Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Neurobiol Dis ; 186: 106273, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37648036

RESUMEN

Epilepsy is one of the most common neurological disorders. Neuroinflammation involving the activation of microglia and astrocytes constitutes an important and common mechanism in epileptogenesis. Transient receptor potential melastatin 2 (TRPM2) is a calcium-permeable, non-selective cation channel that plays pathological roles in various inflammation-related diseases. Our previous study demonstrated that Trpm2 knockout exhibits therapeutic effects on pilocarpine-induced glial activation and neuroinflammation. However, whether TRPM2 in microglia and astrocytes plays a common pathogenic role in this process and the underlying molecular mechanisms remained undetermined. Here, we demonstrate a previously unknown role for microglial TRPM2 in epileptogenesis. Trpm2 knockout in microglia attenuated kainic acid (KA)-induced glial activation, inflammatory cytokines production and hippocampal paroxysmal discharges, whereas Trpm2 knockout in astrocytes exhibited no significant effects. Furthermore, we discovered that these therapeutic effects were mediated by upregulated autophagy via the adenosine monophosphate activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway in microglia. Thus, our findings highlight an important deleterious role of microglial TRPM2 in temporal lobe epilepsy.


Asunto(s)
Microglía , Canales Catiónicos TRPM , Humanos , Proteínas Quinasas Activadas por AMP , Enfermedades Neuroinflamatorias , Canales Catiónicos TRPM/genética , Serina-Treonina Quinasas TOR , Autofagia , Canales de Calcio
2.
Neurobiol Dis ; 172: 105823, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35878745

RESUMEN

Neuroinflammation contributes to the generation of epilepsy and has been proposed as an effective therapeutic target. Recent studies have uncovered the potential effects of the anti-fungal drug miconazole for treating various brain diseases by suppressing neuroinflammation but have not yet been studied in epilepsy. Here, we investigated the effects of different doses of miconazole (5, 20, 80 mg/kg) on seizure threshold, inflammatory cytokines release, and glial cells activation in the pilocarpine (PILO) pentylenetetrazole (PTZ), and intrahippocampal kainic acid (IHKA) models. We demonstrated that 5 and 20 mg/kg miconazole increased seizure threshold, but only 20 mg/kg miconazole reduced inflammatory cytokines release, glial cells activation, and morphological alteration during the early post-induction period (24 h, 3 days). We further investigated the effects of 20 mg/kg miconazole on epilepsy (4 weeks after KA injection). We found that miconazole significantly attenuated cytokines production, glial cells activation, microglial morphological changes, frequency and duration of recurrent hippocampal paroxysmal discharges (HPDs), and neuronal and synaptic damage in the hippocampus during epilepsy. In addition, miconazole suppressed the KA-induced activation of the NF-κB pathway and iNOS production. Our results indicated miconazole to be an effective drug for disease-modifying effects during epilepsy, which may act by attenuating neuroinflammation through the suppression of NF-κB activation and iNOS production. At appropriate doses, miconazole may be a safe and effective approved drug that can easily be repositioned for clinical practice.


Asunto(s)
Epilepsia , FN-kappa B , Citocinas , Epilepsia/tratamiento farmacológico , Humanos , Miconazol/efectos adversos , FN-kappa B/metabolismo , Enfermedades Neuroinflamatorias , Convulsiones/metabolismo
3.
J Neuroinflammation ; 19(1): 226, 2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36104755

RESUMEN

Evidence from experimental and clinical studies implicates immuno-inflammatory responses as playing an important role in epilepsy-induced brain injury. Captopril, an angiotensin-converting enzyme inhibitor (ACEi), has previously been shown to suppress immuno-inflammatory responses in a variety of neurological diseases. However, the therapeutic potential of captopril on epilepsy remains unclear. In the present study, Sprague Dawley (SD) rats were intraperitoneally subjected to kainic acid (KA) to establish a status epilepticus. Captopril (50 mg/kg, i.p.) was administered daily following the KA administration from day 3 to 49. We found that captopril efficiently suppressed the KA-induced epilepsy, as measured by electroencephalography. Moreover, captopril ameliorated the epilepsy-induced cognitive deficits, with improved performance in the Morris water maze, Y-maze and novel objective test. RNA sequencing (RNA-seq) analysis indicated that captopril reversed a wide range of epilepsy-related biological processes, particularly the glial activation, complement system-mediated phagocytosis and the production of inflammatory factors. Interestingly, captopril suppressed the epilepsy-induced activation and abnormal contact between astrocytes and microglia. Immunohistochemical experiments demonstrated that captopril attenuated microglia-dependent synaptic remodeling presumably through C3-C3ar-mediated phagocytosis in the hippocampus. Finally, the above effects of captopril were partially blocked by an intranasal application of recombinant C3a (1.3 µg/kg/day). Our findings demonstrated that captopril reduced the occurrence of epilepsy and cognitive impairment by attenuation of inflammation and C3-mediated synaptic phagocytosis. This approach can easily be adapted to long-term efficacy and safety in clinical practice.


Asunto(s)
Disfunción Cognitiva , Epilepsia , Animales , Captopril/farmacología , Captopril/uso terapéutico , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Ácido Kaínico/toxicidad , Fagocitosis , Ratas , Ratas Sprague-Dawley
4.
Neurobiol Dis ; 160: 105534, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34673151

RESUMEN

Multiple sclerosis (MS) is a disease of the central nervous system (CNS) that is characterized by demyelination, axonal injury and neurological deterioration. Few medications are available for progressive MS, which is associated with neuroinflammation confined to the CNS compartment. Transient receptor potential melastatin 2 (TRPM2) is a calcium-permeable, non-selective cation channel that plays pathological roles in a wide range of neuroinflammatory diseases; however, the underlying molecular mechanisms of TRPM2 remain elusive. Here, we established a cuprizone model that presents hallmark MS pathologies to investigate the role of TRPM2 in progressive MS. We demonstrated that genetic deletion of TRPM2 yields protection from the cuprizone-induced demyelination, synapse loss, microglial activation, NLRP3 inflammasome activation and proinflammatory cytokines production and ultimately leads to an improvement in cognitive decline. Furthermore, we showed that the pharmacological inhibition of NLRP3 ameliorated the demyelination, neuroinflammation and cognitive impairment in the model with no additive effects on the TRPM2 KO mice. Taken together, these results indicated that TRPM2 plays important roles in regulating neuroinflammation in progressive MS via NLRP3 inflammasome, and the results shed light on TRPM2's potential role as a therapeutic target for MS.


Asunto(s)
Disfunción Cognitiva/metabolismo , Inflamasomas/metabolismo , Esclerosis Múltiple/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Canales Catiónicos TRPM/metabolismo , Animales , Disfunción Cognitiva/genética , Cuprizona , Modelos Animales de Enfermedad , Inflamasomas/genética , Ratones , Ratones Noqueados , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/genética , Enfermedades Neuroinflamatorias/genética , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPM/genética
6.
Mol Med Rep ; 29(5)2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38516767

RESUMEN

Acute lung injury (ALI) is an acute inflammatory lung disease associated with both innate and adaptive immune responses. Hexokinase 2 (HK2) is specifically highly expressed in numerous types of inflammation­related diseases and models. In the present study in vitro and in vivo effects of targeted degradation of HK2 on ALI were explored. The degradation of HK2 by the targeting peptide TAT (transactivator of transcription protein of HIV­1)­ataxin 1 (ATXN1)­chaperone­mediated autophagy­targeting motif (CTM) was demonstrated by ELISA and western blotting in vitro and in vivo. The inhibitory effects of TAT­ATXN1­CTM on lipopolysaccharide (LPS)­induced inflammatory responses were examined using ELISAs. The therapeutic effects of TAT­ATXN1­CTM on LPS­induced ALI were examined via histological examination and ELISAs in mice. 10 µM TAT­ATXN1­CTM administration decreased HK2 protein expression and the secretion of proinflammatory cytokines (TNF­α and IL­1ß) without altering HK2 mRNA expression in LPS­treated both in vitro and in vivo, while pathological lung tissue damage and the accumulation of leukocytes, neutrophils, macrophages and lymphocytes in ALI were also significantly suppressed by 10 µM TAT­ATXN1­CTM treatment. TAT­ATXN1­CTM exhibited anti­inflammatory activity in vitro and decreased the severity of ALI in vivo. HK2 degradation may represent a novel therapeutic approach for ALI.


Asunto(s)
Lesión Pulmonar Aguda , Hexoquinasa , Animales , Ratones , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Hexoquinasa/antagonistas & inhibidores , Hexoquinasa/metabolismo , Lipopolisacáridos/efectos adversos , Pulmón/patología
7.
Neurophotonics ; 11(1): 015001, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38125610

RESUMEN

Significance: Comorbidities such as mood and cognitive disorders are often found in individuals with epilepsy after seizures. Cortex processes sensory, motor, and cognitive information. Brain circuit changes can be studied by observing functional network changes in epileptic mice's cortex. Aim: The cortex is easily accessible for non-invasive brain imaging and electroencephalogram recording (EEG). However, the impact of seizures on cortical activity and functional connectivity has been rarely studied in vivo. Approach: Intrinsic optical signal and EEG were used to monitor cortical activity in awake mice within 4 h after pilocarpine induction. It was divided into three periods according to the behavior and EEG of the mice: baseline, onset of seizures (onset, including seizures and resting in between seizure events), and after seizures (post, without seizures). Changes in cortical activity were compared between the baseline and after seizures. Results: Hemoglobin levels increased significantly, particularly in the parietal association cortex (PT), retrosplenial cortex (RS), primary visual cortex (V1), and secondary visual cortex (V2). The network-wide functional connectivity changed post seizures, e.g., hypoconnectivity between PT and visual-associated cortex (e.g., V1 and V2). In contrast, connectivity between the motor-associated cortex and most other regions increased. In addition, the default mode network (DMN) also changed after seizures, with decreased connectivity between primary somatosensory region (SSp) and visual region (VIS), but increased connectivity involving anterior cingulate cortex (AC) and RS. Conclusions: Our results provide references for understanding the mechanisms behind changes in brain circuits, which may explain the profound effects of seizures on comorbid health conditions.

8.
Int Immunopharmacol ; 139: 112744, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39059098

RESUMEN

OBJECTIVE: Sepsis-associated encephalopathy (SAE) can lead to severe cerebral dysfunction as well as cognitive dysfunction, resulting in a significant disease burden. 3-Methyladenine (3-MA) has been confirmed to have anti-inflammatory effects on diseases characterized by enhanced autophagy. However, its role in SAE has not been clarified. METHODS: An SAE mouse model was generated by intraperitoneal injection of lipopolysaccharide (LPS). Mice were given 5, 20, or 80 mg/kg 3-MA to determine the therapeutic dose. The mice in the different groups were given 20 mg/kg 3-MA or saline, and survival, body temperature, body weight and neurobehavioral scores were measured at different time points. The expression of autophagy-related proteins and inflammatory factors was detected by Western blotting, enzyme linked immunosorbent assay (ELISA) and real-time quantitative polymerase chain reaction (RT-qPCR) 12 h after LPS induction. Glial activation and neuronal injury in the hippocampus were detected by immunofluorescence staining and HE staining. The open Field test, novel object recognition (NOR) test, Y-maze test, and Morris water maze (MWM) test were performed to assess cognitive function. RESULTS: Treatment with 20 or 80 mg/kg 3-MA reduced the increase in hippocampal TNF-α, IL-6, and IL-1ß expression in SAE model mice, with 20 mg/kg 3-MA having the greatest therapeutic effect. Treatment with 20 mg/kg 3-MA effectively reduced the expression of hippocampal autophagy-related proteins and mortality, ameliorated hypothermia, decreased body weight and electroencephalography (EEG) performance, and attenuated the activation of neuroglia and neuronal damage. Moreover, it alleviated the cognitive dysfunction 2 weeks after LPS induction. CONCLUSIONS: 3-MA reduced neuroglial activation and neuronal damage, attenuated neuroinflammation, and improved cognitive deficits during recovery period by inhibiting autophagy in SAE.


Asunto(s)
Adenina , Autofagia , Cognición , Lipopolisacáridos , Enfermedades Neuroinflamatorias , Encefalopatía Asociada a la Sepsis , Animales , Encefalopatía Asociada a la Sepsis/tratamiento farmacológico , Autofagia/efectos de los fármacos , Adenina/análogos & derivados , Adenina/farmacología , Masculino , Ratones , Cognición/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/inmunología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Citocinas/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Antiinflamatorios/uso terapéutico , Antiinflamatorios/farmacología
9.
Neurosci Bull ; 40(7): 887-904, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38321347

RESUMEN

Synapse organizers are essential for the development, transmission, and plasticity of synapses. Acting as rare synapse suppressors, the MAM domain containing glycosylphosphatidylinositol anchor (MDGA) proteins contributes to synapse organization by inhibiting the formation of the synaptogenic neuroligin-neurexin complex. A previous analysis of MDGA2 mice lacking a single copy of Mdga2 revealed upregulated glutamatergic synapses and behaviors consistent with autism. However, MDGA2 is expressed in diverse cell types and is localized to both excitatory and inhibitory synapses. Differentiating the network versus cell-specific effects of MDGA2 loss-of-function requires a cell-type and brain region-selective strategy. To address this, we generated mice harboring a conditional knockout of Mdga2 restricted to CA1 pyramidal neurons. Here we report that MDGA2 suppresses the density and function of excitatory synapses selectively on pyramidal neurons in the mature hippocampus. Conditional deletion of Mdga2 in CA1 pyramidal neurons of adult mice upregulated miniature and spontaneous excitatory postsynaptic potentials, vesicular glutamate transporter 1 intensity, and neuronal excitability. These effects were limited to glutamatergic synapses as no changes were detected in miniature and spontaneous inhibitory postsynaptic potential properties or vesicular GABA transporter intensity. Functionally, evoked basal synaptic transmission and AMPAR receptor currents were enhanced at glutamatergic inputs. At a behavioral level, memory appeared to be compromised in Mdga2 cKO mice as both novel object recognition and contextual fear conditioning performance were impaired, consistent with deficits in long-term potentiation in the CA3-CA1 pathway. Social affiliation, a behavioral analog of social deficits in autism, was similarly compromised. These results demonstrate that MDGA2 confines the properties of excitatory synapses to CA1 neurons in mature hippocampal circuits, thereby optimizing this network for plasticity, cognition, and social behaviors.


Asunto(s)
Región CA1 Hipocampal , Plasticidad Neuronal , Células Piramidales , Conducta Social , Sinapsis , Animales , Masculino , Ratones , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Memoria/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal/fisiología , Células Piramidales/fisiología , Células Piramidales/metabolismo , Sinapsis/metabolismo , Sinapsis/fisiología
10.
CNS Neurosci Ther ; 29(10): 2884-2900, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37072932

RESUMEN

BACKGROUND: A consensus has formed that neural circuits in the brain underlie the pathogenesis of temporal lobe epilepsy (TLE). In particular, the synaptic excitation/inhibition balance (E/I balance) has been implicated in shifting towards elevated excitation during the development of TLE. METHODS: Sprague Dawley (SD) rats were intraperitoneally subjected to kainic acid (KA) to generate a model of TLE. Next, electroencephalography (EEG) recording was applied to verify the stability and detectability of spontaneous recurrent seizures (SRS) in rats. Moreover, hippocampal slices from rats and patients with mesial temporal lobe epilepsy (mTLE) were assessed using immunofluorescence to determine the alterations of excitatory and inhibitory synapses and microglial phagocytosis. RESULTS: We found that KA induced stable SRSs 14 days after status epilepticus (SE) onset. Furthermore, we discovered a continuous increase in excitatory synapses during epileptogenesis, where the total area of vesicular glutamate transporter 1 (vGluT1) rose considerably in the stratum radiatum (SR) of cornu ammonis 1 (CA1), the stratum lucidum (SL) of CA3, and the polymorphic layer (PML) of the dentate gyrus (DG). In contrast, inhibitory synapses decreased significantly, with the total area of glutamate decarboxylase 65 (GAD65) in the SL and PML diminishing enormously. Moreover, microglia conducted active synaptic phagocytosis after the formation of SRSs, especially in the SL and PML. Finally, microglia preferentially pruned inhibitory synapses during recurrent seizures in both rat and human hippocampal slices, which contributed to the synaptic alteration in hippocampal subregions. CONCLUSIONS: Our findings elaborately characterize the alteration of neural circuits and demonstrate the selectivity of synaptic phagocytosis mediated by microglia in TLE, which could strengthen the comprehension of the pathogenesis of TLE and inspire potential therapeutic targets for epilepsy treatment.


Asunto(s)
Epilepsia del Lóbulo Temporal , Humanos , Ratas , Animales , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/patología , Microglía/patología , Ratas Sprague-Dawley , Hipocampo/patología , Convulsiones/patología , Región CA1 Hipocampal , Sinapsis/patología , Ácido Kaínico/toxicidad , Modelos Animales de Enfermedad
11.
Mol Neurobiol ; 59(11): 6918-6933, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36053438

RESUMEN

Epilepsy is a chronic brain disease that makes serious cognitive and motor retardation. Ion channels affect the occurrence of epilepsy in various ways, but the mechanisms have not yet been fully elucidated. Transient receptor potential melastain2 (TRPM2) ion channel is a non-selective cationic channel that can permeate Ca2+ and critical for epilepsy. Here, TRPM2 gene knockout mice were used to generate a chronic kindling epilepsy model by PTZ administration in mice. We found that TRPM2 knockout mice were more susceptible to epilepsy than WT mice. Furthermore, the neuronal excitability in the hippocampal CA1 region of TRPM2 knockout mice was significantly increased. Compared with WT group, there were no significant differences in the input resistance and after hyperpolarization of CA1 neurons in TRPM2 knockout mice. Firing adaptation rate of hippocampal CA1 pyramidal neurons of TRPM2 knockout mice was lower than that of WT mice. We also found that activation of Kv7 channel by retigabine reduced the firing frequency of action potential in the hippocampal pyramidal neurons of TRPM2 knockout mice. However, inhibiting Kv7 channel increased the firing frequency of action potential in hippocampal pyramidal neurons of WT mice. The data suggest that activation of Kv7 channel can effectively reduce epileptic seizures in TRPM2 knockout mice. We conclude that genetic knockout of TRPM2 in hippocampal CA1 pyramidal neurons may increase neuronal excitability by inhibiting Kv7 channel, affecting the susceptibility to epilepsy. These findings may provide a potential therapeutic target for epilepsy.


Asunto(s)
Región CA1 Hipocampal , Epilepsia , Células Piramidales , Canales Catiónicos TRPM , Potenciales de Acción , Animales , Región CA1 Hipocampal/citología , Epilepsia/genética , Técnicas de Inactivación de Genes , Ratones , Ratones Noqueados , Células Piramidales/fisiología , Canales Catiónicos TRPM/genética
12.
Int Immunopharmacol ; 87: 106824, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32731181

RESUMEN

Neuroinflammation contributes to the generation of epileptic seizures and is associate with neuropathology and comorbidities. Transient receptor potential melastatin 2 (TRPM2) expresses in various cell types in the brain. It plays a pathological role in a wide range of neuroinflammatory diseases, but has yet been studied in epilepsy. Here, a temporal lobe epilepsy model was generated by pilocarpine administration in mice. At 24 h, knockout (KO) TRPM2 alleviated the level of neuroinflammation, showing a reduction of IL-1ß, TNF-α, CXCL2 and IL-6 mRNA production, NLRP3, ASC, and Caspase-1 protein expression and glial activation. Moreover, KO TRPM2 alleviated neurodegeneration, concurrent with reduced Beclin-1 and ATG5 protein expression. Later, KO TRPM2 ameliorated the epilepsy-induced psychological disorders, with improved performance in the open-field, Y maze and novel object recognition test. Together, these results suggest that TRPM2 facilitates epilepsy-related brain injury and may shed light on its potential as a therapeutic target for epilepsy-associated neuropathology and comorbidities.


Asunto(s)
Cognición , Epilepsia , Canales Catiónicos TRPM , Animales , Conducta Animal , Citocinas/genética , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/patología , Hipocampo/metabolismo , Hipocampo/patología , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/patología , Pilocarpina , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo
13.
Talanta ; 72(5): 1667-74, 2007 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19071814

RESUMEN

The photocatalytic degradation of organochlorine pesticides including alpha-, beta-, gamma-, delta-hexachlorobenzene (BHC), dicofol and cypermethrin were carried out on a nano-TiO(2) coated films under UV irradiation in the air. The photocatalytic conditions, including the amount of TiO(2), irradiation time and the intensity of light were optimized. The pesticides were most effectively degraded under the condition of 2.24 mg/cm(2) on TiO(2) film and a 400W UV irradiation of high-pressure mercury lamp with a wavelength of 365 nm. A typical organochlorine pesticide, 20 microg alpha-BHC, was dipped onto the TiO(2) film surface and degraded completely within 20 min. In addition, the photocatalytic degradation pathways on the nano-TiO(2) coated film were discussed.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA