Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
J Neurosci ; 44(28)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38749703

RESUMEN

Dysregulation of oligodendrocyte progenitor cell (OPC) recruitment and oligodendrocyte differentiation contribute to failure of remyelination in human demyelinating diseases such as multiple sclerosis (MS). Deletion of muscarinic receptor enhances OPC differentiation and remyelination. However, the role of ligand-dependent signaling versus constitutive receptor activation is unknown. We hypothesized that dysregulated acetylcholine (ACh) release upon demyelination contributes to ligand-mediated activation hindering myelin repair. Following chronic cuprizone (CPZ)-induced demyelination (male and female mice), we observed a 2.5-fold increase in ACh concentration. This increase in ACh concentration could be attributed to increased ACh synthesis or decreased acetylcholinesterase-/butyrylcholinesterase (BChE)-mediated degradation. Using choline acetyltransferase (ChAT) reporter mice, we identified increased ChAT-GFP expression following both lysolecithin and CPZ demyelination. ChAT-GFP expression was upregulated in a subset of injured and uninjured axons following intraspinal lysolecithin-induced demyelination. In CPZ-demyelinated corpus callosum, ChAT-GFP was observed in Gfap+ astrocytes and axons indicating the potential for neuronal and astrocytic ACh release. BChE expression was significantly decreased in the corpus callosum following CPZ demyelination. This decrease was due to the loss of myelinating oligodendrocytes which were the primary source of BChE. To determine the role of ligand-mediated muscarinic signaling following lysolecithin injection, we administered neostigmine, a cholinesterase inhibitor, to artificially raise ACh. We identified a dose-dependent decrease in mature oligodendrocyte density with no effect on OPC recruitment. Together, these results support a functional role of ligand-mediated activation of muscarinic receptors following demyelination and suggest that dysregulation of ACh homeostasis directly contributes to failure of remyelination in MS.


Asunto(s)
Enfermedades Desmielinizantes , Oligodendroglía , Transducción de Señal , Animales , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Ratones , Oligodendroglía/metabolismo , Oligodendroglía/efectos de los fármacos , Femenino , Masculino , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Ratones Endogámicos C57BL , Acetilcolina/metabolismo , Cuprizona/toxicidad , Lisofosfatidilcolinas/toxicidad , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Colina O-Acetiltransferasa/metabolismo , Remielinización/fisiología , Remielinización/efectos de los fármacos , Vaina de Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Ratones Transgénicos
2.
Brain Stimul ; 17(3): 575-587, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38648972

RESUMEN

BACKGROUND: Current treatments for Multiple Sclerosis (MS) poorly address chronic innate neuroinflammation nor do they offer effective remyelination. The vagus nerve has a strong regulatory role in inflammation and Vagus Nerve Stimulation (VNS) has potential to affect both neuroinflammation and remyelination in MS. OBJECTIVE: This study investigated the effects of VNS on demyelination and innate neuroinflammation in a validated MS rodent model. METHODS: Lysolecithin (LPC) was injected in the corpus callosum (CC) of 46 Lewis rats, inducing a demyelinated lesion. 33/46 rats received continuously-cycled VNS (cVNS) or one-minute per day VNS (1minVNS) or sham VNS from 2 days before LPC-injection until perfusion at 3 days post-injection (dpi) (corresponding with a demyelinated lesion with peak inflammation). 13/46 rats received cVNS or sham from 2 days before LPC-injection until perfusion at 11 dpi (corresponding with a partial remyelinated lesion). Immunohistochemistry and proteomics analyses were performed to investigate the extend of demyelination and inflammation. RESULTS: Immunohistochemistry showed that cVNS significantly reduced microglial and astrocytic activation in the lesion and lesion border, and significantly reduced the Olig2+ cell count at 3 dpi. Furthermore, cVNS significantly improved remyelination with 57.4 % versus sham at 11 dpi. Proteomic gene set enrichment analyses showed increased activation of (glutamatergic) synapse pathways in cVNS versus sham, most pronounced at 3 dpi. CONCLUSION: cVNS improved remyelination of an LPC-induced lesion. Possible mechanisms might include modulation of microglia and astrocyte activity, increased (glutamatergic) synapses and enhanced oligodendrocyte clearance after initial injury.


Asunto(s)
Enfermedades Desmielinizantes , Lisofosfatidilcolinas , Ratas Endogámicas Lew , Remielinización , Estimulación del Nervio Vago , Animales , Ratas , Remielinización/fisiología , Remielinización/efectos de los fármacos , Lisofosfatidilcolinas/toxicidad , Enfermedades Desmielinizantes/terapia , Enfermedades Desmielinizantes/inducido químicamente , Estimulación del Nervio Vago/métodos , Masculino , Enfermedades Neuroinflamatorias/terapia , Enfermedades Neuroinflamatorias/inducido químicamente , Enfermedades Neuroinflamatorias/etiología , Modelos Animales de Enfermedad , Esclerosis Múltiple/terapia , Esclerosis Múltiple/inducido químicamente , Cuerpo Calloso
3.
Pain ; 164(2): e103-e115, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36638307

RESUMEN

ABSTRACT: Tissue injuries, including burns, are major causes of death and morbidity worldwide. These injuries result in the release of intracellular molecules and subsequent inflammatory reactions, changing the tissues' chemical milieu and leading to the development of persistent pain through activating pain-sensing primary sensory neurons. However, the majority of pain-inducing agents in injured tissues are unknown. Here, we report that, amongst other important metabolite changes, lysophosphatidylcholines (LPCs) including 18:0 LPC exhibit significant and consistent local burn injury-induced changes in concentration. 18:0 LPC induces immediate pain and the development of hypersensitivities to mechanical and heat stimuli through molecules including the transient receptor potential ion channel, vanilloid subfamily, member 1, and member 2 at least partly via increasing lateral pressure in the membrane. As levels of LPCs including 18:0 LPC increase in other tissue injuries, our data reveal a novel role for these lipids in injury-associated pain. These findings have high potential to improve patient care.


Asunto(s)
Lisofosfatidilcolinas , Dolor , Humanos , Lisofosfatidilcolinas/toxicidad
4.
Cereb Cortex ; 33(4): 1403-1411, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35368059

RESUMEN

It has been shown that transcranial ultrasound stimulation (TUS) is capable of attenuating myelin loss and providing neuroprotection in animal models of brain disorders. In this study, we investigated the ability of TUS to promote remyelination in the lysolecithin (LPC)-induced local demyelination in the hippocampus. Demyelination was induced by the micro-injection of 1.5 µL LPC (1%) into the rat hippocampus and the treated group received daily TUS for 5 or 12 days. Magnetic resonance imaging techniques, including magnetization transfer ratio (MTR) and T2-weighted imaging, were used to longitudinally characterize the demyelination model. Furthermore, the therapeutic effects of TUS on LPC-induced demyelination were assessed by Luxol fast blue (LFB) staining. Our data revealed that reductions in MTR values observed during demyelination recover almost completely upon remyelination. The MTR values in demyelinated lesions were significantly higher in TUS-treated rats than in the LPC-only group after undergoing TUS. Form histological observation, TUS significantly reduced the size of demyelinated lesion 7 days after LPC administration. This study demonstrated that MTR was a sensitive and reproducible quantitative marker to assess remyelination process in vivo during TUS treatment. These findings might open new promising treatment strategies for demyelinating diseases such as multiple sclerosis.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Remielinización , Ratas , Animales , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/terapia , Esclerosis Múltiple/patología , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/diagnóstico por imagen , Enfermedades Desmielinizantes/terapia , Lisofosfatidilcolinas/toxicidad , Modelos Animales , Vaina de Mielina , Modelos Animales de Enfermedad
5.
Exp Neurol ; 359: 114262, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36343678

RESUMEN

Multiple sclerosis (MS) is an autoimmune demyelinating disease that leads to axon degeneration as the major cause of everlasting neurological disability. The cis-phosphorylated tau (cis-p-tau) is an isoform of tau phosphorylated on threonine 231 and causes tau fails to bind micro-tubules and promotes assembly. It gains toxic function and forms tangles in the cell which finally leads to cell death. An antibody raised against cis- p-tau (cis mAb) detects this isoform and induces its clearance. Here, we investigated the formation of cis-p-tau in a lysophosphatidylcholine (LPC)-induced prolonged demyelination model as well as the beneficial effects of its clearance using cis mAb. Cis -p-tau was increased in the lesion site, especially in axons and microglia. Behavioral and functional studies were performed using visual cliff test, visual placing test, and visual evoked potential recording. Cis-p-tau clearance resulted in decreased gliosis, protected myelin and reduced axon degeneration. Analysis of behavioral and electrophysiological data showed that clearance of cis-p-tau by cis mAb treatment improved the visual acuity along with the integrity of the optic pathway. Our results highlight the opportunity of using cis mAb as a new therapy for protecting myelin and axons in patients suffering from MS.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Ratones , Animales , Lisofosfatidilcolinas/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Quiasma Óptico/metabolismo , Quiasma Óptico/patología , Potenciales Evocados Visuales , Vaina de Mielina/patología , Esclerosis Múltiple/patología
6.
Environ Toxicol ; 37(11): 2756-2763, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36214341

RESUMEN

Lysophosphatidylcholine (LPC), a major class of glycerophospholipids ubiquitously present in most tissues, plays a dominant role in many diseases, while it is still unknown about the potential mechanism of LPC affecting the testicular Leydig cells. In the present study, mouse TM3 Leydig cells in vitro were treated with LPC for 48 h. LPC was found to significantly induce apoptosis and oxidative stress of mouse TM3 Leydig cells; while inhibition of oxidative stress by N-acetyl-L-cysteine, an inhibitor of oxidative stress, could rescue the induction of apoptosis, indicating that LPC induced apoptosis of mouse TM3 Leydig cells via oxidative stress. Interestingly, LPC was showed to inhibit autophagy; however, induction of autophagy by rapamycin significantly alleviated the induction of apoptosis by LPC. Taken together, oxidative stress was involved in LPC-induced apoptosis of mouse TM3 Leydig cells, and autophagy might play a protective role in LPC-induced apoptosis.


Asunto(s)
Células Intersticiales del Testículo , Lisofosfatidilcolinas , Acetilcisteína , Animales , Apoptosis , Autofagia , Glicerofosfolípidos/metabolismo , Células Intersticiales del Testículo/metabolismo , Lisofosfatidilcolinas/metabolismo , Lisofosfatidilcolinas/toxicidad , Masculino , Ratones , Estrés Oxidativo , Sirolimus
7.
J Neuroinflammation ; 19(1): 194, 2022 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-35902863

RESUMEN

BACKGROUND: Demyelinating diseases in central nervous system (CNS) are a group of diseases characterized by myelin damage or myelin loss. Transforming growth factor beta1 (TGF-ß1) is widely recognized as an anti-inflammatory cytokine, which can be produced by both glial and neuronal cells in CNS. However, the effects of TGF-ß1 on demyelinating diseases and its underlying mechanisms have not been well investigated. METHODS: A demyelinating mouse model using two-point injection of lysophosphatidylcholine (LPC) to the corpus callosum in vivo was established. Exogenous TGF-ß1 was delivered to the lesion via brain stereotactic injection. LFB staining, immunofluorescence, and Western blot were applied to examine the severity of demyelination and pyroptosis process in microglia. Morris water maze test was used to assess the cognitive abilities of experimental mice. Furthermore, lipopolysaccharide (LPS) was applied to induce pyroptosis in primary cultured microglia in vitro, to explore potential molecular mechanism. RESULTS: The degree of demyelination in LPC-modeling mice was found improved with supplement of TGF-ß1. Besides, TGF-ß1 treatment evidently ameliorated the activated proinflammatory pyroptosis of microglia, with downregulated levels of the key pyroptosis effector Gasdermin D (GSDMD), inflammasomes, and cleaved-IL-1ß, which effectively attenuated neuroinflammation in vivo. Evaluated by behavioral tests, the cognitive deficit in LPC-modeling mice was found mitigated with application of TGF-ß1. Mechanistically, TGF-ß1 could reverse pyroptosis-like morphology in LPS-stimulated primary cultured microglia observed by scanning electron microscopy, as well as decrease the protein levels of cleaved-GSDMD, inflammasomes, and cleaved-IL-1ß. Activation of ERK1/2 and NF-κB pathways largely abolished the protective effects of TGF-ß1, which indicated that TGF-ß1 alleviated the pyroptosis possibly via regulating NF-κB/ERK1/2 signal pathways. CONCLUSIONS: Our studies demonstrated TGF-ß1 notably relieved the demyelinating injury and cognitive disorder in LPC-modeling mice, by attenuating the inflammatory pyroptosis of microglia via ERK1/2 and NF-κB pathways. Targeting TGF-ß1 activity might serve as a promising therapeutic strategy in demyelinating diseases.


Asunto(s)
Enfermedades Desmielinizantes , FN-kappa B , Animales , Cognición , Enfermedades Desmielinizantes/patología , Inflamasomas/metabolismo , Lipopolisacáridos/farmacología , Lisofosfatidilcolinas/toxicidad , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Microglía/metabolismo , FN-kappa B/metabolismo , Piroptosis/fisiología , Factor de Crecimiento Transformador beta1/metabolismo
8.
Pain ; 163(10): 1999-2013, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35086123

RESUMEN

ABSTRACT: Rheumatic diseases are often associated to debilitating chronic pain, which remains difficult to treat and requires new therapeutic strategies. We had previously identified lysophosphatidylcholine (LPC) in the synovial fluids from few patients and shown its effect as a positive modulator of acid-sensing ion channel 3 (ASIC3) able to induce acute cutaneous pain in rodents. However, the possible involvement of LPC in chronic joint pain remained completely unknown. Here, we show, from 2 independent cohorts of patients with painful rheumatic diseases, that the synovial fluid levels of LPC are significantly elevated, especially the LPC16:0 species, compared with postmortem control subjects. Moreover, LPC16:0 levels correlated with pain outcomes in a cohort of osteoarthritis patients. However, LPC16:0 do not appear to be the hallmark of a particular joint disease because similar levels are found in the synovial fluids of a second cohort of patients with various rheumatic diseases. The mechanism of action was next explored by developing a pathology-derived rodent model. Intra-articular injections of LPC16:0 is a triggering factor of chronic joint pain in both male and female mice, ultimately leading to persistent pain and anxiety-like behaviors. All these effects are dependent on ASIC3 channels, which drive sufficient peripheral inputs to generate spinal sensitization processes. This study brings evidences from mouse and human supporting a role for LPC16:0 via ASIC3 channels in chronic pain arising from joints, with potential implications for pain management in osteoarthritis and possibly across other rheumatic diseases.


Asunto(s)
Canales Iónicos Sensibles al Ácido , Dolor Crónico , Osteoartritis , Canales Iónicos Sensibles al Ácido/metabolismo , Animales , Artralgia/etiología , Femenino , Humanos , Lisofosfatidilcolinas/toxicidad , Masculino , Ratones , Osteoartritis/complicaciones
9.
Can J Physiol Pharmacol ; 100(2): 107-116, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34935529

RESUMEN

Demyelination disorder is an unusual pathologic event, which occurs in the central nervous system (CNS). Multiple sclerosis (MS) is an inflammatory demyelinating disease that affects the CNS, and it is the leading cause of disability in young adults. Lysolecithin (LPC) is one of the best toxin-induced demyelination models. In this study, a suitable model is created, and the effect of fluoxetine treatment is examined on this model. In this case, it was assumed that daily fluoxetine treatment had increased the endogenous remyelination in the LPC model. This study was focused on investigating the influence of the fluoxetine dose of 5 or 10 mg/kg per day for 1 and 4 weeks on LPC-induced neurotoxicity in the corpus callosum region. It was performed as a demyelinating model in male Wistar rats. After 3 days, fluoxetine was injected intraperitoneally (5 or 10 mg/kg per day) for 1 and 4 weeks in each group. After completing the treatment course, the corpus callosum was removed to examine the gene expression and histological analysis was performed. The results of the histopathological study of hematoxylin and eosin staining of the corpus callosum showed that in 1 and 4-week treatment groups, fluoxetine has reduced the level of inflammation at the LPC injection site (5 and 10 mg/kg per day). Fluoxetine treatment in the luxol fast blue (LFB) staining of the corpus callosum has been led to an increase in myelination capacity in all doses and times. The results of the genetic study showed that the fluoxetine has significantly reduced the expression level of tumor necrosis factor-α, nuclear factor κß, and induced nitric oxide synthase in comparison with the untreated LPC group. Also, the fluoxetine treatment has enhanced the expression level of the forkhead box P3 (FOXP3) gene in comparison with the untreated group. Fluoxetine has increased the expression level of myelination and neurotrophic genes such as myelin basic protein (MBP), oligodendrocyte transcription factor 2 (OLIG2), and brain-derived neurotrophic factor (BDNF). The outcomes demonstrated that fluoxetine reduces inflammation and strengthens the endogenous myelination in the LPC-induced demyelination model; however, supplementary studies are required for specifying the details of its mechanisms.


Asunto(s)
Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Modelos Animales de Enfermedad , Fluoxetina/uso terapéutico , Lisofosfatidilcolinas/efectos adversos , Lisofosfatidilcolinas/toxicidad , Ratas Wistar , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Fluoxetina/administración & dosificación , Fluoxetina/farmacología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Expresión Génica/efectos de los fármacos , Masculino , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/genética , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
10.
FASEB J ; 35(10): e21867, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34499764

RESUMEN

Obesity alters skeletal muscle lipidome and promotes myopathy, but it is unknown whether aberrant muscle lipidome contributes to the reduction in skeletal muscle contractile force-generating capacity. Comprehensive lipidomic analyses of mouse skeletal muscle revealed a very strong positive correlation between the abundance of lysophosphatidylcholine (lyso-PC), a class of lipids that is known to be downregulated with obesity, with maximal tetanic force production. The level of lyso-PC is regulated primarily by lyso-PC acyltransferase 3 (LPCAT3), which acylates lyso-PC to form phosphatidylcholine. Tamoxifen-inducible skeletal muscle-specific overexpression of LPCAT3 (LPCAT3-MKI) was sufficient to reduce muscle lyso-PC content in both standard chow diet- and high-fat diet (HFD)-fed conditions. Strikingly, the assessment of skeletal muscle force-generating capacity ex vivo revealed that muscles from LPCAT3-MKI mice were weaker regardless of diet. Defects in force production were more apparent in HFD-fed condition, where tetanic force production was 40% lower in muscles from LPCAT3-MKI compared to that of control mice. These observations were partly explained by reductions in the cross-sectional area in type IIa and IIx fibers, and signs of muscle edema in the absence of fibrosis. Future studies will pursue the mechanism by which LPCAT3 may alter protein turnover to promote myopathy.


Asunto(s)
1-Acilglicerofosfocolina O-Aciltransferasa/fisiología , Dieta Alta en Grasa/efectos adversos , Lipidómica/métodos , Lisofosfatidilcolinas/toxicidad , Músculo Esquelético/patología , Enfermedades Musculares/patología , Obesidad/fisiopatología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Muscular , Músculo Esquelético/efectos de los fármacos , Enfermedades Musculares/etiología , Enfermedades Musculares/metabolismo
11.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34362845

RESUMEN

Excessive activation of T cells and microglia represents a hallmark of the pathogenesis of human multiple sclerosis (MS). However, the regulatory molecules overactivating these immune cells remain to be identified. Previously, we reported that extracellular IFP35 family proteins, including IFP35 and NMI, activated macrophages as proinflammatory molecules in the periphery. Here, we investigated their functions in the process of neuroinflammation both in the central nervous system (CNS) and the periphery. Our analysis of clinical transcriptomic data showed that expression of IFP35 family proteins was up-regulated in patients with MS. Additional in vitro studies demonstrated that IFP35 and NMI were released by multiple cells. IFP35 and NMI subsequently triggered nuclear factor kappa B-dependent activation of microglia via the TLR4 pathway. Importantly, we showed that both IFP35 and NMI activated dendritic cells and promoted naïve T cell differentiation into Th1 and Th17 cells. Nmi-/- , Ifp35-/- , or administration of neutralizing antibodies against IFP35 alleviated the immune cells' infiltration and demyelination in the CNS, thus reducing the severity of experimental autoimmune encephalomyelitis. Together, our findings reveal a hitherto unknown mechanism by which IFP35 family proteins facilitate overactivation of both T cells and microglia and propose avenues to study the pathogenesis of MS.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/sangre , Péptidos y Proteínas de Señalización Intracelular/genética , Esclerosis Múltiple/patología , Enfermedades Neuroinflamatorias/patología , Animales , Anticuerpos Neutralizantes/farmacología , Estudios de Casos y Controles , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisofosfatidilcolinas/toxicidad , Ratones Endogámicos C57BL , Ratones Mutantes , Microglía/metabolismo , Microglía/patología , Esclerosis Múltiple/genética , Enfermedades Neuroinflamatorias/genética , Células Th17/inmunología , Células Th17/metabolismo
12.
Clin Sci (Lond) ; 135(15): 1845-1858, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34269800

RESUMEN

OBJECTIVE: The mechanisms involved in NOX5 activation in atherosclerotic processes are not completely understood. The present study tested the hypothesis that lysophosphatidylcholine (LPC), a proatherogenic component of oxLDL, induces endothelial calcium influx, which drives NOX5-dependent reactive oxygen species (ROS) production, oxidative stress, and endothelial cell dysfunction. APPROACH: Human aortic endothelial cells (HAEC) were stimulated with LPC (10-5 M, for different time points). Pharmacological inhibition of NOX5 (Melittin, 10-7 M) and NOX5 gene silencing (siRNA) was used to determine the role of NOX5-dependent ROS production in endothelial oxidative stress induced by LPC. ROS production was determined by lucigenin assay and electron paramagnetic spectroscopy (EPR), calcium transients by Fluo4 fluorimetry, and NOX5 activity and protein expression by pharmacological assays and immunoblotting, respectively. RESULTS: LPC increased ROS generation in endothelial cells at short (15 min) and long (4 h) stimulation times. LPC-induced ROS was abolished by a selective NOX5 inhibitor and by NOX5 siRNA. NOX1/4 dual inhibition and selective NOX1 inhibition only decreased ROS generation at 4 h. LPC increased HAEC intracellular calcium, important for NOX5 activation, and this was blocked by nifedipine and thapsigargin. Bapta-AM, selective Ca2+ chelator, prevented LPC-induced ROS production. NOX5 knockdown decreased LPC-induced ICAM-1 mRNA expression and monocyte adhesion to endothelial cells. CONCLUSION: These results suggest that NOX5, by mechanisms linked to increased intracellular calcium, is key to early LPC-induced endothelial oxidative stress and pro-inflammatory processes. Since these are essential events in the formation and progression of atherosclerotic lesions, the present study highlights an important role for NOX5 in atherosclerosis.


Asunto(s)
Aterosclerosis/enzimología , Células Endoteliales/efectos de los fármacos , Lisofosfatidilcolinas/toxicidad , NADPH Oxidasa 5/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Aterosclerosis/patología , Calcio/metabolismo , Señalización del Calcio , Adhesión Celular , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/enzimología , Células Endoteliales/patología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Monocitos/metabolismo , NADPH Oxidasa 5/antagonistas & inhibidores , NADPH Oxidasa 5/genética , Interferencia de ARN
13.
Nat Commun ; 12(1): 2265, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859199

RESUMEN

Nerve-glia (NG2) glia or oligodendrocyte precursor cells (OPCs) are distributed throughout the gray and white matter and generate myelinating cells. OPCs in white matter proliferate more than those in gray matter in response to platelet-derived growth factor AA (PDGF AA), despite similar levels of its alpha receptor (PDGFRα) on their surface. Here we show that the type 1 integral membrane protein neuropilin-1 (Nrp1) is expressed not on OPCs but on amoeboid and activated microglia in white but not gray matter in an age- and activity-dependent manner. Microglia-specific deletion of Nrp1 compromised developmental OPC proliferation in white matter as well as OPC expansion and subsequent myelin repair after acute demyelination. Exogenous Nrp1 increased PDGF AA-induced OPC proliferation and PDGFRα phosphorylation on dissociated OPCs, most prominently in the presence of suboptimum concentrations of PDGF AA. These findings uncover a mechanism of regulating oligodendrocyte lineage cell density that involves trans-activation of PDGFRα on OPCs via Nrp1 expressed by adjacent microglia.


Asunto(s)
Enfermedades Desmielinizantes/patología , Microglía/fisiología , Neuropilina-1/metabolismo , Células Precursoras de Oligodendrocitos/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Remielinización , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Cuerpo Calloso/citología , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/crecimiento & desarrollo , Cuerpo Calloso/patología , Enfermedades Desmielinizantes/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Humanos , Lisofosfatidilcolinas/administración & dosificación , Lisofosfatidilcolinas/toxicidad , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/ultraestructura , Microscopía Electrónica de Transmisión , Modelos Animales , Vaina de Mielina/metabolismo , Neuropilina-1/genética , Oligodendroglía/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Cultivo Primario de Células
14.
J Neurosci Methods ; 352: 109088, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33508411

RESUMEN

BACKGROUND: Multiple Sclerosis (MS) is a demyelinating disease leading to long-term neurological deficit due to unsuccessful remyelination and axonal loss. Currently, there are no satisfactory treatments for progressive MS somewhat due to the lack of an adequate animal model for studying the mechanisms of disease progression and screening new drugs. NEW METHOD: Lysolecithin (LPC) or agarose-gel loaded LPC (AL-LPC) were applied to mouse optic nerve behind the globe via a minor surgery. Agarose loading was used to achieve longer time of LPC exposure and subsequently long-lasting demyelination. RESULTS: The lesion sites characterized by luxol fast blue (LFB), FluoroMyelin, Bielschowsky's staining, and immunostaining showed extensive demyelination and axonal damage. The loss of Retinal ganglion cells (RGCs) in the corresponding retinal layer was shown by immunostaining and H&E staining. Visual evoked potential (VEP) recordings showed a significant increase in the latency of the P1 wave and a decrease in the amplitude of the P1N1 wave. COMPARISON WITH EXISTING METHODS: The new approach with a very minor surgery seems to be more feasible and reproducible compared to stereotaxic LPC injection to optic chiasm. Our data revealed prolonged demyelination, axonal degeneration and RGCs loss in both AL-LPC and LPC groups; however, these pathologies were more extensive in the AL-LPC group. CONCLUSION: The optimized model provides a longer demyelination time frame and axonal damage followed by RGC degeneration; which is of exceptional interest in investigating axonal degeneration mechanisms and screening the new drugs for progressive MS.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Animales , Enfermedades Desmielinizantes/inducido químicamente , Modelos Animales de Enfermedad , Potenciales Evocados Visuales , Lisofosfatidilcolinas/toxicidad , Ratones , Esclerosis Múltiple/inducido químicamente , Vaina de Mielina , Nervio Óptico
15.
J Neuroinflammation ; 17(1): 333, 2020 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-33158440

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is an immune-mediated demyelinated disease of the central nervous system. Activation of microglia is involved in the pathogenesis of myelin loss. OBJECTIVE: This study is focused on the role of Hv1 in regulating demyelination and microglial activation through reactive oxygen species (ROS) production after lysophosphatidylcholine (LPC)-mediated demyelination. We also explored autophagy in this process. METHODS: A model of demyelination using two-point LPC injection into the corpus callosum was established. LFB staining, immunofluorescence, Western blot, and electron microscopy were used to study the severity of demyelination. Microglial phenotype and autophagy were detected by immunofluorescence and Western blot. Morris water maze was used to test spatial learning and memory ability. RESULTS: We have identified that LPC-mediated myelin damage was reduced by Hv1 deficiency. Furthermore, we found that ROS and autophagy of microglia increased in the demyelination region, which was also inhibited by Hv1 knockout. CONCLUSION: These results suggested that microglial Hv1 deficiency ameliorates demyelination through inhibition of ROS-mediated autophagy and microglial phenotypic transformation.


Asunto(s)
Autofagia/fisiología , Enfermedades Desmielinizantes/metabolismo , Canales Iónicos/deficiencia , Lisofosfatidilcolinas/toxicidad , Microglía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Autofagia/efectos de los fármacos , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/patología
16.
Neurochem Int ; 140: 104827, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32853748

RESUMEN

In this study, we investigated whether melatonin treatment prevents development of neuropathic pain via suppression of glial mitogen-activated protein kinases (MAPKs) activation in the cuneate nucleus (CN) in a lysophosphatidylcholine (LPC)-induced median nerve demyelination neuropathy model. Rats were fed orally with melatonin once a day at a dose of 37.5, 75, or 150 mg/kg 30 min before until 3 days after LPC treatment. Subsequently, behavioral tests were conducted on these animals, and immunohistochemistry and immunoblotting were used for qualitative and quantitative analysis of glia and MAPKs, including ERK, JNK, and p38, activation. Enzyme-linked immunosorbent assays were applied to measure pro-inflammatory cytokine responses. Furthermore, intra-CN microinjection of S26131 (MT1 receptor antagonist), 4P-PDOT (MT2 receptor antagonist), or prazosin (MT3 receptor antagonist) were performed to investigate the association between melatonin receptor subtypes and effects of melatonin on demyelination neuropathy. LPC treatment of the median nerve induced a significant increase in glial fibrillary acidic protein (GFAP; an astrocyte marker) and ED1 (an activated microglia marker) immunoreactivity in the ipsilateral CN and led to development of neuropathic pain behavior. Inspection of GFAP-immunoreactive astrocytes revealed that astrocytic hypertrophy, but not proliferation, contributed to increased GFAP immunoreactivity. Double immunofluorescence showed that both GFAP-immunoreactive astrocytes and ED1-immunoreactive microglia co-expressed p-ERK, p-JNK, and p-p38 immunoreactivity. Melatonin administration dose-dependently reduced neuropathic pain behavior, decreased glial and MAPKs activation, and diminished the release of pro-inflammatory cytokines in the ipsilateral CN after LPC treatment. Furthermore, 4P-PDOT, but not S26131 or prazosin, antagonized the therapeutic effects of melatonin. In conclusion, administration of melatonin, via its cognate MT2 receptor, inhibited activation of glial MAPKs, production of pro-inflammatory cytokines, and development of demyelination-induced neuropathic pain behavior.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Lisofosfatidilcolinas/toxicidad , Melatonina/administración & dosificación , Neuralgia/metabolismo , Neuroglía/metabolismo , Receptor de Melatonina MT2/metabolismo , Animales , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Masculino , Microinyecciones/métodos , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Neuroglía/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor de Melatonina MT2/antagonistas & inhibidores , Tetrahidronaftalenos/administración & dosificación
17.
Cell Stress Chaperones ; 25(6): 955-968, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32572784

RESUMEN

Transient receptor potential ankyrin 1 (TRPA1), the non-selective cation channel, was found that can mediate the generation of multiple sclerosis, while the mechanism is still controversial. Lysophosphatidylcholine (LPC) is a critical trigger of multiple sclerosis which results from the syndrome of neuronal inflammation and demyelination. In this work, we suggested that TRPA1 can mediate the LPC-induced oxidative stress and cytotoxicity in OLN-93 oligodendrocyte. The expression of TRPA1 in OLN-93 was detected by using quantitative real-time PCR (qRT-PCR) and immunofluorescence. The calcium overload induced by LPC via TRPA1 was detected by calcium imaging. The mechanism of LPC-induced mitochondrial reactive oxygen species (mtROS) generation, mitochondria membrane depolarization, nitric oxide (NO) increase, and development of superoxide production via TRPA1 was verified by using confocal imaging. The cell injury elicited by LPC via TRPA1 was confirmed by both CCK-8 and LDH cytotoxicity detection. These results indicate that TRPA1 plays an important role of the LPC-induced oxidative stress and cell damage in OLN-93 oligodendrocyte. Therefore, inhibition of TRPA1 may protect the LPC-induced demyelination.


Asunto(s)
Lisofosfatidilcolinas/toxicidad , Oligodendroglía/metabolismo , Estrés Oxidativo , Canal Catiónico TRPA1/metabolismo , Animales , Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Oligodendroglía/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Superóxidos/metabolismo
18.
Pharmacol Res ; 159: 104997, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32534098

RESUMEN

Myelin loss is the hallmark of the demyelinating disease multiple sclerosis (MS) and plays a significant role in multiple neurodegenerative diseases. A common factor in all neuropathologies is the central role of microglia, the intrinsic immune cells of the central nervous system (CNS). Microglia are activated in pathology and can have both pro- and anti-inflammatory functions. Here, we examined the effects of the flavonoid agathisflavone on microglia and remyelination in the cerebellar slice model following lysolecithin induced demyelination. Notably, agathisflavone enhances remyelination and alters microglial activation state, as determined by their morphology and cytokine profile. Furthermore, these effects of agathisflavone on remyelination and microglial activation were inhibited by blockade of estrogen receptor α. Thus, our results identify agathisflavone as a novel compound that may act via ER to regulate microglial activation and enhance remyelination and repair.


Asunto(s)
Antiinflamatorios/farmacología , Biflavonoides/farmacología , Cerebelo/efectos de los fármacos , Microglía/efectos de los fármacos , Vaina de Mielina/metabolismo , Neuroinmunomodulación/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Remielinización/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cerebelo/inmunología , Cerebelo/metabolismo , Cerebelo/patología , Citocinas/metabolismo , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Lisofosfatidilcolinas/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Oligodendroglía/inmunología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Fenotipo , Técnicas de Cultivo de Tejidos
19.
Cardiovasc Eng Technol ; 11(3): 316-327, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32356274

RESUMEN

PURPOSE: Fibrocalcific aortic valve disease (CAVD) is caused by the deposition of calcific nodules in the aortic valve leaflets, resulting in progressive loss of function that ultimately requires surgical intervention. This process is actively mediated by the resident valvular interstitial cells (VICs), which, in response to oxidized lipids, transition from a quiescent to an osteoblast-like state. The purpose of this study was to examine if the ryanodine receptor, an intracellular calcium channel, could be therapeutically targeted to prevent this phenotypic conversion. METHODS: The expression of the ryanodine receptor in porcine aortic VICs was characterized by qRT-PCR and immunofluorescence. Next, the VICs were exposed to lysophosphatidylcholine, an oxidized lipid commonly found in low-density lipoprotein, while the activity of the ryanodine receptor was modulated with ryanodine. The cultures were analyzed for markers of cellular mineralization, alkaline phosphatase activity, proliferation, and apoptosis. RESULTS: Porcine aortic VICs predominantly express isoform 3 of the ryanodine receptors, and this protein mediates the cellular response to LPC. Exposure to LPC caused elevated intracellular calcium concentration in VICs, raised levels of alkaline phosphatase activity, and increased calcific nodule formation, but these changes were reversed when the activity of the ryanodine receptor was blocked. CONCLUSIONS: Our findings suggest blocking the activity of the ryanodine receptor can attenuate the valvular mineralization caused by LPC. We conclude that oxidized lipids, such as LPC, play an important role in the development and progression of CAVD and that the ryanodine receptor is a promising target for pharmacological intervention.


Asunto(s)
Válvula Aórtica/efectos de los fármacos , Calcinosis/inducido químicamente , Agonistas de los Canales de Calcio/toxicidad , Calcio/metabolismo , Lisofosfatidilcolinas/toxicidad , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Apoptosis/efectos de los fármacos , Calcinosis/metabolismo , Calcinosis/patología , Calcinosis/prevención & control , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Canal Liberador de Calcio Receptor de Rianodina/genética , Sus scrofa
20.
J Neurosci ; 40(7): 1483-1500, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31911460

RESUMEN

Myelin loss limits neurological recovery and myelin regeneration and is critical for restoration of function. We recently discovered that global knock-out of the thrombin receptor, also known as Protease Activated Receptor 1 (PAR1), accelerates myelin development. Here we demonstrate that knocking out PAR1 also promotes myelin regeneration. Outcomes in two unique models of myelin injury and repair, that is lysolecithin or cuprizone-mediated demyelination, showed that PAR1 knock-out in male mice improves replenishment of myelinating cells and remyelinated nerve fibers and slows early axon damage. Improvements in myelin regeneration in PAR1 knock-out mice occurred in tandem with a skewing of reactive astrocyte signatures toward a prorepair phenotype. In cell culture, the promyelinating effects of PAR1 loss of function are consistent with possible direct effects on the myelinating potential of oligodendrocyte progenitor cells (OPCs), in addition to OPC-indirect effects involving enhanced astrocyte expression of promyelinating factors, such as BDNF. These findings highlight previously unrecognized roles of PAR1 in myelin regeneration, including integrated actions across the oligodendrocyte and astroglial compartments that are at least partially mechanistically linked to the powerful BDNF-TrkB neurotrophic signaling system. Altogether, findings suggest PAR1 may be a therapeutically tractable target for demyelinating disorders of the CNS.SIGNIFICANCE STATEMENT Replacement of oligodendroglia and myelin regeneration holds tremendous potential to improve function across neurological conditions. Here we demonstrate Protease Activated Receptor 1 (PAR1) is an important regulator of the capacity for myelin regeneration across two experimental murine models of myelin injury. PAR1 is a G-protein-coupled receptor densely expressed in the CNS, however there is limited information regarding its physiological roles in health and disease. Using a combination of PAR1 knock-out mice, oligodendrocyte monocultures and oligodendrocyte-astrocyte cocultures, we demonstrate blocking PAR1 improves myelin production by a mechanism related to effects across glial compartments and linked in part to regulatory actions toward growth factors such as BDNF. These findings set the stage for development of new clinically relevant myelin regeneration strategies.


Asunto(s)
Enfermedades Desmielinizantes/fisiopatología , Regeneración Nerviosa/efectos de los fármacos , Receptor PAR-1/antagonistas & inhibidores , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Axones/patología , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/farmacología , Quelantes/toxicidad , Técnicas de Cocultivo , Cobre , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/patología , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Perfilación de la Expresión Génica , Lisofosfatidilcolinas/toxicidad , Masculino , Ratones , Ratones Noqueados , Vaina de Mielina/fisiología , Regeneración Nerviosa/fisiología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Receptor PAR-1/deficiencia , Receptor PAR-1/fisiología , Prueba de Desempeño de Rotación con Aceleración Constante , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...