1
|
Tork MAB, Fotouhi S, Roozi P, Negah SS. Targeting NLRP3 Inflammasomes: A Trojan Horse Strategy for Intervention in Neurological Disorders. Mol Neurobiol 2024:10.1007/s12035-024-04359-2. [PMID: 39042218 DOI: 10.1007/s12035-024-04359-2] [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: 02/05/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Recently, a growing focus has been on identifying critical mechanisms in neurological diseases that trigger a cascade of events, making it easier to target them effectively. One such mechanism is the inflammasome, an essential component of the immune response system that plays a crucial role in disease progression. The NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3) inflammasome is a subcellular multiprotein complex that is widely expressed in the central nervous system (CNS) and can be activated by a variety of external and internal stimuli. When activated, the NLRP3 inflammasome triggers the production of proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) and facilitates rapid cell death by assembling the inflammasome. These cytokines initiate inflammatory responses through various downstream signaling pathways, leading to damage to neurons. Therefore, the NLRP3 inflammasome is considered a significant contributor to the development of neuroinflammation. To counter the damage caused by NLRP3 inflammasome activation, researchers have investigated various interventions such as small molecules, antibodies, and cellular and gene therapy to regulate inflammasome activity. For instance, recent studies indicate that substances like micro-RNAs (e.g., miR-29c and mR-190) and drugs such as melatonin can reduce neuronal damage and suppress neuroinflammation through NLRP3. Furthermore, the transplantation of bone marrow mesenchymal stem cells resulted in a significant reduction in the levels of pyroptosis-related proteins NLRP3, caspase-1, IL-1β, and IL-18. However, it would benefit future research to have an in-depth review of the pharmacological and biological interventions targeting inflammasome activity. Therefore, our review of current evidence demonstrates that targeting NLRP3 inflammasomes could be a pivotal approach for intervention in neurological disorders.
Collapse
Affiliation(s)
- Mohammad Amin Bayat Tork
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soroush Fotouhi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Roozi
- Department of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran.
| |
Collapse
|
2
|
Mokhtarzadeh Khanghahi A, Rayatpour A, Baharvand H, Javan M. Neuroglial components of brain lesions may provide new therapeutic strategies for multiple sclerosis. Neurol Sci 2023; 44:3795-3807. [PMID: 37410268 DOI: 10.1007/s10072-023-06915-5] [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: 12/30/2022] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune and demyelinating disease of the central nervous system (CNS) which leads to focal demyelinated lesions in the brain and spinal cord. Failure of remyelination contributes to chronic disability in young adults. Characterization of events occurring during the demyelination and remyelination processes and those of which subsequently limit remyelination or contribute to demyelination can provide the possibility of new therapies development for MS. Most of the currently available therapies and investigations modulate immune responses and mediators. Since most therapeutic strategies have unsatisfied outcomes, developing new therapies that enhance brain lesion repair is a priority. A close look at cellular and chemical components of MS lesions will pave the way to a better understanding of lesions pathology and will provide possible opportunities for repair strategies and targeted pharmacotherapy. This review summarizes the lesion components and features, particularly the detrimental elements, and discusses the possibility of suggesting new potential targets as therapies for demyelinating diseases like MS.
Collapse
Affiliation(s)
- Akram Mokhtarzadeh Khanghahi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Atefeh Rayatpour
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Hossein Baharvand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Javan
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran.
- International Collaboration on Repair Discoveries (ICORD), the University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
3
|
Liu W, Yu Z, Wang Z, Waubant EL, Zhai S, Benet LZ. Using an animal model to predict the effective human dose for oral multiple sclerosis drugs. Clin Transl Sci 2023; 16:467-477. [PMID: 36419359 PMCID: PMC10014696 DOI: 10.1111/cts.13458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
The objective of this study was to determine the potential usefulness of an animal model to predict the appropriate dose of newly developed drugs for treating relapsing remitting multiple sclerosis (RRMS). Conversion of the lowest effective dose (LEffD) for mice and rats in the experimental autoimmune encephalomyelitis (EAE) model was used to predict the human effective dose utilizing the body surface area correction factor found in the 2005 US Food and Drug Administration (FDA) Guidance for Industry in selecting safe starting doses for clinical trials. Predictions were also tested by comparison with doses estimated by scaling up the LEffD in the model by the human to animal clearance ratio. Although initial proof-of-concept studies of oral fingolimod tested the efficacy and safety of 1.25 and 5 mg in treating RRMS, the EAE animal model predicted the approved dose of this drug, 0.5 mg daily. This approach would have also provided useful predictions of the approved human oral doses for cladribine, dimethyl fumarate, ozanimod, ponesimod, siponimod, and teriflunomide, drugs developed with more than one supposed mechanism of action. The procedure was not useful for i.v. dosed drugs, including monoclonal antibodies. We maintain that drug development scientists should always examine a simple allometric method to predict the therapeutic effective dose in humans. Then, following clinical studies, we believe that the animal model might be expected to yield useful predictions of other drugs developed to treat the same condition. The methodology may not always be predictive, but the approach is so simple it should be investigated.
Collapse
Affiliation(s)
- Wei Liu
- Department of PharmacyPeking University Third HospitalBeijingChina
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Zhiheng Yu
- Department of PharmacyPeking University Third HospitalBeijingChina
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Ziyu Wang
- Department of PharmacyPeking University Third HospitalBeijingChina
- School of Basic Medical Sciences and Clinical PharmacyChina Pharmaceutical UniversityNanjingChina
| | - Emmanuelle L. Waubant
- Weill Institute for Neurosciences and San Francisco Multiple Sclerosis CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Suodi Zhai
- Department of PharmacyPeking University Third HospitalBeijingChina
| | - Leslie Z. Benet
- Department of PharmacyPeking University Third HospitalBeijingChina
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
4
|
Schroeter CB, Rolfes L, Gothan KSS, Gruchot J, Herrmann AM, Bock S, Fazio L, Henes A, Narayanan V, Pfeuffer S, Nelke C, Räuber S, Huntemann N, Duarte-Silva E, Dobelmann V, Hundehege P, Wiendl H, Raba K, Küry P, Kremer D, Ruck T, Müntefering T, Budde T, Cerina M, Meuth SG. Cladribine treatment improves cortical network functionality in a mouse model of autoimmune encephalomyelitis. J Neuroinflammation 2022; 19:270. [DOI: 10.1186/s12974-022-02588-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/07/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cladribine is a synthetic purine analogue that interferes with DNA synthesis and repair next to disrupting cellular proliferation in actively dividing lymphocytes. The compound is approved for the treatment of multiple sclerosis (MS). Cladribine can cross the blood–brain barrier, suggesting a potential effect on central nervous system (CNS) resident cells. Here, we explored compartment-specific immunosuppressive as well as potential direct neuroprotective effects of oral cladribine treatment in experimental autoimmune encephalomyelitis (EAE) mice.
Methods
In the current study, we compare immune cell frequencies and phenotypes in the periphery and CNS of EAE mice with distinct grey and white matter lesions (combined active and focal EAE) either orally treated with cladribine or vehicle, using flow cytometry. To evaluate potential direct neuroprotective effects, we assessed the integrity of the primary auditory cortex neuronal network by studying neuronal activity and spontaneous synaptic activity with electrophysiological techniques ex vivo.
Results
Oral cladribine treatment significantly attenuated clinical deficits in EAE mice. Ex vivo flow cytometry showed that cladribine administration led to peripheral immune cell depletion in a compartment-specific manner and reduced immune cell infiltration into the CNS. Histological evaluations revealed no significant differences for inflammatory lesion load following cladribine treatment compared to vehicle control. Single cell electrophysiology in acute brain slices was performed and showed an impact of cladribine treatment on intrinsic cellular firing patterns and spontaneous synaptic transmission in neurons of the primary auditory cortex. Here, cladribine administration in vivo partially restored cortical neuronal network function, reducing action potential firing. Both, the effect on immune cells and neuronal activity were transient.
Conclusions
Our results indicate that cladribine exerts a neuroprotective effect after crossing the blood–brain barrier independently of its peripheral immunosuppressant action.
Collapse
|
5
|
Medeiros-Furquim T, Ayoub S, Johnson LJ, Aprico A, Nwoke E, Binder MD, Kilpatrick TJ. Cladribine Treatment for MS Preserves the Differentiative Capacity of Subsequently Generated Monocytes, Whereas Its Administration In Vitro Acutely Influences Monocyte Differentiation but Not Microglial Activation. Front Immunol 2022; 13:678817. [PMID: 35734180 PMCID: PMC9207174 DOI: 10.3389/fimmu.2022.678817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Cladribine (2-chlorodeoxyadenosine, 2CdA) is one of the most effective disease-modifying drugs for multiple sclerosis (MS). Cladribine is a synthetic purine nucleoside analog that induces cell death of lymphocytes and oral cladribine treatment leads to a long-lasting disease stabilization, potentially attributable to immune reconstitution. In addition to its effects on lymphocytes, cladribine has been shown to have immunomodulatory effects on innate immune cells, including dendritic cells and monocytes, which could also contribute to its therapeutic efficacy. However, whether cladribine can modulate human macrophage/microglial activation or monocyte differentiation is currently unknown. The aim of this study was to determine the immunomodulatory effects of cladribine upon monocytes, monocyte-derived macrophages (MDMs) and microglia. We analyzed the phenotype and differentiation of monocytes from MS patients receiving their first course of oral cladribine both before and three weeks after the start of treatment. Flow cytometric analysis of monocytes from MS patients undergoing cladribine treatment revealed that the number and composition of CD14/CD16 monocyte subsets remained unchanged after treatment. Furthermore, after differentiation with M-CSF, such MDMs from treated MS patients showed no difference in gene expression of the inflammatory markers compared to baseline. We further investigated the direct effects of cladribine in vitro using human adult primary MDMs and microglia. GM-CSF-derived MDMs were more sensitive to cell death than M-CSF-derived MDMs. In addition, MDMs treated with cladribine showed increased expression of costimulatory molecules CD80 and CD40, as well as expression of anti-inflammatory, pro-trophic genes IL10 and MERTK, depending on the differentiation condition. Cladribine treatment in vitro did not modulate the expression of activation markers in human microglia. Our study shows that cladribine treatment in vitro affects the differentiation of monocytes into macrophages by modulating the expression of activation markers, which might occur similarly in tissue after their infiltration in the CNS during MS.
Collapse
Affiliation(s)
- Tiago Medeiros-Furquim
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Sinan Ayoub
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Laura J. Johnson
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Andrea Aprico
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Eze Nwoke
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Michele D. Binder
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Department of Neuroscience and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Trevor J. Kilpatrick
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- *Correspondence: Trevor J. Kilpatrick,
| |
Collapse
|
6
|
Cui Y, Yu H, Bu Z, Wen L, Yan L, Feng J. Focus on the Role of the NLRP3 Inflammasome in Multiple Sclerosis: Pathogenesis, Diagnosis, and Therapeutics. Front Mol Neurosci 2022; 15:894298. [PMID: 35694441 PMCID: PMC9175009 DOI: 10.3389/fnmol.2022.894298] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is initiated with an aberrant innate immune response in the central nervous system (CNS) and is involved in many neurological diseases. Inflammasomes are intracellular multiprotein complexes that can be used as platforms to induce the maturation and secretion of proinflammatory cytokines and pyroptosis, thus playing a pivotal role in neuroinflammation. Among the inflammasomes, the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3) inflammasome is well-characterized and contributes to many neurological diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), and ischemic stroke. MS is a chronic autoimmune disease of the CNS, and its hallmarks include chronic inflammation, demyelination, and neurodegeneration. Studies have demonstrated a relationship between MS and the NLRP3 inflammasome. To date, the pathogenesis of MS is not fully understood, and clinical studies on novel therapies are still underway. Here, we review the activation mechanism of the NLRP3 inflammasome, its role in MS, and therapies targeting related molecules, which may be beneficial in MS.
Collapse
|
7
|
2-Chlorodeoxyadenosine (Cladribine) preferentially inhibits the biological activity of microglial cells. Int Immunopharmacol 2022; 105:108571. [DOI: 10.1016/j.intimp.2022.108571] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 02/05/2023]
|
8
|
Correale J, Halfon MJ, Jack D, Rubstein A, Villa A. Acting centrally or peripherally: A renewed interest in the central nervous system penetration of disease-modifying drugs in multiple sclerosis. Mult Scler Relat Disord 2021; 56:103264. [PMID: 34547609 DOI: 10.1016/j.msard.2021.103264] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 09/03/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
With the recent approval of cladribine tablets, siponimod and ozanimod, there has been a renewed interest into the extent to which these current generation disease-modifying therapies (DMTs) are able to cross into the central nervous system (CNS), and how this penetration of the blood-brain barrier (BBB) may influence their ability to treat multiple sclerosis (MS). The integrity of the CNS is maintained by the BBB, blood-cerebrospinal fluid barrier, and the arachnoid barrier, which all play an important role in preserving the immunological environment and homeostasis within the CNS. The integrity of the BBB decreases during the course of MS, with a putative temporal relationship to disease worsening. Furthermore, it is currently considered that progression of the disease is mediated mainly by resident cells of the CNS. The existing literature provides evidence to show that some of the current generation DMTs for MS are able to penetrate the CNS and potentially exert direct effects on CNS-resident cells, in particular the CNS-penetrating prodrugs cladribine and fingolimod, and other sphingosine-1 phosphate receptor modulators; siponimod and ozanimod. Other current generation DMTs appear to be restricted to the periphery due to their high molecular weight or physicochemical properties. As more effective brain penetrant therapies are developed for the treatment of MS, there is a need to understand whether the potential for direct effects within the CNS are of significance, and whether this brings additional benefits over and above treatment effects mediated in the periphery. In turn, this will require an improved understanding of the structure and function of the BBB, the role it plays in MS and subsequent treatments. This narrative review summarizes the data supporting the biological plausibility of a potential benefit from therapeutic molecules entering the CNS, and discusses the potential significance in the current and future treatment of MS.
Collapse
Affiliation(s)
- Jorge Correale
- Department of Neurology, Fleni, Buenos Aires, Argentina.
| | | | - Dominic Jack
- Merck Serono Ltd, Feltham, United Kingdom (an affiliate of Merck KGaA)
| | - Adrián Rubstein
- Merck S.A., Buenos Aires, Argentina (an affiliate of Merck KGaA)
| | - Andrés Villa
- Hospital Ramos Mejía, Universidad de Buenos Aires, Argentina
| |
Collapse
|
9
|
Mathiesen CBK, Rudjord-Levann AM, Gad M, Larsen J, Sellebjerg F, Pedersen AE. Cladribine inhibits secretion of pro-inflammatory cytokines and phagocytosis in human monocyte-derived M1 macrophages in-vitro. Int Immunopharmacol 2020; 91:107270. [PMID: 33360830 DOI: 10.1016/j.intimp.2020.107270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 12/31/2022]
Abstract
Cladribine (Cd) is a purine nucleoside analogue which in an oral formulation is approved for treatment of patients with multiple sclerosis (MS). It is known to mediate the effect through a short-term selective reduction of lymphocytes with minimal effect on the innate immune system. However, a few studies have emerged, that also demonstrate a beneficial immunomodulatory effect of cladribine on monocyte-derived cells. As cladribine crosses the blood-brain barrier this effect could have clinical meaningful impact in the treatment of MS, where recruitment of innate cells such as M1 macrophages play a role in plaque development. Here, we investigated the in-vitro effect on monocyte differentiation into M1 and M2 macrophages and dendritic cells as well as the effect on activation of M1 macrophages. In our experiments, cladribine in therapeutic relevant in-vitro concentrations, did not lead to apoptosis in differentiated M1, M2 macrophages or DCs and did not interfere with the phenotype of these differentiated cells. In M1 macrophages, cladribine reduced the secretion of IL-6 and TNF-α observed after activation with LPS. Similar, cladribine reduced the phagocytic capacity of LPS activated M1 macrophages but did not affect unactivated cells. We conclude, that such reduction of inflammatory potential as well as reduced M1 phagocytic activity, e.g. within an MS plaque, could be an additional clinical meaningful effect of cladribine in the treatment of MS while at the same time it would leave M1 macrophages intact for the protection against infections.
Collapse
Affiliation(s)
| | | | | | | | - Finn Sellebjerg
- The Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Anders Elm Pedersen
- Merck (Denmark), Denmark, an affiliate of Merck KGaA Darmstadt, Germany; Department of Odontology, University of Copenhagen, Denmark.
| |
Collapse
|
10
|
Effect of Cladribine on Neuronal Apoptosis: New Insight of In Vitro Study in Multiple Sclerosis Therapy. Brain Sci 2020; 10:brainsci10080548. [PMID: 32823496 PMCID: PMC7464206 DOI: 10.3390/brainsci10080548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/01/2022] Open
Abstract
Background: Cladribine (2-CdA) can cross the blood–brain barrier, resulting in inhibition of DNA synthesis and repair and disruption of cellular proliferation in actively dividing lymphocytes. No data on effect on neurons are available. Aim: To study “in vitro” 2-CdA apoptotic effects on neurons in healthy donor and multiple sclerosis patient lymphocytes. Methods: Neuroblastoma cells were co-cultured with lymphocytes, with and without 2-CdA. Results: Apoptosis increased in lymphocytes with 2-CdA; increase was also observed when lymphocytes were cultured with neuronal cells. However, neurons were not affected by 2-CdA for apoptosis. Conclusions: 2-CdA causes peripheral and central lymphocyte death preserving neurons, with a reasonable impact on inflammation and neuroprotection.
Collapse
|
11
|
Cladribine Treatment Improved Homocysteine Metabolism and Increased Total Serum Antioxidant Activity in Secondary Progressive Multiple Sclerosis Patients. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1654754. [PMID: 32256946 PMCID: PMC7103043 DOI: 10.1155/2020/1654754] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/02/2020] [Accepted: 02/18/2020] [Indexed: 11/17/2022]
Abstract
Hyperhomocysteinemia plays a crucial role in the pathogenesis of many diseases of the central nervous system (CNS). The nervous system is particularly sensitive to high homocysteine (Hcy) level mainly due to its prooxidative and cytotoxic effects. Cladribine, a drug recently registered for the treatment of multiple sclerosis (MS), possesses additionally neuroprotective effects which are independent of its peripheral immunosuppressant action. Accumulating evidence suggests that oxidative stress and homocysteine thiolactone-mediated protein homocysteinylation play a causal role in MS. Both of these processes may be attenuated by paraoxonase 1 (PON1). Therefore, in the present study, we aimed to examine whether the beneficial effects of the drug in MS patients with a secondary progressive (SP) clinical course, treated with cladribine subcutaneously (s.c.), may be related to its ability to modify serum PON1 activity, Hcy concentration, and protein homocysteinylation, as well as to correct total antioxidant status. A total of 118 subjects were enrolled into the study: (1) patients with a SP type of MS, SP-MS (n = 40); (2) patients with a relapsing-remitting (RR) type of MS, RR-MS (n = 30); and (3) healthy people (n = 48). Patients with SP-MS were treated with cladribine. The drug was given in SP-SM patients s.c. six times every 6 weeks up to a total mean cumulative dose of 1.8 mg/kg. PON1 activity was assessed spectrophotometrically. The level of Hcy, homocysteine thiolactone (HTL) attached to plasma proteins (N-Hcy-protein), and antibodies against homocysteinylated proteins was assessed with an enzyme immunoassay. The total antioxidant activity of the serum was assessed with the ferric-reducing activity of plasma (FRAP) method. Basically, there was no difference in PON1 activity between untreated SP-MS, RR-MS, and control subjects. Serum Hcy was significantly higher in RR-MS patients (p < 0.001) and in SP-MS patients (p < 0.01) compared to the control group. The N-Hcy protein level was higher in RR-MS patients (p < 0.05) in comparison to the control group. Moreover, the elevated level of antibodies against homocysteinylated proteins was observed in the serum of patients with SP-MS. The total antioxidant capacity of serum was lower in MS patients vs. the control group (p < 0.001). After cladribine treatment, the activity of PON1 did not change in SP-MS patients, whereas cladribine treatment decreased the level of total Hcy (p < 0.05). Treatment with cladribine increased the total serum antioxidant activity in SP-MS patients (p < 0.01). The Expanded Disability Status Scale (EDSS) score did not change in SP-MS patients. Cladribine treatment in the SP-MS group attenuates hyperhomocysteinemia-induced protein homocysteinylation (n.s.). It also stabilises the neurological condition of SP-MS patients. The stabilisation of a neurological condition observed in SP-MS patients after cladribine treatment may be partially related to its ability to reduce elevated Hcy level and to improve serum antioxidant potential.
Collapse
|
12
|
Glial Factors Regulating White Matter Development and Pathologies of the Cerebellum. Neurochem Res 2020; 45:643-655. [PMID: 31974933 PMCID: PMC7058568 DOI: 10.1007/s11064-020-02961-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/31/2022]
Abstract
The cerebellum is a brain region that undergoes extremely dynamic growth during perinatal and postnatal development which is regulated by the proper interaction between glial cells and neurons with a complex concert of growth factors, chemokines, cytokines, neurotransmitters and transcriptions factors. The relevance of cerebellar functions for not only motor performance but also for cognition, emotion, memory and attention is increasingly being recognized and acknowledged. Since perturbed circuitry of cerebro-cerebellar trajectories can play a role in many central nervous system pathologies and thereby contribute to neurological symptoms in distinct neurodevelopmental and neurodegenerative diseases, is it the aim with this mini-review to highlight the pathways of glia–glia interplay being involved. The designs of future treatment strategies may hence be targeted to molecular pathways also playing a role in development and disease of the cerebellum.
Collapse
|
13
|
Immune reconstitution therapies: concepts for durable remission in multiple sclerosis. Nat Rev Neurol 2019; 16:56-62. [PMID: 31649335 DOI: 10.1038/s41582-019-0268-z] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2019] [Indexed: 12/29/2022]
Abstract
New so-called immune reconstitution therapies (IRTs) have the potential to induce long-term or even permanent drug-free remission in people with multiple sclerosis (MS). These therapies deplete components of the immune system with the aim of allowing the immune system to renew itself. Haematopoietic stem cell transplantation, the oral formulation cladribine and the monoclonal antibodies alemtuzumab, rituximab and ocrelizumab are frequently categorized as IRTs. However, the evidence that IRTs indeed renew adaptive immune cell repertoires and rebuild a healthy immune system in people with MS is variable. Instead, IRTs might foster the expansion of those cells that survive immunosuppression, and this expansion could be associated with acquisition of new functional phenotypes. Understanding immunological changes induced by IRTs and how they correlate with clinical outcomes will be instrumental in guiding the optimal use of immune reconstitution as a durable therapeutic strategy. This Perspectives article critically discusses the efficacy and potential mechanisms of IRTs in the context of immune system renewal and durable disease remission in MS.
Collapse
|
14
|
Abstract
Multiple sclerosis treatment faces tremendous changes owing to the approval of new medications, some of which are available as oral formulations. Until now, the four orally available medications, fingolimod, dimethylfumarate (BG-12), teriflunomide, and cladribine have received market authorization, whereas laquinimod is still under development. Fingolimod is a sphingosine-1-phosphate inhibitor, which is typically used as escalation therapy and leads to up to 60% reduction of the annualized relapse rate, but might also have neuroprotective properties. In addition, there are three more specific S1P agonists in late stages of development: siponimod, ponesimod, and ozanimod. Dimethylfumarate has immunomodulatory and cytoprotective functions and is used as baseline therapy. Teriflunomide, the active metabolite of the rheumatoid arthritis medication leflunomide, targets the dihydroorotate dehydrogenase, thus inhibiting the proliferation of lymphocytes by depletion of pyrimidines. Here we will review the mechanisms of action, clinical trial data, as well as data about safety and tolerability of the compounds.
Collapse
Affiliation(s)
- Simon Faissner
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| |
Collapse
|
15
|
Madhu K, T P, S M. Bacoside-A inhibits inflammatory cytokines and chemokine in experimental autoimmune encephalomyelitis. Biomed Pharmacother 2018; 109:1339-1345. [PMID: 30551384 DOI: 10.1016/j.biopha.2018.10.188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/27/2018] [Accepted: 10/31/2018] [Indexed: 11/29/2022] Open
Abstract
Chronic inflammation of the myelin sheath is the crucial event behind the progression of multiple sclerosis (MS). Bacoside-A is one of the major constituents obtained from Bacopa monerii (L.) Wettst., and possess neuroprotective as well as anti-inflammatory actions. The current study explores the effect of Bacoside-A in acute and chronic models of Experimental Autoimmune Encephalomyelitis (EAE). The results indicate that the Bacoside-A treated mice produced a significant reduction in disease score compared to disease control in both models. The treatment with Bacoside-A downregulated the inflammatory cytokines (IL-6, IL-17a, and TNFα) and inflammatory chemokine CCL-5 in EAE mice. On the other hand, Bacoside-A treated mice showed a nonsignificant effect on promoting the expressions of NCAM, BDNF1, and FOXP3 in acute and chronic models of EAE. Histopathological analysis revealed that the Bacoside-A treated mice at a dose of 10 mg/kg exhibited a significant reduction in cellular infiltrations, cellular changes, and demyelination in cerebral tissues, but unable to protect at a higher dose in both models. In conclusion, Bacoside-A can able to inhibit the progression of EAE may be by the inhibition of inflammatory cytokines and chemokine evolved during active EAE.
Collapse
Affiliation(s)
- Krishnadas Madhu
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Bengaluru, India.
| | - Prakash T
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Bengaluru, India.
| | - Maya S
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Bengaluru, India
| |
Collapse
|
16
|
Burrows DJ, McGown A, Jain SA, De Felice M, Ramesh TM, Sharrack B, Majid A. Animal models of multiple sclerosis: From rodents to zebrafish. Mult Scler 2018; 25:306-324. [PMID: 30319015 DOI: 10.1177/1352458518805246] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated demyelinating disease of the central nervous system. Animal models of MS have been critical for elucidating MS pathological mechanisms and how they may be targeted for therapeutic intervention. Here we review the most commonly used animal models of MS. Although these animal models cannot fully replicate the MS disease course, a number of models have been developed to recapitulate certain stages. Experimental autoimmune encephalomyelitis (EAE) has been used to explore neuroinflammatory mechanisms and toxin-induced demyelinating models to further our understanding of oligodendrocyte biology, demyelination and remyelination. Zebrafish models of MS are emerging as a useful research tool to validate potential therapeutic candidates due to their rapid development and amenability to genetic manipulation.
Collapse
Affiliation(s)
- David John Burrows
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Alexander McGown
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Saurabh A Jain
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Milena De Felice
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Tennore M Ramesh
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Basil Sharrack
- Academic Department of Neuroscience, The Sheffield NIHR Translational Neuroscience Biomedical Research Centre, University of Sheffield, Sheffield, UK
| | - Arshad Majid
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK/Academic Department of Neuroscience, The Sheffield NIHR Translational Neuroscience Biomedical Research Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
17
|
Role of Inflammasomes in Neuroimmune and Neurodegenerative Diseases: A Systematic Review. Mediators Inflamm 2018; 2018:1549549. [PMID: 29849483 PMCID: PMC5932495 DOI: 10.1155/2018/1549549] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/26/2017] [Accepted: 01/01/2018] [Indexed: 12/31/2022] Open
Abstract
Inflammasomes are multiprotein complexes that can sense pathogen-associated molecular patterns and damage-associated molecular signals. They are involved in the initiation and development of inflammation via activation of IL-1β and IL-18. Many recent studies suggest a strong correlation between inflammasomes and neurological diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), and Parkinson's disease (PD). Several components of inflammasomes, such as nucleotide-binding oligomerization domain- (NOD-) like receptor, absent in melanoma 2- (AIM2-) like receptors (ALRs), apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and caspase-1, as well as the upstream factors and downstream effectors, are associated with the initiation and development of MS and its animal model, experimental autoimmune encephalomyelitis. Additionally, inflammasomes affect the efficacy of interferon-β therapy in patients with MS. Finally, the strong association of inflammasomes with AD and PD needs to be further studied. In this review of latest literatures, we comprehensively tease out diverse roles of different kinds of inflammasomes in neuroimmune and neurodegenerative diseases, especially in the perspective of double roles involved in pathogenesis, and identify future research priorities.
Collapse
|
18
|
Barclay W, Shinohara ML. Inflammasome activation in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Brain Pathol 2018; 27:213-219. [PMID: 27997058 DOI: 10.1111/bpa.12477] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/14/2016] [Indexed: 12/19/2022] Open
Abstract
The aptly named inflammasomes are powerful signaling complexes that sense inflammatory signals under a myriad of conditions, including those from infections and endogenous sources. The inflammasomes promote inflammation by maturation and release of the pro-inflammatory cytokines, IL-1β and IL-18. Several inflammasomes have been identified so far, but this review focuses mainly on the NLRP3 inflammasome. By still ill-defined activation mechanisms, a sensor molecule, NLRP3 (NACHT, LRR and PYD domains-containing protein 3), responds to danger signals and rapidly recruits ASC (apoptosis-associated speck-like protein containing a CARD) and pro-caspase-1 to form a large oligomeric signaling platform-the inflammasome. Involvement of the NLRP3 inflammasome in infections, metabolic disorders, autoinflammation, and autoimmunity, underscores its position as a central player in sensing microbial and damage signals and coordinating pro-inflammatory immune responses. Indeed, evidence in patients with multiple sclerosis (MS) suggests inflammasome activation occurs during disease. Experiments with the mouse model of MS, experimental autoimmune encephalomyelitis (EAE), specifically describe the NLRP3 inflammasome as critical and necessary to disease development. This review discusses recent studies in EAE and MS which describe associations of inflammasome activation with promotion of T cell pathogenicity, infiltration of cells into the central nervous system (CNS) and direct neurodegeneration during EAE and MS.
Collapse
Affiliation(s)
- William Barclay
- Department of Immunology, Duke University Medical School, Durham, NC
| | - Mari L Shinohara
- Department of Immunology, Duke University Medical School, Durham, NC.,Department of Molecular Genetics and Microbiology, Duke University Medical School, Durham, NC
| |
Collapse
|
19
|
Abstract
Cladribine is a deoxyadenosine analogue prodrug that preferentially depletes lymphocytes, key cells underlying multiple sclerosis (MS) pathogenesis. Cladribine tablets (Mavenclad®) represent the first short-course oral disease-modifying drug (DMD) for use in MS. The tablets, administered in two short courses 1 year apart, are indicated for the treatment of adults with highly active relapsing MS on the basis of data from pivotal clinical trials, including the phase 3 study CLARITY and its extension. A cumulative cladribine tablets dose of 3.5 mg/kg administered in this fashion in CLARITY reduced clinical relapse, disability progression and MRI-assessed disease activity and also improved some aspects of health-related quality of life (HR-QOL) versus placebo over 96 weeks in adults with relapsing-remitting MS (RRMS). Moreover, in the 96-week extension (plus 24 weeks' supplemental follow-up), no additional clinical benefit was gained from continuing versus discontinuing cladribine tablets after the first two annual courses of therapy, although MRI activity was more notable in a subset of cladribine tablet recipients who discontinued the drug. In post hoc analyses of CLARITY and/or a phase 2b trial, benefits of cladribine tablets were seen in patients with high disease activity (HDA) relapsing MS that were sometimes greater than in patients without HDA. Cladribine tablets have an acceptable tolerability profile and do not appear to be associated with an increased risk of overall infection or with an increased risk of malignancy (vs. matched reference populations). Active comparisons and longer-term follow-up would be beneficial, although current data indicate that for adults with highly active relapsing MS, cladribine tablets are an effective treatment option with the convenience of low-burden, short-course, oral administration.
Collapse
Affiliation(s)
- Emma D. Deeks
- 0000 0004 0372 1209grid.420067.7Springer, Private Bag 65901, Mairangi Bay, Auckland, 0754 New Zealand
| |
Collapse
|
20
|
Grand'Maison F, Yeung M, Morrow SA, Lee L, Emond F, Ward BJ, Laneuville P, Schecter R. Sequencing of high-efficacy disease-modifying therapies in multiple sclerosis: perspectives and approaches. Neural Regen Res 2018; 13:1871-1874. [PMID: 30233054 PMCID: PMC6183038 DOI: 10.4103/1673-5374.239432] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Multiple sclerosis (MS) is characterized by chronic inflammation in conjunction with neurodegeneration within the central nervous system. Most individuals with MS begin with a relapsing remitting course that later transitions to secondary progressive MS. Currently available disease-modifying therapies (DMTs) for relapsing MS have been demonstrated to reduce disease activity, however most patients require a change in therapy over the course of their disease. Treatment goals include the prevention of relapses and disability accumulation and to achieve this objective requires careful planning. Sequencing of DMTs for individual patients should be designed in such a way to maximize disease control and minimize risk based on the mechanism of action, pharmacokinetic and pharmacodynamic properties of each therapy. This includes the DMT patients are being switched from to those they are being switched to. The reversibility of immune system effects should be a key consideration for DMT sequence selection. This feature varies across DMTs and should factor more prominently in decision making as newer treatments become available for the prevention of disability accumulation in patients with progressive MS. In this short review, we discuss the landscape of existing therapies with an eye to the future when planning for optimal DMT sequencing. While no cure exists for MS, efforts are being directed toward research in neuroregeneration with the hope for positive outcomes.
Collapse
Affiliation(s)
| | - Michael Yeung
- Clinical Neurosciences, Foothills Medical Centre, Calgary, Alberta, Canada
| | - Sarah A Morrow
- London Health Sciences Center (LHSC), Western University, London, Ontario, Canada
| | - Liesly Lee
- Department of Neurology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Francois Emond
- Centre Hospitalier Universitaire de Québec - hôpital de l'Enfant-Jésus, Quebec City, Quebec, Canada
| | - Brian J Ward
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Robyn Schecter
- Novartis Pharmaceuticals Canada Inc., Dorval, Quebec, Canada
| |
Collapse
|
21
|
Holmøy T, Torkildsen Ø, Myhr KM. An update on cladribine for relapsing-remitting multiple sclerosis. Expert Opin Pharmacother 2017; 18:1627-1635. [DOI: 10.1080/14656566.2017.1372747] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Trygve Holmøy
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Univeristy of Oslo, Oslo, Norway
| | - Øivind Torkildsen
- Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Kjell-Morten Myhr
- Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
22
|
Gentile A, Musella A, Bullitta S, Fresegna D, De Vito F, Fantozzi R, Piras E, Gargano F, Borsellino G, Battistini L, Schubart A, Mandolesi G, Centonze D. Siponimod (BAF312) prevents synaptic neurodegeneration in experimental multiple sclerosis. J Neuroinflammation 2016; 13:207. [PMID: 27566665 PMCID: PMC5002118 DOI: 10.1186/s12974-016-0686-4] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/18/2016] [Indexed: 12/31/2022] Open
Abstract
Background Data from multiple sclerosis (MS) and the MS rodent model, experimental autoimmune encephalomyelitis (EAE), highlighted an inflammation-dependent synaptopathy at the basis of the neurodegenerative damage causing irreversible disability in these disorders. This synaptopathy is characterized by an imbalance between glutamatergic and GABAergic transmission and has been proposed to be a potential therapeutic target. Siponimod (BAF312), a selective sphingosine 1-phosphate1,5 receptor modulator, is currently under investigation in a clinical trial in secondary progressive MS patients. We investigated whether siponimod, in addition to its peripheral immune modulation, may exert direct neuroprotective effects in the central nervous system (CNS) of mice with chronic progressive EAE. Methods Minipumps allowing continuous intracerebroventricular (icv) infusion of siponimod for 4 weeks were implanted into C57BL/6 mice subjected to MOG35-55-induced EAE. Electrophysiology, immunohistochemistry, western blot, qPCR experiments, and peripheral lymphocyte counts were performed. In addition, the effect of siponimod on activated microglia was assessed in vitro to confirm the direct effect of the drug on CNS-resident immune cells. Results Siponimod administration (0.45 μg/day) induced a significant beneficial effect on EAE clinical scores with minimal effect on peripheral lymphocyte counts. Siponimod rescued defective GABAergic transmission in the striatum of EAE, without correcting the EAE-induced alterations of glutamatergic transmission. We observed a significant attenuation of astrogliosis and microgliosis together with reduced lymphocyte infiltration in the striatum of EAE mice treated with siponimod. Interestingly, siponimod reduced the release of IL-6 and RANTES from activated microglial cells in vitro, which might explain the reduced lymphocyte infiltration. Furthermore, the loss of parvalbumin-positive (PV+) GABAergic interneurons typical of EAE brains was rescued by siponimod treatment, providing a plausible explanation of the selective effects of this drug on inhibitory synaptic transmission. Conclusions Altogether, our results show that siponimod has neuroprotective effects in the CNS of EAE mice, which are likely independent of its peripheral immune effect, suggesting that this drug could be effective in limiting neurodegenerative pathological processes in MS.
Collapse
Affiliation(s)
- Antonietta Gentile
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Alessandra Musella
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy
| | - Silvia Bullitta
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy
| | - Diego Fresegna
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Francesca De Vito
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Roberta Fantozzi
- Unit of Neurology and Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli, IS, Italy
| | - Eleonora Piras
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia-CERC, 00143, Rome, Italy
| | - Francesca Gargano
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia-CERC, 00143, Rome, Italy
| | | | - Luca Battistini
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia-CERC, 00143, Rome, Italy
| | - Anna Schubart
- Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Georgia Mandolesi
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy.
| | - Diego Centonze
- Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy.,Unit of Neurology and Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli, IS, Italy
| |
Collapse
|