Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 355
Filtrar
Más filtros

Tipo del documento
Publication year range
1.
J Neurosci ; 43(11): 1859-1870, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36725322

RESUMEN

Age-related decline in visual functions is a prevalent health problem among elderly people, and no effective therapies are available up-to-date. Axon degeneration and myelin loss in optic nerves (ONs) are age-dependent and become evident in middle-aged (13-18 months) and old (20-22 months) mice of either sex compared with adult mice (3-8 months), accompanied by functional deficits. Oligodendrocyte (OL) turnover is actively going on in adult ONs. However, the longitudinal change and functional significance of OL turnover in aging ONs remain largely unknown. Here, using cell-lineage labeling and tracing, we reported that oligodendrogenesis displayed an age-dependent decrease in aging ONs. To understand whether active OL turnover is required for maintaining axons and visual function, we conditionally deleted the transcription factor Olig2 in the oligodendrocyte precursor cells of young mice. Genetically dampening OL turnover by Olig2 ablation resulted in accelerated axon loss and retinal degeneration, and subsequently impaired ON signal transmission, suggesting that OL turnover is an important mechanism to sustain axon survival and visual function. To test whether enhancing oligodendrogenesis can prevent age-related visual deficits, 12-month-old mice were treated with clemastine, a pro-myelination drug, or induced deletion of the muscarinic receptor 1 in oligodendrocyte precursor cells. The clemastine treatment or muscarinic receptor 1 deletion significantly increased new OL generation in the aged ONs and consequently preserved visual function and retinal integrity. Together, our data indicate that dynamic OL turnover in ONs is required for axon survival and visual function, and enhancing new OL generation represents a potential approach to reversing age-related declines of visual function.SIGNIFICANCE STATEMENT Oligodendrocyte (OL) turnover has been reported in adult optic nerves (ONs), but the longitudinal change and functional significance of OL turnover during aging remain largely unknown. Using cell-lineage tracing and oligodendroglia-specific manipulation, this study reported that OL generation was active in adult ONs and the efficiency decreased in an age-dependent manner. Genetically dampening OL generation by Olig2 ablation resulted in significant axon loss and retinal degeneration, along with delayed visual signal transmission. Conversely, pro-myelination approaches significantly increased new myelin generation in aging ONs, and consequently preserved retinal integrity and visual function. Our findings indicate that promoting OL generation might be a promising strategy to preserve visual function from age-related decline.


Asunto(s)
Clemastina , Degeneración Retiniana , Ratones , Animales , Clemastina/farmacología , Oligodendroglía/fisiología , Vaina de Mielina/fisiología , Nervio Óptico , Axones , Diferenciación Celular/fisiología
2.
Glia ; 72(9): 1555-1571, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38829008

RESUMEN

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.


Asunto(s)
Clemastina , Glaucoma , Ratones Endogámicos C57BL , Remielinización , Retina , Animales , Clemastina/farmacología , Clemastina/uso terapéutico , Glaucoma/patología , Glaucoma/tratamiento farmacológico , Retina/patología , Retina/efectos de los fármacos , Remielinización/efectos de los fármacos , Remielinización/fisiología , Ratones , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , Modelos Animales de Enfermedad , Enfermedades del Nervio Óptico/tratamiento farmacológico , Enfermedades del Nervio Óptico/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología
3.
Neurobiol Dis ; 190: 106375, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38092269

RESUMEN

Patients with chronic pain often experience memory impairment, but the underlying mechanisms remain elusive. The myelin sheath is crucial for rapid and accurate action potential conduction, playing a pivotal role in the development of cognitive abilities in the central nervous system. The study reveals that myelin degradation occurs in the hippocampus of chronic constriction injury (CCI) mice, which display both chronic pain and memory impairment. Using fiber photometry, we observed diminished task-related neuronal activity in the hippocampus of CCI mice. Interestingly, the repeated administration with clemastine, which promotes myelination, counteracts the CCI-induced myelin loss and reduced neuronal activity. Notably, clemastine specifically ameliorates the impaired memory without affecting chronic pain in CCI mice. Overall, our findings highlight the significant role of myelin abnormalities in CCI-induced memory impairment, suggesting a potential therapeutic approach for treating memory impairments associated with neuropathic pain.


Asunto(s)
Dolor Crónico , Clemastina , Humanos , Animales , Ratones , Clemastina/metabolismo , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/metabolismo , Vaina de Mielina/metabolismo , Sistema Nervioso Central , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Hipocampo/metabolismo
4.
Anal Chem ; 96(17): 6599-6608, 2024 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-38640514

RESUMEN

Antihistamines relieve allergic symptoms by inhibiting the action of histamine. Further understanding of antihistamine transmembrane mechanisms and optimizing the selectivity and real-time monitoring capabilities of drug sensors is necessary. In this study, a micrometer liquid/liquid (L/L) interfacial sensor has served as a biomimetic membrane to investigate the mechanism of interfacial transfer of five antihistamines, i.e., clemastine (CLE), cyproheptadine (CYP), epinastine (EPI), desloratadine (DSL), and cetirizine (CET), and realize the real-time determinations. Cyclic voltammetry (CV) and differential pulse voltammetry (DPV) techniques have been used to uncover the electrochemical transfer behavior of the five antihistamines at the L/L interface. Additionally, finite element simulations (FEMs) have been employed to reveal the thermodynamics and kinetics of the process. Visualization of antihistamine partitioning in two phases at different pH values can be realized by ion partition diagrams (IPDs). The IPDs also reveal the transfer mechanism at the L/L interface and provide effective lipophilicity at different pH values. Real-time determinations of these antihistamines have been achieved through potentiostatic chronoamperometry (I-t), exhibiting good selectivity with the addition of nine common organic or inorganic compounds in living organisms and revealing the potential for in vivo pharmacokinetics. Besides providing a satisfactory surrogate for studying the transmembrane mechanism of antihistamines, this work also sheds light on micro- and nano L/L interfacial sensors for in vivo analysis of pharmacokinetics at a single-cell or single-organelle level.


Asunto(s)
Cetirizina , Clemastina , Ciproheptadina , Imidazoles , Loratadina , Loratadina/análogos & derivados , Loratadina/farmacología , Loratadina/análisis , Loratadina/química , Ciproheptadina/farmacología , Ciproheptadina/análogos & derivados , Ciproheptadina/análisis , Cetirizina/análisis , Cetirizina/farmacología , Cetirizina/química , Clemastina/análisis , Clemastina/farmacología , Clemastina/metabolismo , Antagonistas de los Receptores Histamínicos/farmacología , Antagonistas de los Receptores Histamínicos/química , Antagonistas de los Receptores Histamínicos/análisis , Antagonistas de los Receptores Histamínicos/metabolismo , Técnicas Electroquímicas/métodos , Biomimética , Dibenzazepinas/farmacología , Dibenzazepinas/química
5.
Mol Genet Metab ; 142(3): 108497, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38763041

RESUMEN

Krabbe disease (KD) is a rare inherited demyelinating disorder caused by a deficiency in the lysosomal enzyme galactosylceramide (GalCer) ß-galactosidase. Most patients with KD exhibit fatal cerebral demyelination with apoptotic oligodendrocyte (OL) death and die before the age of 2-4 years. We have previously reported that primary OLs isolated from the brains of twitcher (twi) mice, an authentic mouse model of KD, have cell-autonomous developmental defects and undergo apoptotic death accompanied by abnormal accumulation of psychosine, an endogenous cytotoxic lyso-derivative of GalCer. In this study, we aimed to investigate the effects of the preclinical promyelinating drugs clemastine and Sob-AM2 on KD OL pathologies using primary OLs isolated from the brains of twi mice. Both agents specifically prevented the apoptotic death observed in twi OLs. However, while Sob-AM2 showed higher efficacy in restoring the impaired differentiation and maturation of twi OLs, clemastine more potently reduced the endogenous psychosine levels. These results present the first preclinical in vitro data, suggesting that clemastine and Sob-AM2 can act directly and distinctly on OLs in KD and ameliorate their cellular pathologies associated with myelin degeneration.


Asunto(s)
Apoptosis , Clemastina , Modelos Animales de Enfermedad , Leucodistrofia de Células Globoides , Oligodendroglía , Psicosina , Animales , Leucodistrofia de Células Globoides/patología , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/tratamiento farmacológico , Oligodendroglía/patología , Oligodendroglía/metabolismo , Oligodendroglía/efectos de los fármacos , Ratones , Clemastina/farmacología , Apoptosis/efectos de los fármacos , Psicosina/análogos & derivados , Psicosina/metabolismo , Diferenciación Celular/efectos de los fármacos , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Encéfalo/patología , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Células Cultivadas
6.
Org Biomol Chem ; 22(9): 1812-1820, 2024 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-38328995

RESUMEN

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.


Asunto(s)
Antiprotozoarios , Leishmaniasis Cutánea , Humanos , Animales , Ratones , Clemastina/uso terapéutico , Macrófagos , Leishmaniasis Cutánea/tratamiento farmacológico , Leishmaniasis Cutánea/parasitología , Antiprotozoarios/farmacología , Células Hep G2 , Ratones Endogámicos BALB C
7.
Brain ; 146(8): 3331-3346, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37068912

RESUMEN

Pitt-Hopkins syndrome is an autism spectrum disorder caused by autosomal dominant mutations in the human transcription factor 4 gene (TCF4). One pathobiological process caused by murine Tcf4 mutation is a cell autonomous reduction in oligodendrocytes and myelination. In this study, we show that the promyelinating compounds, clemastine, sobetirome and Sob-AM2 are effective at restoring myelination defects in a Pitt-Hopkins syndrome mouse model. In vitro, clemastine treatment reduced excess oligodendrocyte precursor cells and normalized oligodendrocyte density. In vivo, 2-week intraperitoneal administration of clemastine also normalized oligodendrocyte precursor cell and oligodendrocyte density in the cortex of Tcf4 mutant mice and appeared to increase the number of axons undergoing myelination, as EM imaging of the corpus callosum showed a significant increase in the proportion of uncompacted myelin and an overall reduction in the g-ratio. Importantly, this treatment paradigm resulted in functional rescue by improving electrophysiology and behaviour. To confirm behavioural rescue was achieved via enhancing myelination, we show that treatment with the thyroid hormone receptor agonist sobetirome or its brain penetrating prodrug Sob-AM2, was also effective at normalizing oligodendrocyte precursor cell and oligodendrocyte densities and behaviour in the Pitt-Hopkins syndrome mouse model. Together, these results provide preclinical evidence that promyelinating therapies may be beneficial in Pitt-Hopkins syndrome and potentially other neurodevelopmental disorders characterized by dysmyelination.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Humanos , Animales , Ratones , Clemastina , Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/genética , Preparaciones Farmacéuticas , Discapacidad Intelectual/tratamiento farmacológico , Discapacidad Intelectual/genética
8.
Int J Mol Sci ; 25(15)2024 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-39125778

RESUMEN

Neonatal hypoxic-ischemic encephalopathy is the most common cause of long-term disability in term neonates, and white matter injury is the primary cause of cerebral palsy. Therapies that focus on the neuroprotection of myelination and oligodendrocyte proliferation could potentially ameliorate long-lasting neurological impairments after hypoxic-ischemic encephalopathy. Clemastine, a histamine H1 antagonist, has been shown to exert neuroprotective effects in multiple sclerosis and spinal cord injury by promoting oligodendrogenesis and re-myelination. In this study, we demonstrated the neuroprotective effects of clemastine in our rat model of neonatal hypoxic-ischemic brain injury. Animals received a single intraperitoneal injection of either vehicle or clemastine (10 mg/kg) for 6 consecutive days. Our results showed a significant reduction in white matter loss after treatment, with a clear effect of clemastine on oligodendrocytes, showing a significant increase in the number of Olig2+ cells. We characterized the MAPK/ERK pathway as a potential mechanistic pathway underlying the neuroprotective effects of clemastine. Altogether, our results demonstrate that clemastine is a potential compound for the treatment of hypoxic-ischemic encephalopathy, with a clear neuroprotective effect on white matter injury by promoting oligodendrogenesis.


Asunto(s)
Animales Recién Nacidos , Proliferación Celular , Clemastina , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica , Sistema de Señalización de MAP Quinasas , Fármacos Neuroprotectores , Oligodendroglía , Animales , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Ratas , Clemastina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ratas Sprague-Dawley
9.
Brain ; 145(11): 3943-3952, 2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-35678509

RESUMEN

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.


Asunto(s)
Esclerosis Múltiple , Remielinización , Humanos , Animales , Potenciales Evocados Visuales , Clemastina/uso terapéutico , Vaina de Mielina/metabolismo , Esclerosis Múltiple/patología , Biomarcadores/metabolismo
10.
Proc Natl Acad Sci U S A ; 117(11): 5810-5817, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32127489

RESUMEN

The antihistamine clemastine inhibits multiple stages of the Plasmodium parasite that causes malaria, but the molecular targets responsible for its parasite inhibition were unknown. Here, we applied parallel chemoproteomic platforms to discover the mechanism of action of clemastine and identify that clemastine binds to the Plasmodium falciparum TCP-1 ring complex or chaperonin containing TCP-1 (TRiC/CCT), an essential heterooligomeric complex required for de novo cytoskeletal protein folding. Clemastine destabilized all eight P. falciparum TRiC subunits based on thermal proteome profiling (TPP). Further analysis using stability of proteins from rates of oxidation (SPROX) revealed a clemastine-induced thermodynamic stabilization of the Plasmodium TRiC delta subunit, suggesting an interaction with this protein subunit. We demonstrate that clemastine reduces levels of the major TRiC substrate tubulin in P. falciparum parasites. In addition, clemastine treatment leads to disorientation of Plasmodium mitotic spindles during the asexual reproduction and results in aberrant tubulin morphology suggesting protein aggregation. This clemastine-induced disruption of TRiC function is not observed in human host cells, demonstrating a species selectivity required for targeting an intracellular human pathogen. Our findings encourage larger efforts to apply chemoproteomic methods to assist in target identification of antimalarial drugs and highlight the potential to selectively target Plasmodium TRiC-mediated protein folding for malaria intervention.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Clemastina/farmacología , Antagonistas de los Receptores Histamínicos/farmacología , Proteínas Protozoarias/metabolismo , Sitios de Unión , Línea Celular , Chaperonina con TCP-1/química , Humanos , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Unión Proteica , Proteínas Protozoarias/química , Huso Acromático/efectos de los fármacos
11.
Neurochem Res ; 47(2): 503-515, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34661796

RESUMEN

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.


Asunto(s)
Clemastina , Traumatismos de la Médula Espinal , Animales , Clemastina/metabolismo , Clemastina/farmacología , Clemastina/uso terapéutico , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Ratas , Recuperación de la Función , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología
12.
Med Sci Monit ; 28: e936367, 2022 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-35651299

RESUMEN

BACKGROUND Perioperative hemodynamic instability mediated by anaphylaxis is a life-threatening complication in patients undergoing cardiac surgery with cardiopulmonary bypass (CPB). This study aimed to evaluate the effect of clemastine fumarate in this specific patient population. MATERIAL AND METHODS We enrolled 100 participants who met the inclusion criteria and randomly allocated them to the treatment group and the placebo group. Participants in the treatment group and the placebo group were treated separately with an injection of clemastine fumarate and saline, respectively. Plasma histamine concentration and blood pressure were quantified at 5 timepoints during the perioperative period, and differences between the 2 groups were assessed by repeated-measures ANOVA. The postoperative complications and in-hospital mortality also were evaluated. All participants were followed up for 7 days after cardiac surgery. RESULTS Plasma histamine concentrations increased in both groups but were statistically significantly lower in the treatment group during the perioperative period (P=0.007). Diastolic blood pressure (P=0.014) and mean arterial pressure (P=0.024) in the treatment group were significantly higher than in the placebo group during the perioperative period. The coefficients of variation for systolic (13.9±4.2% vs 17.2±4.4%, P<0.01) and diastolic (12.9±4.9% vs 15.3±5.2%, P=0.02) blood pressure were significantly lower in the treatment group compared with the placebo group. CONCLUSIONS Pretreatment with clemastine fumarate restrains the increase in histamine concentration and provides safer hemodynamics in patients undergoing cardiac surgery with CPB.


Asunto(s)
Clemastina , Hemodinámica , Enfermedades Vasculares , Anafilaxia , Puente Cardiopulmonar , Clemastina/efectos adversos , Clemastina/farmacología , Hemodinámica/efectos de los fármacos , Histamina , Humanos , Atención Perioperativa , Enfermedades Vasculares/tratamiento farmacológico
13.
Br J Cancer ; 124(10): 1647-1652, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33762718

RESUMEN

BACKGROUND: Ranitidine, a histamine 2 blocker, is the standard of care to prevent hypersensitivity reactions (HSRs) caused by paclitaxel infusion. However, the added value of ranitidine in this premedication regimen is controversial. Therefore, we compared the incidence of HSRs during paclitaxel treatment between a standard regimen including ranitidine and a regimen without ranitidine. METHODS: This prospective, pre-post interventional, non-inferiority study compared the standard premedication regimen (N = 183) with dexamethasone, clemastine and ranitidine with a premedication regimen without ranitidine (N = 183). The primary outcome was the incidence of HSR grade ≥3. Non-inferiority was determined by checking whether the upper bound of the two-sided 90% confidence interval (CI) for the difference in HSR rates excluded the +6% non-inferiority margin. RESULTS: In both the pre-intervention (with ranitidine) and post-intervention (without ranitidine) group 183 patients were included. The incidence of HSR grade ≥3 was 4.4% (N = 8) in the pre-intervention group and 1.6% (N = 3) in the post-intervention group: difference -2.7% (90% CI: -6.2 to 0.1). CONCLUSIONS: As the upper boundary of the 90% CI does not exceed the predefined non-inferiority margin of +6%, it can be concluded that a premedication regimen without ranitidine is non-inferior to a premedication regimen with ranitidine. CLINICAL TRIAL REGISTRATION: www.trialregister.nl ; NL8173.


Asunto(s)
Hipersensibilidad a las Drogas/prevención & control , Neoplasias/tratamiento farmacológico , Paclitaxel/efectos adversos , Premedicación/métodos , Ranitidina/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Quimioprevención/efectos adversos , Quimioprevención/métodos , Clemastina/administración & dosificación , Dexametasona/administración & dosificación , Hipersensibilidad a las Drogas/epidemiología , Hipersensibilidad a las Drogas/patología , Quimioterapia Combinada , Estudios de Equivalencia como Asunto , Femenino , Antagonistas de los Receptores H2 de la Histamina/administración & dosificación , Antagonistas de los Receptores H2 de la Histamina/uso terapéutico , Humanos , Infusiones Intravenosas , Masculino , Inutilidad Médica , Persona de Mediana Edad , Neoplasias/epidemiología , Neoplasias/patología , Países Bajos/epidemiología , Paclitaxel/administración & dosificación , Premedicación/efectos adversos , Ranitidina/administración & dosificación , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
14.
Int J Med Sci ; 18(3): 639-645, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33437198

RESUMEN

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.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Clemastina/farmacología , Mediadores de Inflamación/metabolismo , Fármacos Neuroprotectores/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Edema Encefálico/inmunología , Edema Encefálico/patología , Células Cultivadas , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/inmunología , Hemorragia Cerebral/patología , Clemastina/uso terapéutico , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Cultivo Primario de Células , Técnicas Estereotáxicas
15.
Int J Mol Sci ; 23(1)2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-35008492

RESUMEN

Apicomplexan parasites, such as Toxoplasma gondii, Plasmodium spp., Babesia spp., and Cryptosporidium spp., cause significant morbidity and mortality. Existing treatments are problematic due to toxicity and the emergence of drug-resistant parasites. Because protozoan tubulin can be selectively disrupted by small molecules to inhibit parasite growth, we assembled an in vitro testing cascade to fully delineate effects of candidate tubulin-targeting drugs on Toxoplasma gondii and vertebrate host cells. Using this analysis, we evaluated clemastine, an antihistamine that has been previously shown to inhibit Plasmodium growth by competitively binding to the CCT/TRiC tubulin chaperone as a proof-of-concept. We concurrently analyzed astemizole, a distinct antihistamine that blocks heme detoxification in Plasmodium. Both drugs have EC50 values of ~2 µM and do not demonstrate cytotoxicity or vertebrate microtubule disruption at this concentration. Parasite subpellicular microtubules are shortened by treatment with either clemastine or astemizole but not after treatment with pyrimethamine, indicating that this effect is not a general response to antiparasitic drugs. Immunoblot quantification indicates that the total α-tubulin concentration of 0.02 pg/tachyzoite does not change with clemastine treatment. In conclusion, the testing cascade allows profiling of small-molecule effects on both parasite and vertebrate cell viability and microtubule integrity.


Asunto(s)
Antiparasitarios/farmacología , Apicoplastos/efectos de los fármacos , Clemastina/farmacología , Parásitos/efectos de los fármacos , Tubulina (Proteína)/metabolismo , Animales , Células Cultivadas , Antagonistas de los Receptores Histamínicos/farmacología , Humanos , Microtúbulos/metabolismo , Proteínas Protozoarias/metabolismo
16.
J Neuroinflammation ; 17(1): 57, 2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-32061255

RESUMEN

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.


Asunto(s)
Clemastina/farmacología , Enfermedades Desmielinizantes/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Interleucina-1beta/metabolismo , Microglía/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Clemastina/uso terapéutico , Antagonistas de los Receptores Histamínicos H1/farmacología , Antagonistas de los Receptores Histamínicos H1/uso terapéutico , Masculino , Microglía/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
17.
J Pharmacol Sci ; 144(3): 123-128, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32921393

RESUMEN

We assessed the torsadogenic effects of a novel remyelinating drug clemastine for multiple sclerosis using an in vivo proarrhythmia model of acute atrioventricular block rabbit, since the drug has been demonstrated to suppress the human ether-á-go-go related gene (hERG) K+ channels. Bradycardia was induced by atrioventricular node ablation in isoflurane-anesthetized New Zealand White rabbits (n = 5), and the ventricle was electrically driven at 60 beats/min throughout the experiment, except when extrasystoles appeared. Intravenous administration of clinically relevant dose of 0.03 mg/kg of clemastine and 10-times higher dose of 0.3 mg/kg hardly affected the QT interval or duration of the monophasic action potential (MAP) of the ventricle. Additional administration of clemastine at 3 mg/kg significantly increased the QT interval, MAP duration and the short-term variability of repolarization. Meanwhile, the premature ventricular contractions with R on T phenomenon were observed in 3 out of 5 animals, and torsades de pointes arrhythmias were detected in 1 out of 5 animals. These results suggest that the torsadogenic potential of clemastine is obviously observed in the acute atrioventricular block rabbit, which will not appear within the prescribed dose for multiple sclerosis.


Asunto(s)
Clemastina/administración & dosificación , Clemastina/efectos adversos , Síndrome de QT Prolongado/inducido químicamente , Esclerosis Múltiple/tratamiento farmacológico , Torsades de Pointes/inducido químicamente , Potenciales de Acción/efectos de los fármacos , Animales , Bloqueo Atrioventricular/fisiopatología , Bradicardia/fisiopatología , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1/antagonistas & inhibidores , Ventrículos Cardíacos/efectos de los fármacos , Infusiones Intravenosas , Conejos
18.
Cephalalgia ; 39(5): 597-607, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30165750

RESUMEN

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.


Asunto(s)
Clemastina/uso terapéutico , Antagonistas de los Receptores Histamínicos H1/uso terapéutico , Trastornos Migrañosos/inducido químicamente , Trastornos Migrañosos/prevención & control , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/efectos adversos , Adulto , Estudios Cruzados , Método Doble Ciego , Femenino , Cefalea/inducido químicamente , Cefalea/prevención & control , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
19.
Brain ; 141(1): 85-98, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29244098

RESUMEN

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.


Asunto(s)
Clemastina/uso terapéutico , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/etiología , Antagonistas de los Receptores Histamínicos H1/uso terapéutico , Hipoxia Encefálica/complicaciones , Recuperación de la Función/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/ultraestructura , Enfermedades Desmielinizantes/diagnóstico por imagen , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Hipoxia Encefálica/diagnóstico por imagen , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/ultraestructura , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Nervio Óptico/fisiopatología , Oxígeno/farmacología , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo
20.
Lancet ; 390(10111): 2481-2489, 2017 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-29029896

RESUMEN

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.


Asunto(s)
Clemastina/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Adulto , Estudios Cruzados , Método Doble Ciego , Potenciales Evocados Visuales/efectos de los fármacos , Femenino , Humanos , Masculino , Remielinización/efectos de los fármacos , Tomografía de Coherencia Óptica , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda