Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
Glia ; 72(9): 1555-1571, 2024 09.
Article in English | MEDLINE | ID: mdl-38829008

ABSTRACT

As one of the top causes of blindness worldwide, glaucoma leads to diverse optic neuropathies such as degeneration of retinal ganglion cells (RGCs). It is widely accepted that the level of intraocular pressure (IOP) is a major risk factor in human glaucoma, and reduction of IOP level is the principally most well-known method to prevent cell death of RGCs. However, clinical studies show that lowering IOP fails to prevent RGC degeneration in the progression of glaucoma. Thus, a comprehensive understanding of glaucoma pathological process is required for developing new therapeutic strategies. In this study, we provide functional and histological evidence showing that optic nerve defects occurred before retina damage in an ocular hypertension glaucoma mouse model, in which oligodendroglial lineage cells were responsible for the subsequent neuropathology. By treatment with clemastine, an Food and Drug Administration (FDA)-approved first-generation antihistamine medicine, we demonstrate that the optic nerve and retina damages were attenuated via promoting oligodendrocyte precursor cell (OPC) differentiation and enhancing remyelination. Taken together, our results reveal the timeline of the optic neuropathies in glaucoma and highlight the potential role of oligodendroglial lineage cells playing in its treatment. Clemastine may be used in future clinical applications for demyelination-associated glaucoma.


Subject(s)
Clemastine , Glaucoma , Mice, Inbred C57BL , Remyelination , Retina , Animals , Clemastine/pharmacology , Clemastine/therapeutic use , Glaucoma/pathology , Glaucoma/drug therapy , Retina/pathology , Retina/drug effects , Remyelination/drug effects , Remyelination/physiology , Mice , Optic Nerve/drug effects , Optic Nerve/pathology , Disease Models, Animal , Optic Nerve Diseases/drug therapy , Optic Nerve Diseases/pathology , Oligodendroglia/drug effects , Oligodendroglia/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
2.
Org Biomol Chem ; 22(9): 1812-1820, 2024 02 28.
Article in English | MEDLINE | ID: mdl-38328995

ABSTRACT

A library of hybrid molecules is developed based on the common chemical features shared by clemastine and tamoxifen both of which are well known for their antileishmanial activities. In the initial screening against Leishmania major and L. amazonensis promastigotes, as well as cytotoxicity assays using HepG2 cells, several hybrids showed submicromolar activity against the parasite and no toxicity against human cells. The compounds with an EC50 < 2 µM against promastigotes of both species and a selectivity index >10 were further characterized against intracellular amastigotes as well as promastigotes of species that cause both visceral and cutaneous leishmaniasis, such as L. infantum and L. braziliensis, respectively. These sequential screenings revealed the high pan-activity of this class of molecules against these species, with several compounds displaying an EC50 ≤ 2 µM against both promastigotes and intracellular amastigotes. Two of them were identified as the potential templates for lead optimization of this series having shown the highest activities against all species in both stages of parasite. The present findings can serve as a good starting point in the search for novel antileishmanial compounds that are easy to access and highly active.


Subject(s)
Antiprotozoal Agents , Leishmaniasis, Cutaneous , Humans , Animals , Mice , Clemastine/therapeutic use , Macrophages , Leishmaniasis, Cutaneous/drug therapy , Leishmaniasis, Cutaneous/parasitology , Antiprotozoal Agents/pharmacology , Hep G2 Cells , Mice, Inbred BALB C
3.
BMJ Open ; 14(1): e076651, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38296293

ABSTRACT

INTRODUCTION: Remyelination failure hampers symptomatic recovery in multiple sclerosis (MS), underlining the importance of developing remyelinating therapies. Optic neuritis is currently the most established method of measuring remyelination in MS trials. Complementary more generalisable methods of measuring remyelination are required to confirm treatment efficacy. Measuring internuclear ophthalmoplegia (INO) with infrared oculography provides such a method. Moreover, this method can be expanded with a test for selecting likely treatment responders by using fampridine. The aim of this trial is to investigate the (long-term) remyelinating effects of clemastine fumarate in patients with MS and INO and to evaluate if treatment response can be predicted using fampridine. METHODS AND ANALYSIS: RESTORE is a single-centre double-blind randomised placebo-controlled trial of clemastine fumarate versus placebo. Prior to clemastine treatment improvement in oculographic features of INO after a single 10 mg dose of fampridine is measured in all participants and used to predict the treatment response to clemastine. Eighty individuals with MS and INO will be 1:1 randomised to 4 mg of clemastine fumarate two times a day for 6 months or equivalent placebo. Our primary outcome is improvement in the Versional Dysconjugacy Index-area under the curve, measured by infrared oculography after 6 months of treatment. Participants are assessed for persistent treatment effects 6, 18 and 30 months after end of treatment. Secondary outcome measures include other oculography parameters including double-step saccades, retinal imaging, visual acuities, physical disability, cognition and patient-reported outcomes. ETHICS AND DISSEMINATION: Clemastine is a registered and very well-established drug with well-known safety and side effects. The protocol was approved by the medical ethical committee of the Amsterdam UMC, location VUMC and the Dutch Central Committee on Research Involving Human Subject. Written informed consent is obtained from all participants. The results will be published in peer-reviewed medical scientific journals. TRIAL REGISTRATION NUMBER: EudraCT: 2021-003677-66, ClinicalTrials.gov: NCT05338450.


Subject(s)
Multiple Sclerosis , Ocular Motility Disorders , Humans , Multiple Sclerosis/complications , Multiple Sclerosis/drug therapy , Clemastine/therapeutic use , 4-Aminopyridine/therapeutic use , Treatment Outcome , Double-Blind Method , Randomized Controlled Trials as Topic
4.
Transl Res ; 268: 40-50, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38246342

ABSTRACT

Traumatic brain injury (TBI) has a significant impact on cognitive function, affecting millions of people worldwide. Myelin loss is a prominent pathological feature of TBI, while well-functioning myelin is crucial for memory and cognition. Utilizing drug repurposing to identify effective drug candidates for TBI treatment has gained attention. Notably, recent research has highlighted the potential of clemastine, an FDA-approved allergy medication, as a promising pro-myelinating drug. Therefore, in this study, we aim to investigate whether clemastine can enhance myelination and alleviate cognitive impairment following mild TBI using a clinically relevant rat model of TBI. Mild diffuse TBI was induced using the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA). Animals were treated with either clemastine or an equivalent volume of the vehicle from day 1 to day 14 post-injury. Following treatment, memory-related behavioral tests were conducted, and myelin pathology in the cortex and hippocampus was assessed through immunofluorescence staining and ProteinSimple® capillary-based immunoassay. Our results showed that TBI leads to significant myelin loss, axonal damage, glial activation, and a decrease in mature oligodendrocytes in both the cortex and hippocampus. The TBI animals also exhibited notable deficits in memory-related tests. In contrast, animals treated with clemastine showed an increase in mature oligodendrocytes, enhanced myelination, and improved performance in the behavioral tests. These preliminary findings support the therapeutic value of clemastine in alleviating TBI-induced cognitive impairment, with substantial clinical translational potential. Our findings also underscore the potential of remyelinating therapies for TBI.


Subject(s)
Axons , Clemastine , Cognitive Dysfunction , Disease Models, Animal , Myelin Sheath , Rats, Sprague-Dawley , Animals , Clemastine/pharmacology , Clemastine/therapeutic use , Myelin Sheath/drug effects , Myelin Sheath/pathology , Myelin Sheath/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/pathology , Axons/drug effects , Axons/pathology , Male , Rats , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/complications , Brain Injuries, Diffuse/drug effects , Brain Injuries, Diffuse/pathology , Hippocampus/drug effects , Hippocampus/pathology
5.
Biomed Pharmacother ; 157: 113904, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36370521

ABSTRACT

Neurodegenerative diseases have been a weighty problem in elder people who might be stricken with motor or/and cognition defects with lower life quality urging for effective treatment. Drugs are costly from development to market, so that drug repurposing, exploration of existing drugs for novel therapeutic purposes, becomes a wise and popular strategy to raise new treatment options. Clemastine fumarate, different from anti-allergic effect as H1 histamine antagonist, was screened and identified as promising drug for remyelination and autophagy enhancement. Surprisingly, fumarate salt also has similar effect. Hence, whether clemastine fumarate would make a protective impact on neurodegenerative diseases and what contribution fumarate probably makes are intriguing to us. In this review, we summarize the potential mechanism surrounding clemastine fumarate in current literature, and try to distinguish independent or synergistic effect between clemastine and fumarate, aiming to find worthwhile research direction for neurodegeneration diseases.


Subject(s)
Anti-Allergic Agents , Remyelination , Humans , Aged , Clemastine/therapeutic use , Drug Repositioning , Fumarates
6.
Brain ; 145(11): 3943-3952, 2022 11 21.
Article in English | MEDLINE | ID: mdl-35678509

ABSTRACT

Many biomarkers in clinical neuroscience lack pathological certification. This issue is potentially a significant contributor to the limited success of neuroprotective and neurorestorative therapies for human neurological disease-and is evident even in areas with therapeutic promise such as myelin repair. Despite the identification of promising remyelinating candidates, biologically validated methods to demonstrate therapeutic efficacy or provide robust preclinical evidence of remyelination in the CNS are lacking. Therapies with potential to remyelinate the CNS constitute one of the most promising and highly anticipated therapeutic developments in the pipeline to treat multiple sclerosis and other demyelinating diseases. The optic nerve has been proposed as an informative pathway to monitor remyelination in animals and human subjects. Recent clinical trials using visual evoked potential have had promising results, but without unequivocal evidence about the cellular and molecular basis for signal changes on visual evoked potential, the interpretation of these trials is constrained. The visual evoked potential was originally developed and used in the clinic as a diagnostic tool but its use as a quantitative method for assessing therapeutic response requires certification of its biological specificity. Here, using the tools of experimental pathology we demonstrate that quantitative measurements of myelination using both histopathological measures of nodal structure and ultrastructural assessments correspond to visual evoked potential latency in both inflammatory and chemical models of demyelination. Visual evoked potential latency improves after treatment with a tool remyelinating compound (clemastine), mirroring both quantitative and qualitative myelin assessment. Furthermore, clemastine does not improve visual evoked potential latency following demyelinating injury when administered to a transgenic animal incapable of forming new myelin. Therefore, using the capacity for therapeutic enhancement and biological loss of function we demonstrate conclusively that visual evoked potential measures myelin status and is thereby a validated tool for preclinical verification of remyelination.


Subject(s)
Multiple Sclerosis , Remyelination , Humans , Animals , Evoked Potentials, Visual , Clemastine/therapeutic use , Myelin Sheath/metabolism , Multiple Sclerosis/pathology , Biomarkers/metabolism
7.
Microbiol Spectr ; 10(2): e0054121, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35234502

ABSTRACT

Staphylococcus aureus poses a significant threat to human health due to its virulence and multidrug resistance. In addition, recalcitrant biofilm formation of S. aureus often results in chronic infection and the treatment tolerance toward the traditional antibiotics. Thus, the development of novel antimicrobial agents capable to inhibit or eradicate S. aureus biofilm formation does matter. Here, we demonstrated that clemastine showed slight bacteriostatic activity and enhanced the antibacterial activity of oxacillin against S. aureus. Moreover, the dramatic inhibition of biofilm formation was found in clinical S. aureus strains by clemastine. Clemastine inhibited the release of eDNA during the biofilm formation and decreased the S. aureus hemolytic activity. Moreover, the S. aureus SA113 treated with clemastine displayed the decreased transcriptional level of the biofilm formation relevant genes (fnbB, icaA, and icaB), virulence genes (hlg, hld, lukde, lukpvl, beta-PSM, delta-PSM, and cap5A), and the regulatory genes agrA. The proteomics analysis of SA113 treated with clemastine demonstrated the significant changes in levels of biofilm-related proteins (stress response regulators ClpB and GroS, ATP-binding proteins, and urease metabolism), virulence-related proteins (SspA, superantigen, and VWbp), and methicillin resistance-related proteins (glutamine metabolism). The genetic mutations on gdpP (cyclic di-AMP phosphodiesterase) were found in the clemastine-induced tolerant derivative isolate by whole-genome sequencing. Furthermore, the interaction between clemastine and GdpP protein was demonstrated by the molecular docking, gdpP overexpression experiment, and thermal stability assay. Conclusively, clemastine might exert its inhibitory effects against the biofilm formation and hemolysis in S. aureus through targeting GdpP protein. IMPORTANCE The biofilm formation, which protects bacteria from stresses, including antibiotics and host immune responses, can be commonly found in clinical S. aureus isolates worldwide. Treatment failure of traditional antibiotics in biofilm-associated S. aureus infections remains a serious challenge. The novel anti-biofilm drug is urgently needed to address the looming crisis. In this study, clemastine, which is a histamine receptor H1 (HRH1) antagonist, was found to have a novel role of the significant inhibition against the biofilm formation and hemolytic activity of S. aureus and enhanced antibacterial activity against S. aureus when used in combination with oxacillin by targeting the GdpP protein. The discovery of this study identified novel use and mechanism of action of clemastine as a potential anti-biofilm drug for clinical application for S. aureus infectious.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms , Clemastine/pharmacology , Clemastine/therapeutic use , Hemolysis , Humans , Methicillin-Resistant Staphylococcus aureus/genetics , Microbial Sensitivity Tests , Molecular Docking Simulation , Oxacillin/pharmacology , Oxacillin/therapeutic use , Staphylococcal Infections/microbiology , Staphylococcus aureus
8.
Neuroscience ; 484: 66-79, 2022 02 21.
Article in English | MEDLINE | ID: mdl-35007691

ABSTRACT

With the improvement of cancer treatment techniques, increasing attention has been given to chemotherapy-induced cognitive impairment through white matter injury. Clemastine fumarate has been shown to enhance white matter integrity in cuprizone- or hypoxia-induced demyelination mouse models. However, whether clemastine can be beneficial for reversing chemotherapy-induced cognitive impairment remains unexplored. In this study, the mice received oral administration of clemastine after chemotherapy. The open-field test and Morris water maze test were used to evaluate their anxiety, locomotor activity and cognitive function. Luxol Fast Blue staining and transmission electron microscopy were used to detect the morphological damage to the myelin. Demyelination and damage to the mature oligodendrocytes and axons were observed by immunofluorescence and western blotting. Clemastine significantly improved their cognitive function and ameliorated white matter injury in the chemotherapy-treated mice. Clemastine enhanced myelination, promoted oligodendrocyte precursor cell differentiation and increased the neurofilament 200 protein levels in the corpus callosum and hippocampus. We concluded that clemastine rescues cognitive function damage caused by chemotherapy through improving white matter integrity. Remyelination, oligodendrocyte differentiation and the increase of neurofilament protein promoted by clemastine are potential strategies for reversing the cognitive dysfunction caused by chemotherapy.


Subject(s)
Chemotherapy-Related Cognitive Impairment , Demyelinating Diseases , White Matter , Animals , Clemastine/pharmacology , Clemastine/therapeutic use , Corpus Callosum , Cuprizone , Demyelinating Diseases/chemically induced , Mice , Mice, Inbred C57BL , Myelin Sheath/metabolism , Oligodendroglia/metabolism
9.
Neurochem Res ; 47(2): 503-515, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34661796

ABSTRACT

Recent evidence has shown that demyelination occurs along with axonal degeneration in spinal cord injury (SCI) during the secondary injury phase. Oligodendrocyte precursor cells (OPC) are present in the lesions but fail to differentiate into mature oligodendrocytes and form new myelin. Given the limited recovery of neuronal functions after SCI in adults without effective treatment available so far, it remains unknown whether enhancing OPC differentiation and myelination could benefit the recovery of SCI. To show the significance of myelin regeneration after SCI, the injury was treated with clemastine in the rat model. Clemastine is an FDA-approved drug that is potent in promoting oligodendrocyte differentiation and myelination in vivo, for four weeks following SCI. Motor function was assessed using sloping boards and grid walking tests and scored according to the Basso, Beattie, and Bresnahan protocol. The myelin integrity and protein expression were evaluated using transmission electron microscopy and immunofluorescence, respectively. The results indicated that clemastine treatment preserves myelin integrity, decreases loss of axons and improves functional recovery in the rat SCI model. The presented data suggest that myelination-enhancing strategies may serve as a potential therapeutic approach for the functional recovery in SCI.


Subject(s)
Clemastine , Spinal Cord Injuries , Animals , Clemastine/metabolism , Clemastine/pharmacology , Clemastine/therapeutic use , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Rats , Recovery of Function , Spinal Cord/metabolism , Spinal Cord Injuries/pathology
10.
Exp Neurol ; 342: 113742, 2021 08.
Article in English | MEDLINE | ID: mdl-33965410

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder with limited available drugs for treatment. Enhancing autophagy attenuates AD pathology in various AD model mice. Thus, development of potential drugs which enhance autophagy may bring beneficial effects in AD therapy. In the present study, we show clemastine, a first-generation histamine H1R antagonist and being originally marketed for the treatment of allergic rhinitis, ameliorates AD pathogenesis in APP/PS1 transgenic mice. Chronic treatment with clemastine orally reduced amyloid-ß (Aß) load, neuroinflammation and cognitive deficits of APP/PS1 transgenic mice. Clemastine decreases Aß generation via reducing the levels of BACE1, CTFs of APP. Mechanistically, clemastine enhances autophagy concomitant with a suppression of mTOR signaling. Therefore, we propose that clemastine attenuates AD pathology via enhancing mTOR-mediated autophagy.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Autophagy/drug effects , Clemastine/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Autophagy/physiology , Clemastine/pharmacology , Dose-Response Relationship, Drug , HeLa Cells , Histamine H1 Antagonists/pharmacology , Histamine H1 Antagonists/therapeutic use , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1 , TOR Serine-Threonine Kinases/metabolism
11.
Int J Med Sci ; 18(3): 639-645, 2021.
Article in English | MEDLINE | ID: mdl-33437198

ABSTRACT

Intracerebral hemorrhage (ICH) represents a common acute cerebrovascular event that imparts high rates of disability. The microglia-mediated inflammatory response is a critical factor in determining cerebral damage post-ICH. Clemastine (CLM) is a histamine receptor H1 (HRH1) antagonist that has been shown to modulate the inflammatory response. However, the effects of CLM on ICH and the underlying mechanism remain to be determined. This investigation reveals that CLM resulted in reduction of cerebral hematoma volume, decreased cerebral edema and lower rates of neuronal apoptosis as well as improved behavioral scores in an acute ICH murine model. CLM treatment was noted to decrease pro-inflammatory effectors and increased anti-inflammatory effectors post-ICH. In addition, CLM reduced the deleterious effects of activated microglia on neurons in a transwell co-culture system. Our findings show that CLM likely mediates its therapeutic effect through inhibition of microglia-induced inflammatory response and apoptosis, thereby enhancing restoration of neuronal function.


Subject(s)
Brain Edema/drug therapy , Cerebral Hemorrhage/drug therapy , Clemastine/pharmacology , Inflammation Mediators/metabolism , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Brain Edema/immunology , Brain Edema/pathology , Cells, Cultured , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Clemastine/therapeutic use , Coculture Techniques , Disease Models, Animal , Male , Mice , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Neuroprotective Agents/therapeutic use , Primary Cell Culture , Stereotaxic Techniques
12.
ACS Infect Dis ; 7(1): 47-63, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33291887

ABSTRACT

Current chemotherapeutics for leishmaniasis have multiple deficiencies, and there is a need for new safe, efficacious, and affordable medicines. This study describes a successful drug repurposing approach that identifies the over-the-counter antihistamine, clemastine fumarate, as a potential antileishmanial drug candidate. The screening for inhibitors of the sphingolipid synthase (inositol phosphorylceramide synthase, IPCS) afforded, following secondary screening against Leishmania major (Lmj) promastigotes, 16 active compounds. Further refinement through the dose response against LmjIPCS and intramacrophage L. major amastigotes identified clemastine fumarate with good activity and selectivity with respect to the host macrophage. On target engagement was supported by diminished sensitivity in a sphingolipid-deficient L. major mutant (ΔLmjLCB2) and altered phospholipid and sphingolipid profiles upon treatment with clemastine fumarate. The drug also induced an enhanced host cell response to infection indicative of polypharmacology. The activity was sustained across a panel of Old and New World Leishmania species, displaying an in vivo activity equivalent to the currently used drug, glucantime, in a mouse model of L. amazonensis infection. Overall, these data validate IPCS as an antileishmanial drug target and indicate that clemastine fumarate is a candidate for repurposing for the treatment of leishmaniasis.


Subject(s)
Antiprotozoal Agents , Leishmaniasis , Pharmaceutical Preparations , Animals , Antiprotozoal Agents/pharmacology , Clemastine/therapeutic use , Inositol , Leishmaniasis/drug therapy , Mice
13.
Clin Neurol Neurosurg ; 193: 105741, 2020 06.
Article in English | MEDLINE | ID: mdl-32145678

ABSTRACT

OBJECTIVES: Optic neuritis (ON) is the most common cause of optic neuropathy; typically presenting with a unilateral visual loss in young adults, with incidence of 1-5 in 100,000 per year. We evaluated the effect of Clemastine, a first-generation and CNS (central nervous system)-penetrant H1 receptor antagonist on visual evoked potential (VEP), retinal nerve fibre layer (RNFL) and ganglion cell layer (GCL) complex in patients with optic neuritis. PATIENTS AND METHODS: This is a prospective comparative interventional case series in 25 patients with acute optic neuritis. Patients were randomly assigned to group 1 (treated with Clemastine 1 mg orally twice a day for 90 days; 16 patients) or group 2 (received placebo for 90 days; 9 patients) and both groups received standard treatment of optic neuritis. We recorded VEP and peripapillary OCT (optical coherence tomography) of patients before and after three months of treatment. RESULTS: In contrast to patients treated with Clemastine, RNFL thickness loss between base line phase and after three months follow up in control group were statistically significant in temporal, supra temporal, Infrotemporal and almost global sections of RNFL map. The reduction in GCL thickness between base line phase and after three months follow up in control group were significant, while it did not reach significance in treatment group except in inferior region. CONCLUSION: In contrast to treatment group, RNFL and GCL thickness of most quadrants are decreased significantly after three months in patients with ON in control group. In contrast to control group, p100 wave's amplitude recovered in a significant manner in treatment group.


Subject(s)
Clemastine/therapeutic use , Evoked Potentials, Visual/drug effects , Nerve Fibers/pathology , Optic Neuritis/drug therapy , Optic Neuritis/pathology , Retinal Ganglion Cells/pathology , Adult , Clemastine/adverse effects , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Middle Aged , Multiple Sclerosis/complications , Prospective Studies , Tomography, Optical Coherence , Treatment Outcome , Vision Disorders/drug therapy , Young Adult
14.
J Neuroinflammation ; 17(1): 57, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-32061255

ABSTRACT

BACKGROUND: Microglia activation is associated with the development of hypoxic-ischemic brain injury (HIBI). Neuroinflammation suppression might be a suitable therapeutic target in hypoxic oligodendrocyte injury. This study aims to determine whether clemastine can improve hypomyelination by suppressing the activated microglia and promoting the maturation of oligodendrocyte progenitor cells (OPCs) in HIBI. METHODS: A bilateral common carotid artery occlusion (BCCAO) rat model that received continuous intraperitoneal injection (1 mg/kg) for 14 days was employed to elaborate the neuroprotection effects of clemastine. Interleukin-1ß (IL-1ß), nod-like receptor protein 3 (NLRP3), histamine H1 receptor, and OPC differentiation levels in the corpus callosum were measured. Primary cultured OPCs and co-culture of microglia and OPCs were used to explore the link between microglia activation and hypomyelination. Data were evaluated by one-way ANOVA with Fisher's protected least significant difference test. RESULTS: Clemastine treatment could reverse hypomyelination and restrain the upregulation of IL-1ß and NLRP3 in the corpus callosum of BCCAO rats. Primary cultured OPCs treated with IL-1ß showed failed maturation. However, clemastine could also reverse the OPC maturation arrest by activating the extracellular signal-regulated kinase (ERK) signaling pathway. Co-culture of microglia and OPCs with oxygen glucose deprivation treatment exhibited IL-1ß and NLRP3 upregulation. Clemastine could downregulate NLRP3 and IL-1ß and reverse hypomyelination by inhibiting the p38 signaling pathway. CONCLUSIONS: Clemastine could restrain microglia activation, improve axonal hypomyelination in BCCAO rats, and thus might be a viable strategy to inhibit hypomyelination in the corpus callosum of patients with HIBI.


Subject(s)
Clemastine/pharmacology , Demyelinating Diseases/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , Interleukin-1beta/metabolism , Microglia/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Clemastine/therapeutic use , Histamine H1 Antagonists/pharmacology , Histamine H1 Antagonists/therapeutic use , Male , Microglia/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
15.
Cephalalgia ; 39(5): 597-607, 2019 04.
Article in English | MEDLINE | ID: mdl-30165750

ABSTRACT

OBJECTIVE: To investigate the effect of the H1-antihistamine clemastine on the migraine-inducing abilities of pituitary adenylate cyclase activating peptide-38. METHODS: We conducted a double-blind, randomized, placebo controlled two-way cross-over study. Twenty migraine without aura patients were randomly allocated to receive bolus clemastine 2 mg (1 mg/ml) or bolus saline 2 ml intravenously over 2 min on two study days. Following each bolus injection, 10 pmol/kg/min of pituitary adenylate cyclase activating peptide-38 was administered intravenously over 20 min. We recorded migraine/headache characteristics every 10 min until 90 min after the start of infusion, and collected blood to investigate mast cell degranulation and the inflammation markers tryptase and tumor necrosis factor-alpha before and after infusion of pituitary adenylate cyclase activating peptide-38. RESULTS: After clemastine pretreatment, five out of 20 participants developed a migraine-like attack in response to a pituitary adenylate cyclase activating peptide-38 infusion compared to nine out of 20 after placebo pretreatment ( p = 0.288). Following clemastine pretreatment, 15 out of 20 participants reported headache in response to a pituitary adenylate cyclase activating peptide-38 infusion, whereas 19 out of 20 participants did so following placebo pretreatment ( p = 0.221). We found no difference in area under the curve 12 h for headache intensity between the two experimental days ( p = 0.481). We found no difference in area under the curve 180 min for tryptase ( p = 0.525) or tumor necrosis factor-alpha ( p = 0.487) between clemastine and placebo pretreatment days. CONCLUSION: H1-antihistamine, clemastine, failed to prevent migraine or headache after pituitary adenylate cyclase activating peptide-38 infusion, thus making a role for histamine release or mast cell degranulation in pituitary adenylate cyclase activating peptide-38-induced migraine less likely.


Subject(s)
Clemastine/therapeutic use , Histamine H1 Antagonists/therapeutic use , Migraine Disorders/chemically induced , Migraine Disorders/prevention & control , Pituitary Adenylate Cyclase-Activating Polypeptide/adverse effects , Adult , Cross-Over Studies , Double-Blind Method , Female , Headache/chemically induced , Headache/prevention & control , Humans , Male , Middle Aged , Young Adult
16.
Br J Clin Pharmacol ; 84(5): 1020-1028, 2018 05.
Article in English | MEDLINE | ID: mdl-29385232

ABSTRACT

AIMS: Intravenous high-dose free methylprednisolone (MP) hemisuccinate is the primary treatment for an acute relapse in relapsing-remitting multiple sclerosis. However, it is inconvenient and its side effects are undesirable. Both dose and dosing frequency can be reduced by incorporating free MP in glutathione-PEGylated liposomes, creating a slow-release formulation with reduced toxicity and prolonged peripheral efficacy. This first-in-human study was designed to assess the safety, pharmacokinetics and pharmacodynamics of glutathione-PEGylated liposomes containing MP (2B3-201). METHODS: The first part was a double-blind, three-way cross over study in 18 healthy male subjects, receiving ascending doses of 2B3-201, active comparator (free MP) or placebo. Part 2 of the study was an open-label infusion of 2B3-201 (different doses), exploring pretreatment with antihistamines and different infusion schedules in another 18 healthy male subjects, and a cross-over study in six healthy female subjects. MP plasma concentrations, lymphocyte counts, adrenocorticotropic hormone, osteocalcin and fasting glucose were determined. Safety and tolerability profiles were assessed based on adverse events, safety measurements and central nervous system tests. RESULTS: The most frequent recorded AE related to 2B3-201 was an infusion related reaction (89%). 2B3-201 was shown to have a plasma half-life between 24 and 37 h and caused a prolonged decrease in the lymphocyte count, adrenocorticotropic hormone and osteocalcin, and a rise in fasting glucose. CONCLUSION: 2B3-201 is considered safe, with no clinically relevant changes in central nervous system safety parameters and no serious adverse events. In addition, 2B3-201 shows a long plasma half-life and prolonged immunosuppressive effects.


Subject(s)
Delayed-Action Preparations/pharmacology , Glutathione/chemistry , Liposomes/chemistry , Methylprednisolone/pharmacology , Methylprednisolone/pharmacokinetics , Adrenocorticotropic Hormone/blood , Adult , Anti-Allergic Agents/therapeutic use , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Blood Glucose , Clemastine/therapeutic use , Cross-Over Studies , Delayed-Action Preparations/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Drug Compounding/methods , Drug Therapy, Combination/adverse effects , Female , Healthy Volunteers , Humans , Liposomes/adverse effects , Liposomes/pharmacokinetics , Liposomes/pharmacology , Lymphocyte Count , Male , Methylprednisolone/adverse effects , Methylprednisolone/chemistry , Osteocalcin/blood , Polyethylene Glycols/chemistry
17.
Brain ; 141(1): 85-98, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29244098

ABSTRACT

Hypoxia can injure brain white matter tracts, comprised of axons and myelinating oligodendrocytes, leading to cerebral palsy in neonates and delayed post-hypoxic leukoencephalopathy (DPHL) in adults. In these conditions, white matter injury can be followed by myelin regeneration, but myelination often fails and is a significant contributor to fixed demyelinated lesions, with ensuing permanent neurological injury. Non-myelinating oligodendrocyte precursor cells are often found in lesions in plentiful numbers, but fail to mature, suggesting oligodendrocyte precursor cell differentiation arrest as a critical contributor to failed myelination in hypoxia. We report a case of an adult patient who developed the rare condition DPHL and made a nearly complete recovery in the setting of treatment with clemastine, a widely available antihistamine that in preclinical models promotes oligodendrocyte precursor cell differentiation. This suggested possible therapeutic benefit in the more clinically prevalent hypoxic injury of newborns, and we demonstrate in murine neonatal hypoxic injury that clemastine dramatically promotes oligodendrocyte precursor cell differentiation, myelination, and improves functional recovery. We show that its effect in hypoxia is oligodendroglial specific via an effect on the M1 muscarinic receptor on oligodendrocyte precursor cells. We propose clemastine as a potential therapy for hypoxic brain injuries associated with white matter injury and oligodendrocyte precursor cell maturation arrest.


Subject(s)
Clemastine/therapeutic use , Demyelinating Diseases/drug therapy , Demyelinating Diseases/etiology , Histamine H1 Antagonists/therapeutic use , Hypoxia, Brain/complications , Recovery of Function/drug effects , Action Potentials/drug effects , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/ultrastructure , Demyelinating Diseases/diagnostic imaging , Demyelinating Diseases/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental/drug effects , Humans , Hypoxia, Brain/diagnostic imaging , Male , Mice , Mice, Knockout , Middle Aged , Myelin Sheath/drug effects , Myelin Sheath/ultrastructure , Oligodendrocyte Precursor Cells/drug effects , Optic Nerve/physiopathology , Oxygen/pharmacology , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M1/metabolism
18.
Lancet ; 390(10111): 2481-2489, 2017 Dec 02.
Article in English | MEDLINE | ID: mdl-29029896

ABSTRACT

BACKGROUND: Multiple sclerosis is a degenerative inflammatory disease of the CNS characterised by immune-mediated destruction of myelin and progressive neuroaxonal loss. Myelin in the CNS is a specialised extension of the oligodendrocyte plasma membrane and clemastine fumarate can stimulate differentiation of oligodendrocyte precursor cells in vitro, in animal models, and in human cells. We aimed to analyse the efficacy and safety of clemastine fumarate as a treatment for patients with multiple sclerosis. METHODS: We did this single-centre, 150-day, double-blind, randomised, placebo-controlled, crossover trial (ReBUILD) in patients with relapsing multiple sclerosis with chronic demyelinating optic neuropathy on stable immunomodulatory therapy. Patients who fulfilled international panel criteria for diagnosis with disease duration of less than 15 years were eligible. Patients were randomly assigned (1:1) via block randomisation using a random number generator to receive either clemastine fumarate (5·36 mg orally twice daily) for 90 days followed by placebo for 60 days (group 1), or placebo for 90 days followed by clemastine fumarate (5·36 mg orally twice daily) for 60 days (group 2). The primary outcome was shortening of P100 latency delay on full-field, pattern-reversal, visual-evoked potentials. We analysed by intention to treat. The trial is registered with ClinicalTrials.gov, number NCT02040298. FINDINGS: Between Jan 1, 2014, and April 11, 2015, we randomly assigned 50 patients to group 1 (n=25) or group 2 (n=25). All patients completed the study. The primary efficacy endpoint was met with clemastine fumarate treatment, which reduced the latency delay by 1·7 ms/eye (95% CI 0·5-2·9; p=0·0048) when analysing the trial as a crossover. Clemastine fumarate treatment was associated with fatigue, but no serious adverse events were reported. INTERPRETATION: To our knowledge, this is the first randomised controlled trial to document efficacy of a remyelinating drug for the treatment of chronic demyelinating injury in multiple sclerosis. Our findings suggest that myelin repair can be achieved even following prolonged damage. FUNDING: University of California, San Francisco and the Rachleff Family.


Subject(s)
Clemastine/therapeutic use , Multiple Sclerosis/drug therapy , Adult , Cross-Over Studies , Double-Blind Method , Evoked Potentials, Visual/drug effects , Female , Humans , Male , Remyelination/drug effects , Tomography, Optical Coherence , Treatment Outcome
19.
Swiss Dent J ; 127(4): 324-325, 2017.
Article in French | MEDLINE | ID: mdl-28480948

ABSTRACT

Anaphylaxis is defined as a serious allergic or hypersensitivity reaction in the most cases as a result of an IgE-mediated allergic reaction that is rapid in onset and may cause death. Common triggers are foods, insect stings, and medications. The medical treatment includes, epinephrine, glucocorticoids, antihistamines and inhaled bronchodilators, with the aim to prevent progression to life-threatening respiratory and/or cardiovascular symptoms.


Subject(s)
Anaphylaxis/chemically induced , Anaphylaxis/drug therapy , Clemastine/therapeutic use , Epinephrine/therapeutic use , Histamine Antagonists/therapeutic use , Methylprednisolone Hemisuccinate/therapeutic use , Resuscitation , Combined Modality Therapy , Humans
20.
Exp Neurol ; 288: 114-121, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27865736

ABSTRACT

Primary progressive multiple sclerosis (PPMS) is a chronic demyelinating disease of the central nervous system (CNS) currently lacking any effective treatment. Promoting endogenous brain repair offers a potential strategy to halt and possibly restore neurologic function in PPMS. To understand how the microenvironment within white matter lesions plays a role in repair we have focused on neural progenitor cells (NPCs) since these are found in lesions in PPMS and have been found to influence oligodendrocyte progenitor cell maturation (OPCs). To better understand the cellular nature of NPCs in PPMS we developed iPS cells from blood samples of PPMS patients and age matched non-disease spouse or blood relative controls. Using these iPS cell lines we determined that the NPCs from PPMS cases provided no neuroprotection against active CNS demyelination compared to NPCs from control iPS lines which were capable of completely preventing injury. Conditioned media (CM) from PPMS NPCs provides no protection to OPCs and prevents maturation of OPCs into oligodendrocytes in vitro. We also found that CM from PPMS iPS NPCs elicited patient-specific differences in the response to compounds that should foster oligodendrocyte (OL) maturation. Together, these data establish a new model for understanding the nature of myelination defects in PPMS which may lead to novel targeted approaches for preventing demyelination in these patients.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Multiple Sclerosis, Chronic Progressive/pathology , Myelin Sheath/pathology , Aged , Animals , Apoptosis/drug effects , Axons/pathology , Axons/ultrastructure , Cell Differentiation/drug effects , Clemastine/pharmacology , Clemastine/therapeutic use , Culture Media, Conditioned/pharmacology , Cuprizone/toxicity , Female , Humans , Induced Pluripotent Stem Cells/chemistry , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/ultrastructure , Male , Mice, Inbred C57BL , Miconazole/pharmacology , Miconazole/therapeutic use , Middle Aged , Monoamine Oxidase Inhibitors/toxicity , Multiple Sclerosis, Chronic Progressive/chemically induced , Myelin Basic Protein/metabolism , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/metabolism , Oligodendroglia/drug effects , Oligodendroglia/pathology , Oligodendroglia/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL