Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38746334

RESUMEN

White matter injury (WMI) is thought to be a major contributor to long-term cognitive dysfunctions after traumatic brain injury (TBI). This damage occurs partly due to apoptotic death of oligodendrocyte lineage cells (OLCs) after the injury, triggered directly by the trauma or in response to degenerating axons. Recent research suggests that the gut microbiota modulates the inflammatory response through the modulation of peripheral immune cell infiltration after TBI. Additionally, T-cells directly impact OLCs differentiation and proliferation. Therefore, we hypothesized that the gut microbiota plays a critical role in regulating the OLC response to WMI influencing T-cells differentiation and activation. Gut microbial depletion early after TBI chronically reduced re-myelination, acutely decreased OLCs proliferation, and was associated with increased myelin debris accumulation. Surprisingly, the absence of T-cells in gut microbiota depleted mice restored OLC proliferation and remyelination after TBI. OLCs co-cultured with T-cells derived from gut microbiota depleted mice resulted in impaired proliferation and increased expression of MHC-II compared with T cells from control-injured mice. Furthermore, MHC-II expression in OLCs appears to be linked to impaired proliferation under gut microbiota depletion and TBI conditions. Collectively our data indicates that depletion of the gut microbiota after TBI impaired remyelination, reduced OLCs proliferation with concomitantly increased OLC MHCII expression and required the presence of T cells. This data suggests that T cells are an important mechanistic link by which the gut microbiota modulate the oligodendrocyte response and white matter recovery after TBI.

2.
Neurotherapeutics ; 20(6): 1859-1874, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37768487

RESUMEN

Traumatic brain injury (TBI) can result in axonal loss and demyelination, leading to persistent damage in the white matter. Demyelinated axons are vulnerable to pathologies related to an abnormal myelin structure that expose neurons to further damage. Oligodendrocyte progenitor cells (OPCs) mediate remyelination after recruitment to the injury site. Often this process is inefficient due to inadequate OPC proliferation. To date, no effective treatments are currently available to stimulate OPC proliferation in TBI. Recombinant human erythropoietin (rhEPO) is a pleiotropic neuroprotective cytokine, and its receptor is present in all stages of oligodendroglial lineage cell differentiation. Therefore, we hypothesized that rhEPO administration would enhance remyelination after TBI through the modulation of OPC response. Utilizing a murine model of controlled cortical impact and a primary OPC culture in vitro model, we characterized the impact of rhEPO on remyelination and proliferation of oligodendrocyte lineage cells. Myelin black gold II staining of the peri-contusional corpus callosum revealed an increase in myelinated area in association with an increase in BrdU-positive oligodendrocytes in injured mice treated with rhEPO. Furthermore, morphological analysis of OPCs showed a decrease in process length in rhEPO-treated animals. RhEPO treatment increased OPC proliferation after in vitro CSPG exposure. Erythropoietin receptor (EPOr) gene knockdown using siRNA prevented rhEPO-induced OPC proliferation, demonstrating that the rhEPO effect on OPC response is EPOr activation dependent. Together, our findings demonstrate that rhEPO administration may promote myelination by increasing oligodendrocyte lineage cell proliferation after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Eritropoyetina , Células Precursoras de Oligodendrocitos , Ratones , Humanos , Animales , Células Precursoras de Oligodendrocitos/patología , Oligodendroglía , Vaina de Mielina , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Proteínas Recombinantes/farmacología , Proliferación Celular , Hipoxia/patología , Eritropoyetina/farmacología , Diferenciación Celular
3.
Neurosci Lett ; 804: 137216, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36997018

RESUMEN

Erythropoietin (EPO) is a hypoxia-responsive cytokine that induces neuroprotective effect in hypoxic-ischaemic, traumatic, excitotoxic and inflammatory injuries. Recently, utilizing a clinically relevant murine model of TBI and delayed hypoxemia, we have found that ongoing recombinant human EPO (rhEPO) administration influenced neurogenesis, neuroprotection, synaptic density and, behavioral outcomes early after TBI, and the impact on long-lasting outcomes 6 months after injury. We also demonstrated that the 1-month behavioral improvement was associated with mitogen-activated protein kinase (MAPK)/cAMP response element-binding protein (CREB) signaling activation and increased of excitatory synaptic density in the amygdala. However, we did not uncover which type of cells were involved in fear memory response enhancement after rhEPO treatment in the setting of TBI with delayed hypoxemia. In this report, using chemogenetic tools in our controlled cortical impact (CCI) model, we were able to inactivate excitatory neurons and eliminate rhEPO-induced fear memory recall enhancement. In summary, these data demonstrate that rhEPO treatment initiated after TBI enhances contextual fear memory in the injured brain via activation of excitatory neurons in the amygdala.


Asunto(s)
Lesiones Encefálicas , Eritropoyetina , Ratones , Humanos , Animales , Lesiones Encefálicas/metabolismo , Eritropoyetina/farmacología , Neuronas/metabolismo , Hipoxia , Miedo
4.
J Neurotrauma ; 40(7-8): 772-787, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36259455

RESUMEN

Traumatic brain injury (TBI) patients are at high risk for disruption of the gut microbiome. Previously, we have demonstrated that broad-spectrum antibiotic exposure after TBI drastically alters the gut microbiota and modulates neuroinflammation, neurogenesis, and long-term fear memory. However, these data did not determine if the impact of antibiotic exposure on the brain's response to injury was mediated directly by antibiotics or indirectly via modulation of the gut microbiota. We designed two different approaches to address this knowledge gap. One was utilizing fecal microbiota transplantation (FMT) from control and antibiotic-treated mice (treated with vancomycin, neomycin, ampicillin, and metronidazole [VNAM]) into germ-free (GF) mice prior to injury, and the other was exposing specific pathogen-free (SPF) mice to a 2-week period of antibiotics prior to injury but discontinuing antibiotics 72 h prior to injury. GF mice receiving FMT from VNAM-treated mice (GF-VNAM) demonstrated reduced gut bacterial alpha diversity and richness compared with GF mice receiving control FMT. At 7 days post-injury, GF-VNAM had increased microglial activation, reduced infiltration of T cells, and decreased neurogenesis. Similarly, SPF mice exposed to antibiotics prior to but not after injury demonstrated similar alterations in neuroinflammation and neurogenesis compared with control mice. These data support our hypothesis implicating the gut microbiota as an important modulator of the neuroinflammatory process and neurogenesis after TBI and provide an exciting new approach for neuroprotective therapeutics for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Microbioma Gastrointestinal , Ratones , Animales , Microbioma Gastrointestinal/fisiología , Enfermedades Neuroinflamatorias , Antibacterianos/farmacología , Metronidazol , Lesiones Traumáticas del Encéfalo/terapia , Ampicilina , Ratones Endogámicos C57BL
5.
Brain Res ; 1795: 148074, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36075467

RESUMEN

Therapeutic interventions targeting secondary insults, such as delayed hypoxemia, provide a unique opportunity for treatment in severe traumatic brain injury (TBI). Erythropoietin (EPO) is a hypoxia-responsive cytokine with important roles in neurodevelopment, neuroprotection and neuromodulation. We hypothesized that recombinant human erythropoietin (rhEPO) administration would mitigate injury in a combined injury model of TBI and delayed hypoxemia. Utilizing a clinically relevant murine model of TBI and delayed hypoxemia, we characterized how ongoing rhEPO administration influenced neurogenesis, neuroprotection, synaptic density and, behavioral outcomes early after TBI, and the impact on long-lasting outcomes 6 months after injury. We employed novel object recognition (NOR) and fear conditioning to assess long-term memory. At 1-month post-injury, we observed a significant increase in cued-fear memory response in the rhEPO-injured mice compared with vehicle-injured mice. This was associated with neuroprotection and neurogenesis in the hippocampus and mitogen-activated protein kinase (MAPK)/cAMP response element-binding protein (CREB) signaling activation and increased of excitatory synaptic density in the amygdala. Early rhEPO treatment after injury reduced neurodegeneration and increased excitatory synaptic density in the hippocampus and amygdala at 6 months post-injury. However at 6 months post-injury (4 months after discontinuation of rhEPO), we did not observe changes in behavioral assessments nor MAPK/CREB pathway activation. In summary, these data demonstrate that ongoing rhEPO treatment initiated at a clinically feasible time point improves neurological, cognitive, and histological outcomes after TBI in the setting of secondary hypoxemic insults.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Eritropoyetina , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Miedo , Humanos , Hipoxia/complicaciones , Hipoxia/tratamiento farmacológico , Ratones , Proteínas Quinasas Activadas por Mitógenos , Neuroprotección , Proteínas Recombinantes
6.
Acta Neuropathol Commun ; 10(1): 10, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35090569

RESUMEN

Clinical trials of therapeutics for traumatic brain injury (TBI) demonstrating preclinical efficacy for TBI have failed to replicate these results in humans, in part due to the absence of clinically feasible therapeutic windows for administration. Minocycline, an inhibitor of microglial activation, has been shown to be neuroprotective when administered early after experimental TBI but detrimental when administered chronically to human TBI survivors. Rather than focusing on the rescue of primary injury with early administration of therapeutics which may not be clinically feasible, we hypothesized that minocycline administered at a clinically feasible time point (24 h after injury) would be neuroprotective in a model of TBI plus delayed hypoxemia. We first explored several different regimens of minocycline dosing with the initial dose 24 h after injury and 2 h prior to hypoxemia, utilizing short-term neuropathology to select the most promising candidate. We found that a short course of minocycline reduced acute microglial activation, monocyte infiltration and hippocampal neuronal loss at 1 week post injury. We then conducted a preclinical trial to assess the long-term efficacy of a short course of minocycline finding reductions in hippocampal neurodegeneration and synapse loss, preservation of white matter myelination, and improvements in fear memory performance at 6 months after injury. Timing in relation to injury and duration of minocycline treatment and its impact on neuroinflammatory response may be responsible for extensive neuroprotection observed in our studies.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Minociclina/farmacología , Fármacos Neuroprotectores/farmacología , Recuperación de la Función/efectos de los fármacos , Animales , Femenino , Masculino , Memoria/efectos de los fármacos , Ratones , Minociclina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico
7.
Neural Regen Res ; 17(5): 1007-1008, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-34558522
8.
Acta Neuropathol Commun ; 9(1): 40, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33691793

RESUMEN

The influence of the gut microbiota on traumatic brain injury (TBI) is presently unknown. This knowledge gap is of paramount clinical significance as TBI patients are highly susceptible to alterations in the gut microbiota by antibiotic exposure. Antibiotic-induced gut microbial dysbiosis established prior to TBI significantly worsened neuronal loss and reduced microglia activation in the injured hippocampus with concomitant changes in fear memory response. Importantly, antibiotic exposure for 1 week after TBI reduced cortical infiltration of Ly6Chigh monocytes, increased microglial pro-inflammatory markers, and decreased T lymphocyte infiltration, which persisted through 1 month post-injury. Moreover, microbial dysbiosis was associated with reduced neurogenesis in the dentate gyrus 1 week after TBI. By 3 months after injury (11 weeks after discontinuation of the antibiotics), we observed increased microglial proliferation, increased hippocampal neuronal loss, and modulation of fear memory response. These data demonstrate that antibiotic-induced gut microbial dysbiosis after TBI impacts neuroinflammation, neurogenesis, and fear memory and implicate gut microbial modulation as a potential therapeutic intervention for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Disbiosis/complicaciones , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad , Neurogénesis , Animales , Bacterias/genética , Modelos Animales de Enfermedad , Disbiosis/microbiología , Disbiosis/fisiopatología , Hipocampo/patología , Masculino , Memoria , Ratones , Ratones Endogámicos C57BL , Microglía
9.
Exp Neurol ; 335: 113507, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33065076

RESUMEN

Preclinical investigations into neuroprotective agents for traumatic brain injury (TBI) have shown promise when administered before or very early after experimental TBI. However clinical trials of therapeutics demonstrating preclinical efficacy for TBI have failed to replicate these results in humans, a lost in translation phenomenon. N-acetylcysteine (NAC) is a potent anti-oxidant with demonstrated efficacy in pre-clinical TBI when administered early after primary injury. Utilizing our clinically relevant mouse model, we hypothesized that NAC administration in a clinically relevant timeframe could improve the brain's resilience to the secondary insult of hypoxemia. NAC or vehicle administered daily starting 2 h prior to hypoxemia (24 h after controlled cortical impact) for 3 doses in male mice reduced short-term axonal injury and hippocampal neuronal loss. Six month behavioral assessments including novel object recognition, socialization, Barnes maze, and fear conditioning did not reveal performance differences between sham controls and injured mice receiving NAC or saline vehicle. At 7 months after injury, NAC administered mice had reduced hippocampal neuronal loss but no reduction in lesion volume. In summary, our preclinical trial to test the neuroprotective efficacy of NAC against a secondary hypoxic insult after TBI demonstrated short and long-term neuropathological evidence of neuroprotection but a lack of detectable differences in long-term behavioral assessments between sham controls and injured mice limits conclusions on its impact on long-term neurobehavioral outcomes.


Asunto(s)
Acetilcisteína/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Axones/patología , Conducta Animal/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/psicología , Miedo , Glutatión/metabolismo , Hipocampo/patología , Hipoxia/psicología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , Desempeño Psicomotor , Conducta Social
10.
Neuropharmacology ; 125: 319-332, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28807673

RESUMEN

The GPR55 receptor is expressed abundantly in the brain, especially in the striatum, suggesting it might fulfill a role in motor function. Indeed, motor behavior is impaired in mice lacking GPR55, which also display dampened inflammatory responses. Abnormal-cannabidiol (Abn-CBD), a synthetic cannabidiol (CBD) isomer, is a GPR55 agonist that may serve as a therapeutic agent in the treatment of inflammatory diseases. In this study, we explored whether modulating GPR55 could also represent a therapeutic approach for the treatment of Parkinson's disease (PD). The distribution of GPR55 mRNA was first analyzed by in situ hybridization, localizing GPR55 transcripts to neurons in brain nuclei related to movement control, striatum, globus pallidus, subthalamic nucleus, substantia nigra and cortex. Striatal expression of GPR55 was downregulated in parkinsonian conditions. When Abn-CBD and CBD (5 mg/kg) were chronically administered to mice treated over 5 weeks with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and probenecid (MPTPp), Abn-CBD but not CBD prevented MPTPp induced motor impairment. Although Abn-CBD protected dopaminergic cell bodies, it failed to prevent degeneration of the terminals or preserve dopamine levels in the striatum. Both compounds induced morphological changes in microglia that were compatible with an anti-inflammatory phenotype that did not correlate with a neuroprotective activity. The symptomatic relief of Abn-CBD was further studied in the haloperidol-induced catalepsy mouse model. Abn-CBD had an anti-cataleptic effect that was reversed by CBD and PSB1216, a newly synthesized GPR55 antagonist, and indeed, two other GPR55 agonists also displayed anti-cataleptic effects (CID1792197 and CID2440433). These results demonstrate for the first time that activation of GPR55 might be beneficial in combating PD.


Asunto(s)
Antiparkinsonianos/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/metabolismo , Receptores de Cannabinoides/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Ganglios Basales/efectos de los fármacos , Ganglios Basales/metabolismo , Ganglios Basales/patología , Proteínas de Unión al Calcio/metabolismo , Cannabidiol/análogos & derivados , Cannabidiol/farmacología , Catalepsia/tratamiento farmacológico , Catalepsia/metabolismo , Catalepsia/patología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Dopamina/metabolismo , Haloperidol , Ácido Homovanílico/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Trastornos Parkinsonianos/patología , ARN Mensajero/metabolismo
11.
Neuropharmacology ; 113(Pt A): 110-123, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27693549

RESUMEN

Neuroinflammation involves the activation of glial cells, which is associated to the progression of neurodegeneration in Parkinson's disease. Recently, we and other researchers demonstrated that dopamine receptor D3 (D3R)-deficient mice are completely refractory to neuroinflammation and consequent neurodegeneration associated to the acute intoxication with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). In this study we examined the therapeutic potential and underlying mechanism of a D3R-selective antagonist, PG01037, in mice intoxicated with a chronic regime of administration of MPTP and probenecid (MPTPp). Biodistribution analysis indicated that intraperitoneally administered PG01037 crosses the blood-brain barrier and reaches the highest concentration in the brain 40 min after the injection. Furthermore, the drug was preferentially distributed to the brain in comparison to the plasma. Treatment of MPTPp-intoxicated mice with PG01037 (30 mg/kg, administrated twice a week for five weeks) attenuated the loss of dopaminergic neurons in the substantia nigra pars compacta, as evaluated by stereological analysis, and the loss of striatal dopaminergic terminals, as determined by densitometric analyses of tyrosine hydroxylase and dopamine transporter immunoreactivities. Accordingly, the treatment resulted in significant improvement of motor performance of injured animals. Interestingly, the therapeutic dose of PG01037 exacerbated astrogliosis and resulted in increased ramification density of microglial cells in the striatum of MPTPp-intoxicated mice. Further analyses suggested that D3R expressed in astrocytes favours a beneficial astrogliosis with anti-inflammatory consequences on microglia. Our findings indicate that D3R-antagonism exerts a therapeutic effect in parkinsonian animals by reducing the loss of dopaminergic neurons in the nigrostriatal pathway, alleviating motor impairments and modifying the pro-inflammatory phenotype of glial cells.


Asunto(s)
Benzamidas/administración & dosificación , Neuronas Dopaminérgicas/efectos de los fármacos , Encefalitis/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Trastornos Parkinsonianos/prevención & control , Piridinas/administración & dosificación , Receptores de Dopamina D3/antagonistas & inhibidores , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Benzamidas/sangre , Benzamidas/farmacología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Neuronas Dopaminérgicas/patología , Encefalitis/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/prevención & control , Trastornos Parkinsonianos/complicaciones , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Porción Compacta de la Sustancia Negra/patología , Piridinas/sangre , Piridinas/farmacología , Receptores de Dopamina D3/metabolismo
12.
Neurosci Biobehav Rev ; 76(Pt B): 380-395, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27639448

RESUMEN

Public opinion surrounding the recreational use and therapeutic potential of cannabis is shifting. This review describes new work examining the behavioural and neural effects of cannabis and the endocannabinoid system, highlighting key regions within corticolimbic brain circuits. First, we consider the role of human genetic factors and cannabis strain chemotypic differences in contributing to interindividual variation in the response to cannabinoids, such as THC, and review studies demonstrating that THC-induced impairments in decision-making processes are mediated by actions at prefrontal CB1 receptors. We further describe evidence that signalling through prefrontal or ventral hippocampal CB1 receptors modulates mesolimbic dopamine activity, aberrations of which may contribute to emotional processing deficits in schizophrenia. Lastly, we review studies suggesting that endocannabinoid tone in the amygdala is a critical regulator of anxiety, and report new data showing that FAAH activity is integral to this response. Together, these findings underscore the importance of cannabinoid signalling in the regulation of cognitive and affective behaviours, and encourage further research given their social, political, and therapeutic implications.


Asunto(s)
Fumar , Animales , Cannabis , Endocannabinoides , Humanos , Receptor Cannabinoide CB1
13.
Brain Behav Immun ; 57: 94-105, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27318096

RESUMEN

Elements of the endocannabinoid system are strongly expressed in the basal ganglia where they suffer profound rearrangements after dopamine depletion. Modulation of the levels of the endocannabinoid 2-arachidonoyl-glycerol by inhibiting monoacylglycerol lipase alters glial phenotypes and provides neuroprotection in a mouse model of Parkinson's disease. In this study, we assessed whether inhibiting fatty acid amide hydrolase could also provide beneficial effects on the time course of this disease. The fatty acid amide hydrolase inhibitor, URB597, was administered chronically to mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and probenecid (MPTPp) over 5weeks. URB597 (1mg/kg) prevented MPTPp induced motor impairment but it did not preserve the dopamine levels in the nigrostriatal pathway or regulate glial cell activation. The symptomatic relief of URB597 was confirmed in haloperidol-induced catalepsy assays, where its anti-cataleptic effects were both blocked by antagonists of the two cannabinoid receptors (CB1 and CB2), and abolished in animals deficient in these receptors. Other fatty acid amide hydrolase inhibitors, JNJ1661010 and TCF2, also had anti-cataleptic properties. Together, these results demonstrate an effect of fatty acid amide hydrolase inhibition on the motor symptoms of Parkinson's disease in two distinct experimental models that is mediated by cannabinoid receptors.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Ácidos Araquidónicos/metabolismo , Benzamidas/farmacología , Agonistas de Receptores de Cannabinoides/metabolismo , Carbamatos/farmacología , Endocannabinoides/metabolismo , Trastornos Parkinsonianos/tratamiento farmacológico , Alcamidas Poliinsaturadas/metabolismo , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB2/antagonistas & inhibidores , Animales , Benzamidas/administración & dosificación , Carbamatos/administración & dosificación , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL
14.
J Plast Reconstr Aesthet Surg ; 69(3): 417-26, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26683008

RESUMEN

Several techniques have been described for smile restoration after facial nerve paralysis. When a nerve other than the contralateral facial nerve is used to restore the smile, some controversy appears because of the nonphysiological mechanism of smile recovering. Different authors have reported natural results with the masseter nerve. The physiological pathways which determine whether this is achieved continue to remain unclear. Using functional magnetic resonance imaging, brain activation pattern measuring blood-oxygen-level-dependent (BOLD) signal during smiling and jaw clenching was recorded in a group of 24 healthy subjects (11 females). Effective connectivity of premotor regions was also compared in both tasks. The brain activation pattern was similar for smile and jaw-clenching tasks. Smile activations showed topographic overlap though more extended for smile than clenching. Gender comparisons during facial movements, according to kinematics and BOLD signal, did not reveal significant differences. Effective connectivity results of psychophysiological interaction (PPI) from the same seeds located in bilateral facial premotor regions showed significant task and gender differences (p < 0.001). The hypothesis of brain plasticity between the facial nerve and masseter nerve areas is supported by the broad cortical overlap in the representation of facial and masseter muscles.


Asunto(s)
Encéfalo/patología , Nervio Facial/anatomía & histología , Parálisis Facial/cirugía , Músculo Masetero/inervación , Transferencia de Nervios/métodos , Plasticidad Neuronal/fisiología , Adulto , Estudios de Casos y Controles , Estimulación Eléctrica , Expresión Facial , Parálisis Facial/diagnóstico , Parálisis Facial/rehabilitación , Femenino , Voluntarios Sanos , Humanos , Maxilares/fisiología , Imagen por Resonancia Magnética/métodos , Persona de Mediana Edad , Oxígeno/sangre , Valores de Referencia , Sonrisa/fisiología , Adulto Joven
15.
Mol Neurobiol ; 53(4): 2312-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-25976369

RESUMEN

Growing evidence suggests that the endocannabinoid system plays a role in neuroprotection in Parkinson's disease. Recently, we have shown the neuroprotective effect of monoacylglycerol lipase (MAGL) inhibition with JZL184 in the chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model. However, further investigation is needed to determine the neuroprotective mechanisms of the endocannabinoid system on the nigrostriatal pathway. The aim of this work was to investigate whether the neuroprotective effect of JZL184 in mice could be extended to an in vitro cellular model to further understand the mechanism of action of the drug. The SH-SY5Y cell line was selected based on its dopaminergic-like phenotype and its susceptibility to 1-methyl-4-phenylpyridinium iodide (MPP(+)) toxicity. Furthermore, SH-SY5Y cells express both cannabinoid receptors, CB1 and CB2. The present study describes the neuroprotective effect of MAGL inhibition with JZL184 in SH-SY5Y cells treated with MPP(+). The effect of JZL184 in cell survival was blocked by AM630, a CB2 receptor antagonist, and it was mimicked with JWH133, a CB2 receptor agonist. Rimonabant, a CB1 receptor antagonist, did not affect JZL184-induced cell survival. These results demonstrate that the neuroprotective effect of MAGL inhibition with JZL184 described in animal models of Parkinson's disease could be extended to in vitro models such as SH-SY5Y cells treated with MPP(+). This represents a useful tool to study mechanisms of neuroprotection mediated by MAGL inhibition, and we provide evidence for the possible involvement of CB2 receptors in the improvement of cell survival.


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Benzodioxoles/farmacología , Fármacos Neuroprotectores/farmacología , Piperidinas/farmacología , Receptor Cannabinoide CB2/metabolismo , Amidohidrolasas/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Humanos , Monoacilglicerol Lipasas/antagonistas & inhibidores , Monoacilglicerol Lipasas/metabolismo , Neurotoxinas/toxicidad , Receptor Cannabinoide CB1/metabolismo
16.
Pain Rep ; 1(3)2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28210712

RESUMEN

INTRODUCTION: Epidermal keratinocytes are increasingly recognized as active participants in the sensory transduction of itch and pain, processes known to involve primary afferent glutamatergic neurons. However the role of keratinocyte glutamate signaling in sensory functioning is not fully understood. Here, we present the observation of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid type glutamate receptors (AMPAR) in epidermal keratinocytes. METHODS: Immunohistochemical and in situ hybridization analyses were conducted to assess the expression of AMPAR subunits in epidermal keratinocytes in mouse and human skin samples, and in organotypic cultures of human keratinocytes. In addition, RTPCR further confirmed the expression of GluA4-containing AMPAR in epidermal keratinocytes. RESULTS: We found prominent immunolabeling (IL) for the GluA4 subunit of AMPAR in keratinocytes of glabrous and hairy skin of mouse epidermis, as well as in human epidermal keratinocytes. RTPCR confirmed Gria4 transcript expression in epidermal mouse keratinocytes. In addition, expression of GRIA4 mRNA was confirmed in epidermal human keratinocytes by in situ hybridization. Immunohistochemical studies conducted in human skin biopsies from patients with atopic dermatitis (AD) and postherpetic neuralgia (PHN) demonstrate that keratinocyte expression of GluA4 can be altered under pathological conditions. Moreover, a decrease of GluA4 expression was observed in organotypic cultures of human keratinocytes after direct application of algogenic agents. CONCLUSIONS: We provide evidence that GluA4-containing AMPAR are expressed in epidermal keratinocytes, that human pruritic and painful dermatopathologies have alterations in the keratinocyte expression levels of GluA4-containing AMPAR, and that itch and pain producing substances can directly regulate their production in keratinocytes.

17.
Neurobiol Aging ; 35(11): 2603-2616, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24973119

RESUMEN

Changes in cannabinoid receptor expression and concentration of endocannabinoids have been described in Parkinson's disease; however, it remains unclear whether they contribute to, or result from, the disease process. To evaluate whether targeting the endocannabinoid system could provide potential benefits in the treatment of the disease, the effect of a monoacylglycerol lipase inhibitor that prevents degradation of 2-arachidonyl-glycerol was tested in mice treated chronically with probenecid and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTPp). Chronic administration of the compound, JZL184 (8 mg/kg), prevented MPTPp-induced motor impairment and preserved the nigrostriatal pathway. Furthermore, none of the hypokinetic effects associated with cannabinoid receptor agonism were observed. In the striatum and substantia nigra pars compacta, MPTPp animals treated with JZL184 exhibited astroglial and microglial phenotypic changes that were accompanied by increases in TGFß messenger RNA expression and in glial cell-derived neurotrophic factor messenger RNA and protein levels. JZL184 induced an increase in ß-catenin translocation to the nucleus, implicating the Wnt/catenin pathway. Together, these results demonstrate a potent neuroprotective effect of JZL184 on the nigrostriatal pathway of parkinsonian animals, likely involving restorative astroglia and microglia activation and the release of neuroprotective and antiinflammatory molecules.


Asunto(s)
Benzodioxoles/uso terapéutico , Monoacilglicerol Lipasas/antagonistas & inhibidores , Neuroglía/patología , Fármacos Neuroprotectores , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/patología , Piperidinas/uso terapéutico , Animales , Antiinflamatorios/metabolismo , Ácidos Araquidónicos/metabolismo , Benzodioxoles/farmacología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Endocannabinoides/metabolismo , Proteína Ácida Fibrilar de la Glía , Glicéridos/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Vías Nerviosas/patología , Vías Nerviosas/fisiopatología , Fármacos Neuroprotectores/metabolismo , Trastornos Parkinsonianos/fisiopatología , Piperidinas/farmacología , Probenecid , Sustancia Negra/metabolismo , Sustancia Negra/patología , Sustancia Negra/fisiopatología
18.
J Neuropathol Exp Neurol ; 71(11): 973-82, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23044920

RESUMEN

The external segment of the globus pallidus (GPe) in humans and the equivalent structure in rodents, the globus pallidus (GP), influence signal processing in the basal ganglia under normal and pathological conditions. Parvalbumin (PV) immunoreactivity defines 2 main neuronal subpopulations in the GP/GPe: PV-immunopositive cells that project mainly to the subthalamic nucleus and the internal segment of the GP and PV-negative cells that mainly project to the striatum. We evaluated the number of neurons in the GP/GPe in animal models of Parkinson disease. In rats, dopaminergic denervation with 6-hydroxydopamine (6-OHDA) provoked a significant decrease in the number of GP neurons (12% ± 4%, p < 0.05), which specifically affected the PV subpopulation. A similar trend was observed in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys. Markers of GABAergic activity (GAD65 and GAD67 mRNA) were not different from those of controls in 6-OHDA-lesioned rats. Taken together, these findings provide evidence for nondopaminergic neuronal cell loss in the basal ganglia of 6-OHDA-lesioned rats and suggest that a similar loss may occur in the MPTP monkey. These data suggest that in patients with Parkinson disease, the loss of GABAergic neurons projecting to the subthalamic nucleus may contribute to the hyperactivity of this nucleus despite the absence of gross alterations in GAD mRNA expression.


Asunto(s)
Neuronas GABAérgicas/patología , Globo Pálido/patología , Trastornos Parkinsonianos/patología , Parvalbúminas/biosíntesis , Animales , Biomarcadores/metabolismo , Muerte Celular/fisiología , Modelos Animales de Enfermedad , Neuronas GABAérgicas/metabolismo , Globo Pálido/metabolismo , Macaca fascicularis , Masculino , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...