanistic studies and new S1PRs targeted therapies in atherosclerosis and ischemia in the future.
Collapse
Affiliation(s)
- Shishu Lu
- Hengyang Medical College, University of South China, Hengyang, China
| | - Meihua She
- Hengyang Medical College, University of South China, Hengyang, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China.
| | - Qun Zeng
- Hengyang Medical College, University of South China, Hengyang, China
| | - Guanghui Yi
- Hengyang Medical College, University of South China, Hengyang, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Jiawei Zhang
- Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
39
|
Motyl JA, Strosznajder JB, Wencel A, Strosznajder RP. Recent Insights into the Interplay of Alpha-Synuclein and Sphingolipid Signaling in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22126277. [PMID: 34207975 PMCID: PMC8230587 DOI: 10.3390/ijms22126277] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 01/22/2023] Open
Abstract
Molecular studies have provided increasing evidence that Parkinson’s disease (PD) is a protein conformational disease, where the spread of alpha-synuclein (ASN) pathology along the neuraxis correlates with clinical disease outcome. Pathogenic forms of ASN evoke oxidative stress (OS), neuroinflammation, and protein alterations in neighboring cells, thereby intensifying ASN toxicity, neurodegeneration, and neuronal death. A number of evidence suggest that homeostasis between bioactive sphingolipids with opposing function—e.g., sphingosine-1-phosphate (S1P) and ceramide—is essential in pro-survival signaling and cell defense against OS. In contrast, imbalance of the “sphingolipid biostat” favoring pro-oxidative/pro-apoptotic ceramide-mediated changes have been indicated in PD and other neurodegenerative disorders. Therefore, we focused on the role of sphingolipid alterations in ASN burden, as well as in a vast range of its neurotoxic effects. Sphingolipid homeostasis is principally directed by sphingosine kinases (SphKs), which synthesize S1P—a potent lipid mediator regulating cell fate and inflammatory response—making SphK/S1P signaling an essential pharmacological target. A growing number of studies have shown that S1P receptor modulators, and agonists are promising protectants in several neurological diseases. This review demonstrates the relationship between ASN toxicity and alteration of SphK-dependent S1P signaling in OS, neuroinflammation, and neuronal death. Moreover, we discuss the S1P receptor-mediated pathways as a novel promising therapeutic approach in PD.
Collapse
Affiliation(s)
- Joanna A. Motyl
- Department of Hybrid Microbiosystems Engineering, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland; (J.A.M.); (A.W.)
| | - Joanna B. Strosznajder
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawinskiego St., 02-106 Warsaw, Poland;
| | - Agnieszka Wencel
- Department of Hybrid Microbiosystems Engineering, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland; (J.A.M.); (A.W.)
| | - Robert P. Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawinskiego St., 02-106 Warsaw, Poland
- Correspondence:
| |
Collapse
|
40
|
Kuczynski AM, Oh J. Ozanimod for the treatment of relapsing forms of multiple sclerosis. Neurodegener Dis Manag 2021; 11:207-220. [PMID: 34011158 DOI: 10.2217/nmt-2021-0005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease that causes chronic neurological disability in young adults. Modulation of sphingosine 1-phosphate (S1P) receptors, a group of receptors that, among other things, regulate egression of lymphocytes from lymph nodes, has proven to be effective in treating relapsing MS. Fingolimod, the first oral S1P receptor modulator, has demonstrated potent efficacy and tolerability, but can cause undesirable side effects due to its interaction with a wide range of S1P receptor subtypes. This review will focus on ozanimod, a more selective S1P receptor modulator, which has recently received approval for relapsing MS. We summarize ozanimod's mechanism of action, and efficacy and safety from clinical trials that demonstrate its utility as another treatment option for relapsing MS.
Collapse
Affiliation(s)
- Andrea M Kuczynski
- Department of Medicine, Division of Neurology, St. Michael's Hospital University of Toronto, Toronto, Canada
| | - Jiwon Oh
- Department of Medicine, Division of Neurology, St. Michael's Hospital University of Toronto, Toronto, Canada
| |
Collapse
|
41
|
Fronza M, Lorefice L, Frau J, Cocco E. An Overview of the Efficacy and Safety of Ozanimod for the Treatment of Relapsing Multiple Sclerosis. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:1993-2004. [PMID: 34007159 PMCID: PMC8123972 DOI: 10.2147/dddt.s240861] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/14/2021] [Indexed: 12/20/2022]
Abstract
Multiple sclerosis (MS) is a complex disease of the central nervous system that can cause permanent disability in young adults. A large armamentarium is available for its management and is increasing over time. Ozanimod is an oral drug belonging to the sphingosine-1-phosphate receptor (S1PR) modulator family recently approved in different countries for MS with active disease. It selectively modulates S1PR1 and S1PR5 to prevent autoreactive lymphocytes from entering the central nervous system (CNS), where they can determine inflammation and neurodegeneration. Ozanimod was tested in one Phase II and two Phase III pivotal trials and was shown to be effective and well tolerated. Moreover, further investigations, including comparative trials with other S1P modulators and MS disease-modifying drugs, are needed to better define placement in MS treatment. Furthermore, ozanimod is currently under evaluation for inflammatory bowel diseases, such as ulcerative colitis and Crohn’s disease, in international phase III studies. This article retraces the itinerary leading to the approval of ozanimod for MS treatment and its peculiarities and potentiality inside the S1PR modulator family.
Collapse
Affiliation(s)
- Marzia Fronza
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Lorena Lorefice
- Multiple Sclerosis Center, Binaghi Hospital, ATS Sardegna, ASSL Cagliari, Cagliari, Italy
| | - Jessica Frau
- Multiple Sclerosis Center, Binaghi Hospital, ATS Sardegna, ASSL Cagliari, Cagliari, Italy
| | - Eleonora Cocco
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy.,Multiple Sclerosis Center, Binaghi Hospital, ATS Sardegna, ASSL Cagliari, Cagliari, Italy
| |
Collapse
|
42
|
Ma B, Guckian KM, Liu XG, Yang C, Li B, Scannevin R, Mingueneau M, Drouillard A, Walzer T. Novel Potent Selective Orally Active S1P5 Receptor Antagonists. ACS Med Chem Lett 2021; 12:351-355. [PMID: 33738061 DOI: 10.1021/acsmedchemlett.0c00631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/13/2021] [Indexed: 11/29/2022] Open
Abstract
S1P5 is one of the five sphingosine-1-phosphate (S1P) receptors which play important roles in immune and CNS cell homeostasis, growth, and differentiation. Little is known about the effect of modulation of S1P5 due to the lack of S1P5 specific modulators with suitable druglike properties. Here we describe the discovery and optimization of a novel series of potent selective S1P5 antagonists and the identification of an orally active brain-penetrant tool compound 15.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Annabelle Drouillard
- Université Lyon 1, Lyon 69007, France
- International Center for Infectiology Research, Lyon 69007, France
| | - Thierry Walzer
- Université Lyon 1, Lyon 69007, France
- International Center for Infectiology Research, Lyon 69007, France
| |
Collapse
|
43
|
Chun J, Giovannoni G, Hunter SF. Sphingosine 1-phosphate Receptor Modulator Therapy for Multiple Sclerosis: Differential Downstream Receptor Signalling and Clinical Profile Effects. Drugs 2021; 81:207-231. [PMID: 33289881 PMCID: PMC7932974 DOI: 10.1007/s40265-020-01431-8] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Lysophospholipids are a class of bioactive lipid molecules that produce their effects through various G protein-coupled receptors (GPCRs). Sphingosine 1-phosphate (S1P) is perhaps the most studied lysophospholipid and has a role in a wide range of physiological and pathophysiological events, via signalling through five distinct GPCR subtypes, S1PR1 to S1PR5. Previous and continuing investigation of the S1P pathway has led to the approval of three S1PR modulators, fingolimod, siponimod and ozanimod, as medicines for patients with multiple sclerosis (MS), as well as the identification of new S1PR modulators currently in clinical development, including ponesimod and etrasimod. S1PR modulators have complex effects on S1PRs, in some cases acting both as traditional agonists as well as agonists that produce functional antagonism. S1PR subtype specificity influences their downstream effects, including aspects of their benefit:risk profile. Some S1PR modulators are prodrugs, which require metabolic modification such as phosphorylation via sphingosine kinases, resulting in different pharmacokinetics and bioavailability, contrasting with others that are direct modulators of the receptors. The complex interplay of these characteristics dictates the clinical profile of S1PR modulators. This review focuses on the S1P pathway, the characteristics and S1PR binding profiles of S1PR modulators, the mechanisms of action of S1PR modulators with regard to immune cell trafficking and neuroprotection in MS, together with a summary of the clinical effectiveness of the S1PR modulators that are approved or in late-stage development for patients with MS. Sphingosine 1-phosphate receptor modulator therapy for multiple sclerosis: differential downstream receptor signalling and clinical profile effects (MP4 65540 kb).
Collapse
Affiliation(s)
- Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark St, London, E1 2AT UK
| | - Samuel F. Hunter
- Advanced Neurosciences Institute, 101 Forrest Crossing Blvd STE 103, Franklin, TN 37064 USA
| |
Collapse
|
44
|
Singh S, Singh TG, Rehni AK. An Insight into Molecular Mechanisms and Novel Therapeutic Approaches in Epileptogenesis. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 19:750-779. [PMID: 32914725 DOI: 10.2174/1871527319666200910153827] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 11/22/2022]
Abstract
Epilepsy is the second most common neurological disease with abnormal neural activity involving the activation of various intracellular signalling transduction mechanisms. The molecular and system biology mechanisms responsible for epileptogenesis are not well defined or understood. Neuroinflammation, neurodegeneration and Epigenetic modification elicit epileptogenesis. The excessive neuronal activities in the brain are associated with neurochemical changes underlying the deleterious consequences of excitotoxicity. The prolonged repetitive excessive neuronal activities extended to brain tissue injury by the activation of microglia regulating abnormal neuroglia remodelling and monocyte infiltration in response to brain lesions inducing axonal sprouting contributing to neurodegeneration. The alteration of various downstream transduction pathways resulted in intracellular stress responses associating endoplasmic reticulum, mitochondrial and lysosomal dysfunction, activation of nucleases, proteases mediated neuronal death. The recently novel pharmacological agents modulate various receptors like mTOR, COX-2, TRK, JAK-STAT, epigenetic modulators and neurosteroids are used for attenuation of epileptogenesis. Whereas the various molecular changes like the mutation of the cell surface, nuclear receptor and ion channels focusing on repetitive episodic seizures have been explored by preclinical and clinical studies. Despite effective pharmacotherapy for epilepsy, the inadequate understanding of precise mechanisms, drug resistance and therapeutic failure are the current fundamental problems in epilepsy. Therefore, the novel pharmacological approaches evaluated for efficacy on experimental models of epilepsy need to be identified and validated. In addition, we need to understand the downstream signalling pathways of new targets for the treatment of epilepsy. This review emphasizes on the current state of novel molecular targets as therapeutic approaches and future directions for the management of epileptogenesis. Novel pharmacological approaches and clinical exploration are essential to make new frontiers in curing epilepsy.
Collapse
Affiliation(s)
- Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Ashish Kumar Rehni
- Cerebral Vascular Disease Research Laboratories, Department of Neurology and Neuroscience Program, University of Miami School of Medicine, Miami, Florida 33101, United States
| |
Collapse
|
45
|
Brown D, Moezzi D, Dong Y, Koch M, Yong VW. Combination of Hydroxychloroquine and Indapamide Attenuates Neurodegeneration in Models Relevant to Multiple Sclerosis. Neurotherapeutics 2021; 18:387-400. [PMID: 33410109 PMCID: PMC8116375 DOI: 10.1007/s13311-020-01002-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2020] [Indexed: 01/10/2023] Open
Abstract
As the underlying pathophysiology of progressive forms of multiple sclerosis (MS) remains unclear, current treatment strategies are inadequate. Progressive MS is associated with increased oxidative stress and neuronal damage in lesions along with an extensive representation of activated microglia/macrophages. To target these disease mechanisms, we tested the novel combination of generic medications, hydroxychloroquine (HCQ), and indapamide, in tissue culture and in mice. HCQ is an anti-malarial medication found to inhibit microglial activation and to ameliorate disease activity in experimental autoimmune encephalomyelitis. We are currently completing a phase II trial of HCQ in primary progressive MS ( ClinicalTrials.gov Identifier: NCT02913157). Indapamide is an antihypertensive previously discovered in our laboratory drug screen to be an anti-oxidant. As these medications have a different spectrum of activities on disease mechanisms relevant to progressive MS, their use in combination may be more effective than either alone. We thus sought preclinical data for the effectiveness of this combination. In vitro, indapamide had robust hydroxyl scavenging activity, while HCQ and indapamide alone and in combination protected against iron-induced neuronal killing; TNF-α levels in activated microglia were reduced by either drug alone, without additional combination effects. In mice with a lysolecithin lesion that manifests demyelination and axonal loss in the spinal cord, the combination but not individual treatment of HCQ and indapamide reduced CD68+ microglia/macrophage representation in lesions, attenuated axonal injury, and lowered levels of lipid peroxidation. Our study supports the combination of indapamide and HCQ as a new treatment strategy targeting multiple facets of progressive MS.
Collapse
Affiliation(s)
- Dennis Brown
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - Dorsa Moezzi
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - Yifei Dong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - Marcus Koch
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
46
|
Neuroinflammation in intracerebral haemorrhage: immunotherapies with potential for translation. Lancet Neurol 2020; 19:1023-1032. [DOI: 10.1016/s1474-4422(20)30364-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 09/12/2020] [Accepted: 09/24/2020] [Indexed: 12/22/2022]
|
47
|
Randez-Gil F, Bojunga L, Estruch F, Winderickx J, Del Poeta M, Prieto JA. Sphingolipids and Inositol Phosphates Regulate the Tau Protein Phosphorylation Status in Humanized Yeast. Front Cell Dev Biol 2020; 8:592159. [PMID: 33282871 PMCID: PMC7705114 DOI: 10.3389/fcell.2020.592159] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/21/2020] [Indexed: 01/08/2023] Open
Abstract
Hyperphosphorylation of protein tau is a hallmark of Alzheimer's disease (AD). Changes in energy and lipid metabolism have been correlated with the late onset of this neurological disorder. However, it is uncertain if metabolic dysregulation is a consequence of AD or one of the initiating factors of AD pathophysiology. Also, it is unclear whether variations in lipid metabolism regulate the phosphorylation state of tau. Here, we show that in humanized yeast, tau hyperphosphorylation is stimulated by glucose starvation in coincidence with the downregulation of Pho85, the yeast ortholog of CDK5. Changes in inositol phosphate (IP) signaling, which has a central role in energy metabolism, altered tau phosphorylation. Lack of inositol hexakisphosphate kinases Kcs1 and Vip1 (IP6 and IP7 kinases in mammals) increased tau hyperphosphorylation. Similar effects were found by mutation of IPK2 (inositol polyphosphate multikinase), or PLC1, the yeast phospholipase C gene. These effects may be explained by IP-mediated regulation of Pho85. Indeed, this appeared to be the case for plc1, ipk2, and kcs1. However, the effects of Vip1 on tau phosphorylation were independent of the presence of Pho85, suggesting additional mechanisms. Interestingly, kcs1 and vip1 strains, like pho85, displayed dysregulated sphingolipid (SL) metabolism. Moreover, genetic and pharmacological inhibition of SL biosynthesis stimulated the appearance of hyperphosphorylated forms of tau, while increased flux through the pathway reduced its abundance. Finally, we demonstrated that Sit4, the yeast ortholog of human PP2A protein phosphatase, is a downstream effector of SL signaling in mediating the tau phosphorylation state. Altogether, our results add new knowledge on the molecular effectors involved in tauopathies and identify new targets for pharmacological intervention.
Collapse
Affiliation(s)
- Francisca Randez-Gil
- Department of Biotechnology, Instituto de Agroquímica y Tecnología de los Alimentos, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - Lino Bojunga
- Department of Biotechnology, Instituto de Agroquímica y Tecnología de los Alimentos, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - Francisco Estruch
- Departament of Biochemistry and Molecular Biology, Universitat de València, Valencia, Spain
| | | | - Maurizio Del Poeta
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
- Veterans Administration Medical Center, Northport, NY, United States
| | - Jose A. Prieto
- Department of Biotechnology, Instituto de Agroquímica y Tecnología de los Alimentos, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| |
Collapse
|
48
|
Impact of S1P Mimetics on Mesenteric Ischemia/Reperfusion Injury. Pharmaceuticals (Basel) 2020; 13:ph13100298. [PMID: 33050288 PMCID: PMC7601119 DOI: 10.3390/ph13100298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/24/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenteric ischemia/reperfusion (I/R), following the transient deprivation of blood flow to the gut, triggers an acute flogistic process involving the disruption of endothelial and epithelial barriers integrity, the activation of immune cells, and the abundant release of inflammatory mediators. Among them, the lipid mediator sphingosine-1-phosphate (S1P) is involved in maintaining epithelial and endothelial barrier integrity and in governing the migration of immune cells through the interaction with S1P1–5 receptors. Therefore, the present work aims to investigate the involvement of S1P signaling in intestinal I/R-induced injury by studying the effects of FTY720, the non-selective S1P1,3–5 agonist, and comparing them with the responses to ozanimod, selective S1P1,5 agonist, in a murine model of gut I/R. Intestinal edema, gut and lung neutrophil infiltration, and oxidative stress were evaluated through biochemical and morphological assays. The collected results highlight the protective action of FTY720 against the inflammatory cascade elicited by mesenteric I/R injury, mainly through the control of vascular barrier integrity. While these beneficial effects were mimicked by ozanimod and can be therefore attributed largely to the effects exerted by FTY720 on S1P1, the recruitment of myeloid cells to the injured areas, limited by FTY720 but not by ozanimod, rather suggests the involvement of other receptor subtypes.
Collapse
|
49
|
Esaki K, Balan S, Iwayama Y, Shimamoto-Mitsuyama C, Hirabayashi Y, Dean B, Yoshikawa T. Evidence for Altered Metabolism of Sphingosine-1-Phosphate in the Corpus Callosum of Patients with Schizophrenia. Schizophr Bull 2020; 46:1172-1181. [PMID: 32346731 PMCID: PMC7505171 DOI: 10.1093/schbul/sbaa052] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The disturbed integrity of myelin and white matter, along with dysregulation of the lipid metabolism, may be involved in schizophrenia pathophysiology. Considering the crucial role of sphingolipids in neurodevelopment, particularly in oligodendrocyte differentiation and myelination, we examined the role of sphingolipid dynamics in the pathophysiology of schizophrenia. We performed targeted mass spectrometry-based analysis of sphingolipids from the cortical area and corpus callosum of postmortem brain samples from patients with schizophrenia and controls. We observed lower sphingosine-1-phosphate (S1P) levels, specifically in the corpus callosum of patients with schizophrenia, but not in major depressive disorder or bipolar disorder, when compared with the controls. Patient data and animal studies showed that antipsychotic intake did not contribute to the lowered S1P levels. We also found that lowered S1P levels in the corpus callosum of patients with schizophrenia may stem from the upregulation of genes for S1P-degrading enzymes; higher expression of genes for S1P receptors suggested a potential compensatory mechanism for the lowered S1P levels. A higher ratio of the sum of sphingosine and ceramide to S1P, which can induce apoptosis and cell-cycle arrest, was also observed in the samples of patients with schizophrenia than in controls. These results suggest that an altered S1P metabolism may underlie the deficits in oligodendrocyte differentiation and myelin formation, leading to the structural and molecular abnormalities of white matter reported in schizophrenia. Our findings may pave the way toward a novel therapeutic strategy.
Collapse
Affiliation(s)
- Kayoko Esaki
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Shabeesh Balan
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Yoshimi Iwayama
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
- Support Unit for Bio-Material Analysis, Research Division, RIKEN Center for Brain Science, Saitama, Japan
| | | | - Yoshio Hirabayashi
- Cellular Informatics Laboratory, RIKEN Cluster for Pioneering Research, Saitama, Japan
| | - Brian Dean
- The Florey Institute of Neuroscience and Mental Health, Howard Florey Laboratories, The University of Melbourne, Victoria, Australia
- The Centre for Mental Health, Swinburne University, Victoria, Australia
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
50
|
Fabri JHTM, de Sá NP, Malavazi I, Del Poeta M. The dynamics and role of sphingolipids in eukaryotic organisms upon thermal adaptation. Prog Lipid Res 2020; 80:101063. [PMID: 32888959 DOI: 10.1016/j.plipres.2020.101063] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 01/09/2023]
Abstract
All living beings have an optimal temperature for growth and survival. With the advancement of global warming, the search for understanding adaptive processes to climate changes has gained prominence. In this context, all living beings monitor the external temperature and develop adaptive responses to thermal variations. These responses ultimately change the functioning of the cell and affect the most diverse structures and processes. One of the first structures to detect thermal variations is the plasma membrane, whose constitution allows triggering of intracellular signals that assist in the response to temperature stress. Although studies on this topic have been conducted, the underlying mechanisms of recognizing thermal changes and modifying cellular functioning to adapt to this condition are not fully understood. Recently, many reports have indicated the participation of sphingolipids (SLs), major components of the plasma membrane, in the regulation of the thermal stress response. SLs can structurally reinforce the membrane or/and send signals intracellularly to control numerous cellular processes, such as apoptosis, cytoskeleton polarization, cell cycle arresting and fungal virulence. In this review, we discuss how SLs synthesis changes during both heat and cold stresses, focusing on fungi, plants, animals and human cells. The role of lysophospholipids is also discussed.
Collapse
Affiliation(s)
- João Henrique Tadini Marilhano Fabri
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA; Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Nivea Pereira de Sá
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Iran Malavazi
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA; Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, USA; Veterans Administration Medical Center, Northport, New York, USA.
| |
Collapse
|