Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Am J Emerg Med ; 2023 Dec 10.
Article En | MEDLINE | ID: mdl-38155031

BACKGROUND: Homocysteine (Hcy) is widely recognized as a significant risk factor for cardiovascular and cerebrovascular diseases. However, our research has uncovered a novel perspective, suggesting that elevated levels of Hcy could serve as an indicator for neurological diseases. This article presents a unique case of Subacute Combined Degeneration of the spinal cord(SCD), characterized by high homocysteine levels, yet normal vitamin B12 and imaging results. This discovery could facilitate early detection and ensure timely referral of patients to specialized departments for further treatment.

2.
Med Sci Monit ; 27: e934043, 2021 Dec 06.
Article En | MEDLINE | ID: mdl-34866132

BACKGROUND Being refractory to drugs remains an urgent treatment problem in status epilepticus (SE). The fact that γ-aminobutyric acid A receptors (GABAARs) become internalized and inactive, N-methyl-D-aspartate receptors (NMDARs) become externalized and active during SE may explain the refractoriness to benzodiazepine. However, the real-time dynamic efficacy of antiepileptic drugs remains unclear. Therefore, we propose a hypothesis that diazepam monotherapy or diazepam-ketamine dual therapy could terminate seizures and reduce mortality in the SE model at different time points during ongoing SE. MATERIAL AND METHODS An SE model was established in adult Sprague-Dawley rats with lithium and pilocarpine. The GABAAR agonist diazepam was injected at 5, 10, 20, or 30 min when SE continued. In addition, diazepam and the NMDAR antagonist ketamine were injected at 10 to 60 min at 6 different time points. We measured seizure-free rates, seizure duration, degree of behavioral seizure, and mortality. RESULTS Diazepam monotherapy at 5 min and 10 min from the beginning of SE was able to terminate seizures and improved survival rates. Diazepam-ketamine dual therapy at 10 min, 20 min, and 30 min from the beginning of SE terminated seizures and achieved high survival rates. CONCLUSIONS In this parallel randomized controlled trial with a rat model, we found that diazepam monotherapy was an effective antiepileptic strategy at the early stage of SE less than 10 min after SE onset. If SE lasts more than 10 min but less than 30 min, the diazepam-ketamine dual therapy strategy may be an appropriate choice.


Diazepam/pharmacology , Ketamine/pharmacology , Seizures/drug therapy , Status Epilepticus/drug therapy , Status Epilepticus/mortality , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/pharmacology , Diazepam/administration & dosage , Disease Models, Animal , Drug Administration Schedule , Drug Therapy, Combination/methods , Female , Ketamine/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Seizures/mortality , Treatment Outcome
3.
Exp Ther Med ; 22(4): 1045, 2021 Oct.
Article En | MEDLINE | ID: mdl-34434259

Cerebral ischemia is among the leading causes of death and long-term disability worldwide. The aim of the present study was to investigate the effects of aloperine (ALO) on cerebral ischemia/reperfusion (I/R) injury in rats and elucidate the possible underlying mechanisms. Therefore, a rat model of reversible middle cerebral artery occlusion (MCAO) was established to induce cerebral I/R injury. Following pretreatment with different doses of ALO, the histopathological changes in the brain tissue were evaluated by hematoxylin and eosin staining. The degree of cerebral infarction was determined using by 2,3,5-triphenyltetrazolium chloride staining. Additionally, the levels of oxidative stress- and inflammation-related factors were measured using commercially available kits. Cell apoptosis was assessed by TUNEL staining, while the expression levels of apoptosis- and PI3K/AKT signaling pathway-related proteins were determined by western blot analysis. The results demonstrated that ALO alleviated histopathological injury in the brain tissue and the area of cerebral infarction in a dose-dependent manner. Furthermore, significantly reduced levels of reactive oxygen species and malondialdehyde were observed in the ALO-treated rats post-MCAO/reperfusion, accompanied by increased levels of superoxide dismutase, catalase and glutathione. Consistently, treatment with ALO notably decreased the concentration of inflammatory factors, including TNF-α, IL-1ß and IL-6, in a dose-dependent manner. In addition, ALO attenuated neuronal cell apoptosis, downregulated the expression of Bax and upregulated that of Bcl-2. I/R markedly reduced the expression levels of phosphorylated (p-)PI3K and p-AKT, which were dose-dependently restored by ALO intervention. Collectively, the aforementioned findings indicated that ALO could improve cerebral I/R injury and alleviate oxidative stress, inflammation and cell apoptosis via activating the PI3K/AKT signaling pathway, thus supporting the therapeutic potential of ALO against cerebral I/R injury in ischemic stroke.

4.
Front Neurosci ; 14: 737, 2020.
Article En | MEDLINE | ID: mdl-32848539

Epilepsy is one of the most common chronic neurological diseases in the world, with a high incidence, a high risk of sudden unexplained death, and diagnostic challenges. Exosomes are nanosized extracellular vesicles that are released into physical environments and carry a variety of biological information. Moreover, exosomes can also be synthesized and released from brain cells, passing through the blood-brain barrier, and can be detected in peripheral blood or cerebrospinal fluid. Our study using the tandem mass tag (TMT) approach showed that a total of 76 proteins were differentially expressed in serum exosomes between epilepsy patients and healthy controls, with 6 proteins increasing and 70 proteins decreasing. Analysis of large clinical samples and two mouse models of chronic epilepsy indicated that two significantly differentially expressed serum exosomal proteins, coagulation factor IX (F9) and thrombospondin-1 (TSP-1), represent promising biomarkers for the diagnosis of epilepsy, with area under the curve (AUC) values of up to 0.7776 (95% CI, 0.7306-0.8246) and 0.8534 (95% CI, 0.8152-0.8916), respectively. This is the first study of exosomal proteins in epilepsy, and it suggests that exosomes are promising new tools for the diagnosis of epilepsy.

5.
Biochem Biophys Res Commun ; 524(4): 1057-1063, 2020 04 16.
Article En | MEDLINE | ID: mdl-32067740

Adherens junction-associated protein-1 (AJAP1), also called SHREW1, was first discovered as a novel component of adherens junctions in 2004. In later studies, AJAP1 was found to suppress invasion and predict recurrence of some tumors. Apart from its function as a putative tumor suppressor, AJAP1 is still poorly understood. Schwenk et al. recently found that AJAP1 was tightly associated with the γ-Aminobutyric acid type B receptor subunit 1(GABABR1). It is well known that GABABR plays a vital role in epilepsy as an inhibitory transmitter receptor. Structurally adjacent, possibly functionally interacting, therefore, we hypothesize that AJAP1 participates in the onset and progression of epilepsy. We designed this experiment to investigate the expression and location of AJAP1 in temporal lobe epilepsy (TLE) patients and kainic acid(KA)-induced epilepsy animal models by immunofluorescence and Western blot analyses. We overexpressed and inhibited AJAP1 through lentiviruses in KA-induced models and observed the corresponding effects on epileptic animals. Double-label immunofluorescence showed that AJAP1 was expressed mainly in neurons. Western blot analysis revealed that AJAP1 expression was downregulated in the neocortex of TLE patients and the hippocampus and neocortex of epileptic animal models. The overexpression of AJAP1 can reduce the frequency of spontaneous seizures, whereas the inhibition of AJAP1 expression can increase the incidence rate. Our study demonstrated that AJAP1 may be involved in the pathogenic process of epilepsy and may represent a novel antiepileptic target.


Epilepsy/metabolism , Receptors, GABA-B/metabolism , Adolescent , Adult , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/metabolism , Child , Epilepsy/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Receptors, GABA-B/analysis , Young Adult
6.
Cell Death Dis ; 10(12): 897, 2019 11 27.
Article En | MEDLINE | ID: mdl-31772151

Epilepsy is a serious neurological disease characterized by recurrent unprovoked seizures. The exact etiology of epilepsy is not fully understood. Protrudin is a neural membrane protein and is found to be mutated in hereditary spastic paraplegia that characterized by symptoms like seizures. Here, we reported that the expression of protrudin was downregulated in the temporal neocortex of epileptic patients and in the hippocampus and cortex of pentylenetetrazol and kainic acid-kindled epileptic mouse models. Behavioral and electroencephalogram analyses indicated that overexpression of protrudin in the mouse hippocampus increased the latency of the seizure and decreased the frequency and duration of seizure activity. Using whole-cell patch clamp, overexpression of protrudin in the mouse hippocampus resulted in a reduction in action potential frequency and an increase in gamma-aminobutyric acid (GABA)ergic inhibitory current amplitude. Moreover, western blot analysis showed that the membrane expression of the GABA A receptor ß2/3 subunit was also upregulated after protrudin overexpression, and coimmunoprecipitation resulted in a protein-protein interaction between protrudin, GABAARß2/3 and GABA receptor-associated protein in the hippocampus of epileptic mice. These findings suggest that protrudin probably inhibits the occurrence and development of epilepsy through the regulation of GABAA receptor-mediated synaptic transmission, and protrudin might be a promising target for the treatment of epilepsy.


Receptors, GABA-A/metabolism , Seizures/metabolism , Vesicular Transport Proteins/metabolism , Action Potentials , Adolescent , Adult , Animals , Child , Disease Models, Animal , Epilepsy/metabolism , Epilepsy, Temporal Lobe/metabolism , Female , Green Fluorescent Proteins/metabolism , Hippocampus/pathology , Humans , Inhibitory Postsynaptic Potentials , Lentivirus , Male , Mice , Middle Aged , Neural Inhibition , Protein Binding , Synaptic Transmission , Young Adult
7.
J Clin Invest ; 129(9): 3864-3876, 2019 09 03.
Article En | MEDLINE | ID: mdl-31424425

The expression of the transmembrane protein 25 gene (Tmem25) is strongly influenced by glutamate ionotropic receptor kainate type subunit 4, and its function remains unknown. Here, we showed that TMEM25 was primarily localized to late endosomes in neurons. Electrophysiological experiments suggested that the effects of TMEM25 on neuronal excitability were likely mediated by N-methyl-d-aspartate receptors. TMEM25 affected the expression of the N-methyl-d-aspartate receptor NR2B subunit and interacted with NR2B, and both were colocalized to late endosome compartments. TMEM25 induced acidification changes in lysosome compartments and accelerated the degradation of NR2B. Furthermore, TMEM25 expression was decreased in brain tissues from patients with epilepsy and epileptic mice. TMEM25 overexpression attenuated the behavioral phenotypes of epileptic seizures, whereas TMEM25 downregulation exerted the opposite effect. These results provide some insights into TMEM25 biology in the brain and the functional relationship between TMEM25 and epilepsy.


Brain/metabolism , Endosomes/metabolism , Epilepsy/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission , Animals , Endosomes/genetics , Epilepsy/genetics , HEK293 Cells , Humans , Mice , Receptors, N-Methyl-D-Aspartate/genetics
8.
Brain Res ; 1721: 146332, 2019 10 15.
Article En | MEDLINE | ID: mdl-31301272

BACKGROUND: To explore the causative genes and pathogenesis of autosomal dominant partial epilepsy with auditory features in a large Chinese family that includes 7 patients over four generations. METHODS: We used targeted exome sequencing and Sanger sequencing to validate the mutation. Zebrafish were used to explore the epileptic behavior caused by the mutation. Primary cortical neuronal culturing and in utero electroporation were used to observe the influences of the mutation on neuronal polarity and migration. RESULTS: We report the identification of a novel missense mutation, c.128C > G (p. Pro43Arg), in exon 1 of LGI1. The heterozygous missense mutation, which cosegregated with the syndrome, was absent in 300 unrelated and matched-ancestor controls. The mutation inhibited the secretion of LGI1 and could not rescue the hyperactivity caused by lgi1a knockdown in zebrafish. In vitro, mutant LGI1 interrupts normal cell polarity. In agreement with these findings, dysfunctional cortical neuron migration was observed using in utero electroporation technology, which is reminiscent of the subtle structural changes in the lateral temporal region observed in the proband of this family. CONCLUSION: Our findings enrich the spectrum of LGI1 mutations and support the pathogenicity of the mutation. Furthermore, additional information regarding the role of LGI1 in the development of temporal lobe epilepsy was elucidated, and a potential relationship was established between cortical neuronal migration dysfunction and autosomal dominant partial epilepsy with auditory features.


Epilepsies, Partial/genetics , Epilepsies, Partial/pathology , Intracellular Signaling Peptides and Proteins/genetics , Adult , Asian People , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/pathology , Exons/genetics , Family , Female , Genotype , Heterozygote , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Mutation/genetics , Mutation, Missense/genetics , Pedigree , Phenotype , Seizures/genetics , Seizures/pathology
10.
Cell Death Dis ; 9(11): 1058, 2018 10 17.
Article En | MEDLINE | ID: mdl-30333479

The proprotein convertase Furin plays crucial roles in the pathology of many diseases. However, the specific role of furin in epilepsy remains unclear. In our study, furin protein was increased in the temporal neocortex of epileptic patients and in the hippocampus and cortex of epileptic mice. The furin transgenic (TG) mice showed increased susceptibility to epilepsy and heightened epileptic activity compared with wild-type (WT) mice. Conversely, lentivirus-mediated knockdown of furin restrained epileptic activity. Using whole-cell patch clamp, furin knockdown and overexpression influenced neuronal inhibitory by regulating postsynaptic gamma-aminobutyric acid A receptor (GABAAR)-mediated synaptic transmission. Importantly, furin influenced the expression of GABAAR ß2/3 membrane and total protein in epileptic mice by changing transcription level of GABAAR ß2/3, not the protein degradation. These results reveal that furin may regulate GABAAR-mediated inhibitory synaptic transmission by altering the transcription of GABAAR ß2/3 subunits in epilepsy; this finding could provide new insight into epilepsy prevention and treatment.


Epilepsy/genetics , Furin/genetics , Genetic Predisposition to Disease , Receptors, GABA-A/genetics , Receptors, GABA/genetics , Synaptic Transmission/genetics , Action Potentials/genetics , Adolescent , Adult , Aged , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Child , Epilepsy/metabolism , Epilepsy/physiopathology , Female , Furin/antagonists & inhibitors , Furin/metabolism , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Neurons/metabolism , Neurons/pathology , Oligonucleotides/genetics , Oligonucleotides/metabolism , Patch-Clamp Techniques , Receptors, GABA/metabolism , Receptors, GABA-A/metabolism , Synapses/genetics , Synapses/metabolism , Synapses/pathology , Transcription, Genetic , Transgenes
11.
Epilepsia Open ; 3(3): 331-339, 2018 Sep.
Article En | MEDLINE | ID: mdl-30187003

Research findings on the molecular mechanisms of epilepsy almost always originate from animal experiments, and the development of induced pluripotent stem cell (iPSC) technology allows the use of human cells with genetic defects for studying the molecular mechanisms of genetic epilepsy (GE) for the first time. With iPSC technology, terminally differentiated cells collected from GE patients with specific genetic etiologies can be differentiated into many relevant cell subtypes that carry all of the GE patient's genetic information. iPSCs have opened up a new research field involving the pathogenesis of GE. Using this approach, studies have found that gene mutations induce GE by altering the balance between neuronal excitation and inhibition, which is associated. among other factors, with neuronal developmental disturbances, ion channel abnormalities, and synaptic dysfunction. Simultaneously, astrocyte activation, mitochondrial dysfunction, and abnormal signaling pathway activity are also important factors in the molecular mechanisms of GE.

12.
Epilepsy Res ; 146: 112-120, 2018 10.
Article En | MEDLINE | ID: mdl-30096484

Lithium, a classic mood stabilizer, prevents apoptosis-dependent cellular death and has garnered considerable interest as a neuroprotective agent that is efficacious in the treatment of many neurological diseases. However, the effects of lithium in epilepsy remain controversial. We found that different doses of lithium affect epileptic seizure activity and bidirectionally modulate the susceptibility to and severity of seizures induced by pilocarpine in rats. Recently, it has been demonstrated that systematically administered lithium affects the powers of hippocampal gamma and theta oscillations in baseline electroencephalograms. Low-dose lithium (10 mg/kg) administered to pilocarpine-treated rats markedly increased the powers of basal gamma (30-80 Hz) and theta (4-12 Hz) oscillations, decreased the proportion of Racine stage 4-5 seizures, extended latency until seizure onset, and significantly reduced the frequency of lower-class seizures (p < 0.05). Conversely, when the dose was increased to 40 mg/kg, lithium reduced the frequency of lower-class seizures compared to control treatment (p < 0.05). Further, at this high dose, lithium reduced the power of basal gamma oscillations and markedly increased the susceptibility to and severity of pilocarpine-induced seizures and enhanced ripple rhythms (80-200 Hz) postictally. Our results provide a framework for further investigations of the underlying electrophysiological mechanisms of lithium-induced imbalances in excitatory and inhibitory neural circuits that regulate seizure activity in rats. In conclusion, the observed in vivo changes in the powers of basal gamma and theta oscillations in response to different doses of lithium may reflect hippocampal neural network responsiveness.


Anticonvulsants/pharmacology , Epilepsy/drug therapy , Hippocampus/drug effects , Lithium Compounds/pharmacology , Neuroprotective Agents/pharmacology , Seizures/drug therapy , Animals , Anticonvulsants/adverse effects , Brain Waves/drug effects , Dose-Response Relationship, Drug , Epilepsy/chemically induced , Epilepsy/physiopathology , Hippocampus/physiopathology , Lithium Compounds/adverse effects , Male , Neuroprotective Agents/adverse effects , Pilocarpine , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/physiopathology
13.
Cell Death Dis ; 9(8): 795, 2018 07 23.
Article En | MEDLINE | ID: mdl-30038264

Epilepsy is one of the most prevalent and drug-refractory neurological disorders. Zinc finger DHHC-type containing 8 (ZDHHC8) is a putative palmitoyltransferase that is highly expressed in the brain. However, the impact of ZDHHC8 on seizures remains unclear. We aimed to explore the association of ZDHHC8 with epilepsy and investigate its in epileptogenesis in in vivo and in vitro models through behavioral, electrophysiological, and pathological studies. We used kainic acid- and pilocarpine-induced C57BL/6 mice and magnesium-free-induced pyramidal neurons as experimental epileptic models in this study. We first found increased ZDHHC8 expression in the brains of temporal lobe epilepsy (TLE) patients, similar to that observed in chronic epileptic mice, strongly suggesting that ZDHHC8 is correlated with human epilepsy. In the in vitro seizure models, knocking down ZDHHC8 using recombinant adeno-associated virus (rAAV) delayed seizure precipitation and decreased chronic spontaneous recurrent seizures (SRSs) and epileptiform-like discharges, while ZDHHC8 overexpression had the opposite effect. ZDHHC8 levels were consistent with seizure susceptibility in induced mice with SRSs. In an in vitro magnesium-free model, neuronal hyperexcitability and hypersynchrony were reduced in ZDHHC8-knockdown neurons but were increased in ZDHHC8-overexpressing neurons. To further explore the potential mechanisms, we observed that ZDHHC8 had a significant modulatory effect on 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl) propanoic acid (AMPA) receptor-related excitatory, but not inhibitory, glutamatergic synaptic neurotransmission, further affecting the inward rectification of AMPA currents in acute hippocampal slices in whole-cell recordings. ZDHHC8 facilitated GluA1 trafficking to the neuronal surface in the hippocampus, as shown by immunoprecipitation and Western blotting. These results suggest that ZDHHC8 may promote the generation and propagation of seizures in humans and that knocking down ZDHHC8 might produce anti-epileptogenic effects in drug-resistant epilepsy. Our study provides evidence that may facilitate the development of an alternative approach for the treatment of epilepsy by modulating AMPA/GluA1-mediated neurotransmission.


Acyltransferases/metabolism , Epilepsy, Temporal Lobe/pathology , Acyltransferases/antagonists & inhibitors , Acyltransferases/genetics , Adolescent , Adult , Animals , Brain/metabolism , Child, Preschool , Disease Models, Animal , Epilepsy/chemically induced , Epilepsy/metabolism , Epilepsy/pathology , Epilepsy, Temporal Lobe/metabolism , Female , Hippocampus/drug effects , Hippocampus/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA Interference , RNA, Small Interfering/metabolism , Receptors, AMPA/metabolism , Seizures/chemically induced , Seizures/metabolism , Seizures/pathology , Synaptic Transmission , Young Adult , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
14.
Int J Neurosci ; 126(4): 326-34, 2016.
Article En | MEDLINE | ID: mdl-25539181

OBJECTIVES: During the last decade, experimental evidence has demonstrated an important role of hypoxia, which leads to neuronal cell death and angiogenesis, in the mechanisms of seizure precipitation and recurrence. MicroRNA-199 targets hypoxia-inducible factor-1alpha (HIF-1α), which has recently been implicated in the pathophysiology of the hypoxic state and brain injury. However, little is known about the roles of MicroRNA-199 and HIF-1α in the human epileptogenic process. DESIGN AND METHODS: In this study, we investigated the expression of miR-199a-5p, miR-199b-5p and HIF-1α using real-time PCR, immunohistochemistry and western blots in the temporal neocortex of twenty four patients with intractable epilepsy and twelve control subjects. RESULTS: Compared with the control group, the expression of miR-199a-5p and miR-199b-5p was significantly lower in epileptic brain tissues (p < 0.05). The levels of HIF-1α mRNA and protein were highly up-regulated in epileptic brain tissues compared with those of control subjects (p < 0.05). CONCLUSION: These data suggest that the abnormal expression of miR-199 and HIF-1α in epileptic brain tissue may be involved in the pathophysiology of human epilepsy and that the expression of HIF-1α may be regulated by miR-199. These findings may provide new insights into the treatment of epilepsy.


Drug Resistant Epilepsy/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/biosynthesis , Temporal Lobe/metabolism , Adolescent , Adult , Case-Control Studies , Female , Gene Expression , Humans , Male , Middle Aged , Young Adult
...