Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32.460
Filter
1.
Pak J Pharm Sci ; 37(2(Special)): 435-442, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38822547

ABSTRACT

Depression is a common non-motor symptom of Parkinson's disease. Previous studies demonstrated that hydroxysafflor yellow A had properties of improving motor symptoms of Parkinson's disease. The effect of hydroxysafflor yellow A on depression in Parkinson's disease mice is investigated in this study. To induce Parkinson's disease model, male Swiss mice were exposed to rotenone (30 mg/kg) for 6 weeks. The chronic unpredictable mild stress was employed to induce depression from week 3 to week 6. Sucrose preference, tail suspension, and forced swimming tests were conducted. Golgi and Nissl staining of hippocampus were carried out. The levels of dopamine, 5-hydroxytryptamine and the expression of postsynaptic density protein 95, brain-derived neurotrophic factor in hippocampus were assayed. It showed that HSYA improved the depression-like behaviors of Parkinson's disease mice. Hydroxysafflor yellow A attenuated the injury of nerve and elevated contents of dopamine, 5-hydroxytryptamine in hippocampus. Treatment with hydroxysafflor yellow A also augmented the expression of postsynaptic density protein 95 and brain-derived neurotrophic factor. These findings suggest that hydroxysafflor yellow A ameliorates depression-like behavior in Parkinson's disease mice through regulating the contents of postsynaptic density protein 95 and brain-derived neurotrophic factor, therefore protecting neurons and neuronal dendrites of the hippocampus.


Subject(s)
Behavior, Animal , Brain-Derived Neurotrophic Factor , Chalcone , Depression , Hippocampus , Quinones , Serotonin , Animals , Quinones/pharmacology , Quinones/therapeutic use , Chalcone/analogs & derivatives , Chalcone/pharmacology , Chalcone/therapeutic use , Male , Mice , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Depression/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Behavior, Animal/drug effects , Serotonin/metabolism , Dopamine/metabolism , Rotenone/pharmacology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/psychology
2.
Cell Biol Toxicol ; 40(1): 41, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833095

ABSTRACT

Hippocampal neurons maintain the ability of proliferation throughout life to support neurogenesis. Deoxynivalenol (DON) is a mycotoxin that exhibits brain toxicity, yet whether and how DON affects hippocampal neurogenesis remains unknown. Here, we use mouse hippocampal neuron cells (HT-22) as a model to illustrate the effects of DON on neuron proliferation and to explore underlying mechanisms. DON exposure significantly inhibits the proliferation of HT-22 cells, which is associated with an up-regulation of cell cycle inhibitor p21 at both mRNA and protein levels. Global and site-specific m6A methylation levels on the 3'UTR of p21 mRNA are significantly increased in response to DON treatment, whereas inhibition of m6A hypermethylation significantly alleviates DON-induced cell cycle arrest. Further mechanistic studies indicate that the m6A readers YTHDF1 and IGF2BP1 are responsible for m6A-mediated increase in p21 mRNA stability. Meanwhile, 3'UTR of E3 ubiquitin ligase TRIM21 mRNA is also m6A hypermethylated, and another m6A reader YTHDF2 binds to the m6A sites, leading to decreased TRIM21 mRNA stability. Consequently, TRIM21 suppression impairs ubiquitin-mediated p21 protein degradation. Taken together, m6A-mediated upregulation of p21, at both post-transcriptional and post-translational levels, contributes to DON-induced inhibition of hippocampal neuron proliferation. These results may provide new insights for epigenetic therapy of neurodegenerative diseases.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21 , Hippocampus , Neurons , Trichothecenes , Up-Regulation , Animals , Trichothecenes/toxicity , Trichothecenes/pharmacology , Hippocampus/metabolism , Hippocampus/drug effects , Hippocampus/cytology , Mice , Neurons/drug effects , Neurons/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Up-Regulation/drug effects , Cell Proliferation/drug effects , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Line , 3' Untranslated Regions/genetics , Neurogenesis/drug effects , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA Stability/drug effects , Cell Cycle Checkpoints/drug effects , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Methylation/drug effects
3.
Gynecol Endocrinol ; 40(1): 2360066, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38833172

ABSTRACT

Menopausal transition in women involves complex neurobiochemical changes linked to ovarian dysfunction, resulting in symptoms like vasomotor symptoms (VMS), sleep disturbances, anxiety, and cognitive impairments. Hormone replacement therapy is the first-line treatment. However, many women are reluctant to use HRT or have contraindications toward HRT and seek for alternatives. Non-hormonal therapies with extracts of Cimicifuga racemosa rhizomes like the isopropanolic extract (iCR, black cohosh) offer a promising alternative. A preclinical pilot study exploring iCR's effects on gene expression in the hippocampus and hypothalamus of ovarectomized (OVX) rats mimicking menopausal conditions identified important signaling pathways and CNS-based contributions to the multitargeted modes of action of iCR. Especially in the hippocampus, iCR compensated effects of OVX on gene expression profiles. These changes are reflected by the genes AVPR1A, GAL, CALCA, HCRT, PNOC, ESR1, ESR2 and TAC3 contributing to the formation of hot flushes or thermoregulation as well as to secondary effects such as blood pressure, metabolism, hormonal regulation, homeostasis, mood regulation, neuroendocrine modulation, regulation of sleep and arousal, and in learning, memory and cognition. To understand the mechanisms in the brain of estrogen-depressed animals (OVX) and subsequent iCR treatment we combined the results of the pilot study with those of up-to-date literature and tried to transfer the current knowledge to humans during menopausal transition and adaptation. Focus was laid on changes in the hippocampal function, that is disturbed by hormonal fluctuations, but can also be brought back into balance by iCR.


Subject(s)
Cimicifuga , Hippocampus , Menopause , Plant Extracts , Cimicifuga/chemistry , Hippocampus/drug effects , Hippocampus/metabolism , Female , Animals , Menopause/drug effects , Plant Extracts/pharmacology , Rats , Pilot Projects , Humans , Ovariectomy
4.
Folia Biol (Praha) ; 70(1): 45-52, 2024.
Article in English | MEDLINE | ID: mdl-38830122

ABSTRACT

Effective treatment of patients with autism spectrum disorder (ASD) is still absent so far. Taurine exhibits therapeutic effects towards the autism-like behaviour in ASD model animals. Here, we determined the mechanism of taurine effect on hippocampal neurogenesis in genetically inbred BTBR T+ tf/J (BTBR) mice, a proposed model of ASD. In this ASD mouse model, we explored the effect of oral taurine supplementation on ASD-like behaviours in an open field test, elevated plus maze, marble burying test, self-grooming test, and three-chamber test. The mice were divided into four groups of normal controls (WT) and models (BTBR), who did or did not receive 6-week taurine supplementation in water (WT, WT+ Taurine, BTBR, and BTBR+Taurine). Neurogenesis-related effects were determined by Ki67 immunofluorescence staining. Western blot analysis was performed to detect the expression of phosphatase and tensin homologue deleted from chromosome 10 (PTEN)/mTOR/AKT pathway-associated proteins. Our results showed that taurine improved the autism-like behaviour, increased the proliferation of hippocampal cells, promoted PTEN expression, and reduced phosphorylation of mTOR and AKT in hippocampal tissue of the BTBR mice. In conclusion, taurine reduced the autism-like behaviour in partially inherited autism model mice, which may be associa-ted with improving the defective neural precursor cell proliferation and enhancing the PTEN-associated pathway in hippocampal tissue.


Subject(s)
Autistic Disorder , Hippocampus , Neurogenesis , PTEN Phosphohydrolase , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Taurine , Animals , Taurine/pharmacology , Hippocampus/metabolism , Hippocampus/drug effects , TOR Serine-Threonine Kinases/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Neurogenesis/drug effects , Autistic Disorder/metabolism , Autistic Disorder/drug therapy , Male , Behavior, Animal/drug effects , Mice , Disease Models, Animal , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/drug therapy , Cell Proliferation/drug effects
5.
Transl Psychiatry ; 14(1): 208, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796566

ABSTRACT

In clinical settings, tumor compression, trauma, surgical injury, and other types of injury can cause hypothalamic damage, resulting in various types of hypothalamic dysfunction. Impaired release of oxytocin can lead to cognitive impairment and affect prognosis and long-term quality of life after hypothalamic injury. Hypothalamic injury-induced cognitive dysfunction was detected in male animals. Behavioral parameters were measured to assess the characteristics of cognitive dysfunction induced by hypothalamic-pituitary stalk lesions. Brains were collected for high-throughput RNA sequencing and immunostaining to identify pathophysiological changes in hippocampal regions highly associated with cognitive function after injury to corresponding hypothalamic areas. Through transcriptomic analysis, we confirmed the loss of oxytocin neurons after hypothalamic injury and the reversal of hypothalamic-induced cognitive dysfunction after oxytocin supplementation. Furthermore, overactivation of the ERK signaling pathway and ß-amyloid deposition in the hippocampal region after hypothalamic injury were observed, and cognitive function was restored after inhibition of ERK signaling pathway overactivation. Our findings suggest that cognitive dysfunction after hypothalamic injury may be caused by ERK hyperphosphorylation in the hippocampal region resulting from a decrease in the number of oxytocin neurons, which in turn causes ß-amyloid deposition.


Subject(s)
Amyloid beta-Peptides , Cognitive Dysfunction , Hippocampus , Hypothalamus , MAP Kinase Signaling System , Oxytocin , Oxytocin/metabolism , Oxytocin/pharmacology , Animals , Hippocampus/metabolism , Hippocampus/drug effects , Male , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , MAP Kinase Signaling System/drug effects , Amyloid beta-Peptides/metabolism , Neurons/drug effects , Neurons/metabolism , Disease Models, Animal , Mice , Phosphorylation
6.
Zhongguo Zhong Yao Za Zhi ; 49(7): 1896-1904, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38812202

ABSTRACT

This study aims to analyze the constituents of Jiaotai Pills migrating to the blood in normal rats by UHPLC-TOF-MS technique and reveal the underlying mechanism of Jiaotai Pills in the treatment of depression by network pharmacology and animal experiments. UHPLC-TOF-MS technique was used to detect the constituents of Jiaotai Pills in the blood of rats after intragastric administration. The intersection target of the constituents and depression was screened by DisGeNET and SwissTargetPrediction database, and the protein-protein interaction(PPI) network was constructed. Key targets were imported into the DAVID platform for Gene Ontology(GO) analysis and Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway annotation. Combined with constituents, targets, and pathways, the "constituent-target-pathway" network was constructed by Cytoscape 3.9.1 software, through which the key targets and pathways of Jiaotai Pills against depression were predicted. The depression model of chronic unpredictable mild stress(CUMS) was established on rats. After that, behavioral experiments were conducted. The expression of inflammatory factors in serum and the neurotransmitters in the brain were detected by ELISA, and the expression of key targets in the hippocampus was detected by Western blot. The results showed that a total of 17 constituents of Jiaotai Pills were identified in the blood, including 10 alkaloids. There were 124 intersection targets between constituents of Jiaotai Pills and depression disorder. A total of 52 core targets were screened according to PPI results, including NLRP3 and caspase-1, etc. KEGG enrichment analysis mainly involved 15 typical pathways such as NOD-like receptor pathway. The results of animal experiments showed that Jiaotai Pills significantly improved the depression-like behavior of CUMS depressive model on rats, decreased the levels of IL-1ß, TNF-α and IL-6 in serum, and increased the expression of neurotransmitters such as 5-hydroxytryptamine(5-HT), dopamine(DA), and norepinephrine(NE) in the brain. Besides, Jiaotai Pills also down-regulated the expression of NLRP3 and caspase-1 proteins in the hippocampus and inhibited the NLRP3-mediated NOD-like receptor signaling pathway. In conclusion, Jiaotai Pills may play a role in the treatment of depression by inhibiting the NLRP3 inflammasome and the NOD-like receptor pathway mediated by NLRP3.


Subject(s)
Depression , Drugs, Chinese Herbal , Network Pharmacology , Rats, Sprague-Dawley , Animals , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Depression/drug therapy , Depression/genetics , Depression/metabolism , Rats , Male , Chromatography, High Pressure Liquid , Protein Interaction Maps , Mass Spectrometry , Humans , Hippocampus/drug effects , Hippocampus/metabolism
7.
Addict Biol ; 29(5): e13402, 2024 May.
Article in English | MEDLINE | ID: mdl-38797559

ABSTRACT

Increases in harmful drinking among older adults indicate the need for a more thorough understanding of the relationship between later-life alcohol use and brain health. The current study investigated the relationships between alcohol use and progressive grey and white matter changes in older adults using longitudinal data. A total of 530 participants (aged 70 to 90 years; 46.0% male) were included. Brain outcomes assessed over 6 years included total grey and white matter volume, as well as volume of the hippocampus, thalamus, amygdala, corpus callosum, orbitofrontal cortex and insula. White matter integrity was also investigated. Average alcohol use across the study period was the main exposure of interest. Past-year binge drinking and reduction in drinking from pre-baseline were additional exposures of interest. Within the context of low-level average drinking (averaging 11.7 g per day), higher average amount of alcohol consumed was associated with less atrophy in the left (B = 7.50, pFDR = 0.010) and right (B = 5.98, pFDR = 0.004) thalamus. Past-year binge-drinking was associated with poorer white matter integrity (B = -0.013, pFDR = 0.024). Consuming alcohol more heavily in the past was associated with greater atrophy in anterior (B = -12.73, pFDR = 0.048) and posterior (B = -17.88, pFDR = 0.004) callosal volumes over time. Across alcohol exposures and neuroimaging markers, no other relationships were statistically significant. Within the context of low-level drinking, very few relationships between alcohol use and brain macrostructure were identified. Meanwhile, heavier drinking was negatively associated with white matter integrity.


Subject(s)
Alcohol Drinking , Atrophy , Brain , Gray Matter , Magnetic Resonance Imaging , White Matter , Humans , Male , Aged , Female , Longitudinal Studies , Brain/diagnostic imaging , Brain/pathology , Brain/drug effects , White Matter/diagnostic imaging , White Matter/pathology , White Matter/drug effects , Aged, 80 and over , Gray Matter/pathology , Gray Matter/diagnostic imaging , Gray Matter/drug effects , Atrophy/pathology , Aging/pathology , Aging/physiology , Binge Drinking/pathology , Binge Drinking/diagnostic imaging , Thalamus/diagnostic imaging , Thalamus/pathology , Thalamus/drug effects , Hippocampus/diagnostic imaging , Hippocampus/pathology , Hippocampus/drug effects , Amygdala/diagnostic imaging , Amygdala/pathology , Corpus Callosum/diagnostic imaging , Corpus Callosum/pathology , Corpus Callosum/drug effects
8.
CNS Neurosci Ther ; 30(5): e14778, 2024 May.
Article in English | MEDLINE | ID: mdl-38801174

ABSTRACT

AIMS: Synaptic vesicle protein 2A (SV2A) is a unique therapeutic target for pharmacoresistant epilepsy (PRE). As seizure-induced neuronal programmed death, parthanatos was rarely reported in PRE. Apoptosis-inducing factor (AIF), which has been implicated in parthanatos, shares a common cytoprotective function with SV2A. We aimed to investigate whether parthanatos participates in PRE and is mitigated by SV2A via AIF. METHODS: An intraperitoneal injection of lithium chloride-pilocarpine was used to establish an epileptic rat model, and phenytoin and phenobarbital sodium were utilized to select PRE and pharmacosensitive rats. The expression of SV2A was manipulated via lentivirus delivery into the hippocampus. Video surveillance was used to assess epileptic ethology. Biochemical tests were employed to test hippocampal tissues following a successful SV2A infection. Molecular dynamic calculations were used to simulate the interaction between SV2A and AIF. RESULTS: Parthanatos core index, PARP1, PAR, nuclear AIF and MIF, γ-H2AX, and TUNEL staining were all increased in PRE. SV2A is bound to AIF to form a stable complex, successfully inhibiting AIF and MIF nuclear translocation and parthanatos and consequently mitigating spontaneous recurrent seizures in PRE. Moreover, parthanatos deteriorated after the SV2A reduction. SIGNIFICANCE: SV2A protected hippocampal neurons and mitigated epileptic seizures by inhibiting parthanatos via binding to AIF in PRE.


Subject(s)
Apoptosis Inducing Factor , Disease Models, Animal , Drug Resistant Epilepsy , Membrane Glycoproteins , Nerve Tissue Proteins , Rats, Sprague-Dawley , Animals , Rats , Apoptosis Inducing Factor/metabolism , Male , Nerve Tissue Proteins/metabolism , Drug Resistant Epilepsy/metabolism , Drug Resistant Epilepsy/drug therapy , Membrane Glycoproteins/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Anticonvulsants/pharmacology
9.
Int Immunopharmacol ; 134: 112147, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38718656

ABSTRACT

The neuronal and renal deteriorations observed in patients exposed to methotrexate (MTX) therapy highlight the need for medical interventions to counteract these complications. Boswellic acid (BA) and apigenin (APG) are natural phytochemicals with prominent neuronal and renal protective impacts in various ailments. However, their impacts on MTX-provoked renal and hippocampal toxicity have not been reported. Thus, the present work is tailored to clarify the ability of BA and APG to counteract MTX-provoked hippocampal and renal toxicity. BA (250 mg/kg) or APG (20 mg/kg) were administered orally in rats once a day for 10 days, while MTX (20 mg/kg, i.p.) was administered once on the sixth day of the study. At the histopathological level, BA and APG attenuated MTX-provoked renal and hippocampal aberrations. They also inhibited astrocyte activation, as proven by the inhibition of glial fibrillary acidic protein (GFAP). These impacts were partially mediated via the activation of autophagy flux, as proven by the increased expression of beclin1, LC3-II, and the curbing of p62 protein, alongside the regulation of the p-AMPK/mTOR nexus. In addition, BA and APG displayed anti-inflammatory features as verified by the damping of NOD-2 and p-NF-κB p65 to reduce TNF-α, IL-6, and NLRP3/IL-1ß cue. These promising effects were accompanied with a notable reduction in one of the gap junction proteins, connexin-43 (Conx-43). These positive impacts endorse BA and APG as adjuvant modulators to control MTX-driven hippocampal and nephrotoxicity.


Subject(s)
Apigenin , Autophagy , Connexin 43 , Hippocampus , Kidney , Methotrexate , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Triterpenes , Animals , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Methotrexate/adverse effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Apigenin/pharmacology , Apigenin/therapeutic use , Triterpenes/pharmacology , Triterpenes/therapeutic use , NF-kappa B/metabolism , Male , Rats , Connexin 43/metabolism , Autophagy/drug effects , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Rats, Wistar , Signal Transduction/drug effects
10.
Neurochem Res ; 49(7): 1823-1837, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38727985

ABSTRACT

Methylglyoxal (MG) is considered a classical biomarker of diabetes mellitus and its comorbidities. However, a role for this compound in exacerbated immune responses, such as septicemia, is being increasingly observed and requires clarification, particularly in the context of neuroinflammatory responses. Herein, we used two different approaches (in vivo and acute hippocampal slice models) to investigate MG as a biomarker of neuroinflammation and the neuroimmunometabolic shift to glycolysis in lipopolysaccharide (LPS) inflammation models. Our data reinforce the hypothesis that LPS-induced neuroinflammation stimulates the cerebral innate immune response by increasing IL-1ß, a classical pro-inflammatory cytokine, and the astrocyte reactive response, via elevating S100B secretion and GFAP levels. Acute neuroinflammation promotes an early neuroimmunometabolic shift to glycolysis by elevating glucose uptake, lactate release, PFK1, and PK activities. We observed high serum and cerebral MG levels, in association with a reduction in glyoxalase 1 detoxification activity, and a close correlation between serum and hippocampus MG levels with the systemic and neuroinflammatory responses to LPS. Findings strongly suggest a role for MG in immune responses.


Subject(s)
Biomarkers , Hippocampus , Lipopolysaccharides , Neuroinflammatory Diseases , Pyruvaldehyde , Pyruvaldehyde/metabolism , Lipopolysaccharides/pharmacology , Animals , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/chemically induced , Biomarkers/metabolism , Male , Hippocampus/metabolism , Hippocampus/drug effects , Rats, Wistar , S100 Calcium Binding Protein beta Subunit/metabolism , Glycolysis/drug effects , Interleukin-1beta/metabolism , Inflammation/metabolism , Inflammation/chemically induced , Glial Fibrillary Acidic Protein/metabolism , Lactoylglutathione Lyase/metabolism , Rats , Astrocytes/metabolism , Astrocytes/drug effects
11.
J Neurosci Res ; 102(5): e25338, 2024 May.
Article in English | MEDLINE | ID: mdl-38706427

ABSTRACT

The enzyme indoleamine 2,3 dioxygenase 1 (IDO1) catalyzes the rate-limiting step in the kynurenine pathway (KP) which produces both neuroprotective and neurotoxic metabolites. Neuroinflammatory signals produced as a result of pathological conditions can increase production of IDO1 and boost its enzymatic capacity. IDO1 and the KP have been implicated in behavioral recovery after human traumatic brain injury (TBI), but their roles in experimental models of TBI are for the most part unknown. We hypothesized there is an increase in KP activity in the fluid percussion injury (FPI) model of TBI, and that administration of an IDO1 inhibitor will improve neurological recovery. In this study, adult male Sprague Dawley rats were subjected to FPI or sham injury and received twice-daily oral administration of the IDO1 inhibitor PF-06840003 (100 mg/kg) or vehicle control. FPI resulted in a significant increase in KP activity, as demonstrated by an increased ratio of kynurenine: tryptophan, in the perilesional neocortex and ipsilateral hippocampus 3 days postinjury (DPI), which normalized by 7 DPI. The increase in KP activity was prevented by PF-06840003. IDO1 inhibition also improved memory performance as assessed in the Barnes maze and anxiety behaviors as assessed in open field testing in the first 28 DPI. These results suggest increased KP activity after FPI may mediate neurological dysfunction, and IDO1 inhibition should be further investigated as a potential therapeutic target to improve recovery.


Subject(s)
Brain Injuries, Traumatic , Indoleamine-Pyrrole 2,3,-Dioxygenase , Kynurenine , Rats, Sprague-Dawley , Animals , Male , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Rats , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Kynurenine/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Disease Models, Animal , Recovery of Function/drug effects , Tryptophan/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Maze Learning/drug effects
12.
Brain Behav ; 14(5): e3515, 2024 May.
Article in English | MEDLINE | ID: mdl-38702895

ABSTRACT

INTRODUCTION: Maternal sleep deprivation (MSD), which induces inflammation and synaptic dysfunction in the hippocampus, has been associated with learning and memory impairment in offspring. Melatonin (Mel) has been shown to have anti-inflammatory, antioxidant, and neuroprotective function. However, the beneficial effect of Mel on MSD-induced cognitive impairment and its mechanisms are unknown. METHODS: In the present study, adult offspring suffered from MSD were injected with Mel (20 mg/kg) once a day during postnatal days 61-88. The cognitive function was evaluated by the Morris water maze test. Levels of proinflammatory cytokines were examined by enzyme-linked immunosorbent assay. The mRNA and protein levels of synaptic plasticity associated proteins were examined using reverse transcription-polymerase chain reaction and western blotting. RESULTS: The results showed that MSD impaired learning and memory in the offspring mice. MSD increased the levels of interleukin (IL)-1creIL-6, and tumor necrosis factor-α and decreased the expression levels of brain-derived neurotrophic factor, tyrosine kinase receptor B, postsynaptic density protein-95, and synaptophysin in the hippocampus. Furthermore, Mel attenuated cognitive impairment and restored markers of inflammation and synaptic plasticity to control levels. CONCLUSIONS: These findings indicated that Mel could ameliorate learning and memory impairment induced by MSD, and these beneficial effects were related to improvement in inflammation and synaptic dysfunction.


Subject(s)
Hippocampus , Melatonin , Memory Disorders , Neuronal Plasticity , Sleep Deprivation , Animals , Melatonin/pharmacology , Melatonin/administration & dosage , Sleep Deprivation/complications , Sleep Deprivation/drug therapy , Sleep Deprivation/physiopathology , Mice , Male , Hippocampus/metabolism , Hippocampus/drug effects , Female , Memory Disorders/drug therapy , Memory Disorders/etiology , Memory Disorders/physiopathology , Neuronal Plasticity/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Pregnancy , Maternal Deprivation , Cognitive Dysfunction/etiology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Neuroinflammatory Diseases/drug therapy
13.
Neurosciences (Riyadh) ; 29(2): 103-112, 2024 May.
Article in English | MEDLINE | ID: mdl-38740397

ABSTRACT

OBJECTIVES: To investigate the fundamental mechanisms of the neuroprotective impact of Astaxanthin (AST) in a mouse model of Alzheimer's disease (AD) induced by scopolamine. METHODS: This research constituted an in vivo animal study encompassing 36 adult male mice, divided into 6 groups: Control, 100 mg/kg AST, 2 mg/kg scopolamine (AD group), 100 mg/kg AST+2 mg/kg scopolamine, 3 mg/kg galantamine+2 mg/kg scopolamine, and 100 mg/kg AST+3 mg/kg galantamine+2 mg/kg scopolamine. After 14 days, the mice's short-term memory, hippocampus tissue, oxidative and inflammatory markers were evaluated. RESULTS: The AST demonstrated a beneficial influence on short-term memory and a reduction in acetylcholinesterase activity in the brain. It exhibited neuroprotective and anti-amyloidogenic properties, significantly decreased pro-inflammatory markers and oxidative stress, and reversed the decline of the Akt-1 and phosphorylated Akt pathway, a crucial regulator of abnormal tau. Furthermore, AST enhanced the effect of galantamine in reducing inflammation and oxidative stress. CONCLUSION: The findings indicate that AST may offer therapeutic benefits against cognitive dysfunction in AD. This is attributed to its ability to reduce oxidative stress, control neuroinflammation, and enhance Akt-1 and pAkt levels, thereby underscoring its potential in AD treatment strategies.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Neuroprotective Agents , Oxidative Stress , Scopolamine , Xanthophylls , Animals , Xanthophylls/pharmacology , Xanthophylls/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/chemically induced , Male , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Acetylcholinesterase/metabolism , Galantamine/pharmacology , Galantamine/therapeutic use , Memory, Short-Term/drug effects
14.
Cell Commun Signal ; 22(1): 269, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745240

ABSTRACT

BACKGROUND: The pathway involving PTEN-induced putative kinase 1 (PINK1) and PARKIN plays a crucial role in mitophagy, a process activated by artesunate (ART). We propose that patients with anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis exhibit insufficient mitophagy, and ART enhances mitophagy via the PINK1/PARKIN pathway, thereby providing neuroprotection. METHODS: Adult female mice aged 8-10 weeks were selected to create a passive transfer model of anti-NMDAR encephalitis. We conducted behavioral tests on these mice within a set timeframe. Techniques such as immunohistochemistry, immunofluorescence, and western blotting were employed to assess markers including PINK1, PARKIN, LC3B, p62, caspase3, and cleaved caspase3. The TUNEL assay was utilized to detect neuronal apoptosis, while transmission electron microscopy (TEM) was used to examine mitochondrial autophagosomes. Primary hippocampal neurons were cultured, treated, and then analyzed through immunofluorescence for mtDNA, mtROS, TMRM. RESULTS: In comparison to the control group, mitophagy levels in the experimental group were not significantly altered, yet there was a notable increase in apoptotic neurons. Furthermore, markers indicative of mitochondrial leakage and damage were found to be elevated in the experimental group compared to the control group, but these markers showed improvement following ART treatment. ART was effective in activating the PINK1/PARKIN pathway, enhancing mitophagy, and diminishing neuronal apoptosis. Behavioral assessments revealed that ART ameliorated symptoms in mice with anti-NMDAR encephalitis in the passive transfer model (PTM). The knockdown of PINK1 led to a reduction in mitophagy levels, and subsequent ART intervention did not alleviate symptoms in the anti-NMDAR encephalitis PTM mice, indicating that ART's therapeutic efficacy is mediated through the activation of the PINK1/PARKIN pathway. CONCLUSIONS: At the onset of anti-NMDAR encephalitis, mitochondrial damage is observed; however, this damage is mitigated by the activation of mitophagy via the PINK1/PARKIN pathway. This regulatory feedback mechanism facilitates the removal of damaged mitochondria, prevents neuronal apoptosis, and consequently safeguards neural tissue. ART activates the PINK1/PARKIN pathway to enhance mitophagy, thereby exerting neuroprotective effects and may achieve therapeutic goals in treating anti-NMDAR encephalitis.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis , Artesunate , Disease Models, Animal , Neuroprotective Agents , Protein Kinases , Animals , Artesunate/pharmacology , Artesunate/therapeutic use , Mice , Female , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/pathology , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/drug therapy , Protein Kinases/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Microscopy, Electron, Transmission , Mitophagy/drug effects , Apoptosis/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Hippocampus/pathology , Hippocampus/drug effects , Hippocampus/metabolism
15.
PLoS One ; 19(5): e0302850, 2024.
Article in English | MEDLINE | ID: mdl-38748711

ABSTRACT

BACKGROUND AND AIM: Vascular dementia (VD) is a common type of dementia. This study aimed to evaluate the effects of low and high doses of lutein administration in bilateral-carotid vessel occlusion (2VO) rats. EXPERIMENTAL PROCEDURE: The rats were divided into the following groups: the control, sham-, vehicle (2VO+V) groups, and two groups after 2VO were treated with lutein 0.5 (2VO+LUT-o.5) and 5mg/kg (2VO+LUT-5). The passive-avoidance and Morris water maze were performed to examine fear and spatial memory. The field-potential recording was used to investigate the properties of basal synaptic transmission (BST), paired-pulse ratio (PPR), as an index for measurement of neurotransmitter release, and long-term potentiation (LTP). The hippocampus was removed to evaluate hippocampal cells, volume, and MDA level. RESULT: Treatment with low and high doses improves spatial memory and LTP impairment in VD rats, but only the high dose restores the fear memory, hippocampal cell loss, and volume and MDA level. Interestingly, low-dose, but not high-dose, increased PPR. However, BST recovered only in the high-dose treated group. CONCLUSIONS: Treatment with a low dose might affect neurotransmitter release probability, but a high dose affects postsynaptic processes. It seems likely that low and high doses improve memory and LTP through different mechanisms.


Subject(s)
Dementia, Vascular , Disease Models, Animal , Hippocampus , Long-Term Potentiation , Lutein , Neuronal Plasticity , Animals , Dementia, Vascular/drug therapy , Dementia, Vascular/physiopathology , Rats , Male , Neuronal Plasticity/drug effects , Long-Term Potentiation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Lutein/pharmacology , Lutein/administration & dosage , Lutein/therapeutic use , Memory/drug effects , Rats, Wistar , Spatial Memory/drug effects , Dose-Response Relationship, Drug , Maze Learning/drug effects , Synaptic Transmission/drug effects
16.
Alzheimers Res Ther ; 16(1): 109, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750512

ABSTRACT

BACKGROUND: As one major symptom of Alzheimer's disease (AD), anterograde amnesia describes patients with an inability in new memory formation. The crucial role of the entorhinal cortex in forming new memories has been well established, and the neuropeptide cholecystokinin (CCK) is reported to be released from the entorhinal cortex to enable neocortical associated memory and long-term potentiation. Though several studies reveal that the entorhinal cortex and CCK are related to AD, it is less well studied. It is unclear whether CCK is a good biomarker or further a great drug candidate for AD. METHODS: mRNA expressions of CCK and CCK-B receptor (CCKBR) were examined in two mouse models, 3xTg AD and CCK knock-out (CCK-/-) mice. Animals' cognition was investigated with Morris water maze, novel object recognition test and neuroplasticity with in-vitro electrophysiological recording. Drugs were given intraperitoneally to animals to investigate the rescue effects on cognitive deficits, or applied to brain slices directly to explore the influence in inducement of long-term potentiation. RESULTS: Aged 3xTg AD mice exhibited reduced CCK mRNA expression in the entorhinal cortex but reduced CCKBR expression in the neocortex and hippocampus, and impaired cognition and neuroplasticity comparable with CCK-/- mice. Importantly, the animals displayed improved performance and enhanced long-term potentiation after the treatment of CCKBR agonists. CONCLUSIONS: Here we provide more evidence to support the role of CCK in learning and memory and its potential to treat AD. We elaborated on the rescue effect of a promising novel drug, HT-267, on aged 3xTg AD mice. Although the physiological etiology of CCK in AD still needs to be further investigated, this study sheds light on a potential pharmaceutical candidate for AD and dementia.


Subject(s)
Alzheimer Disease , Amnesia, Anterograde , Cholecystokinin , Disease Models, Animal , Mice, Transgenic , Receptor, Cholecystokinin B , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Mice , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/deficiency , Amnesia, Anterograde/drug therapy , Cholecystokinin/metabolism , Entorhinal Cortex/drug effects , Entorhinal Cortex/metabolism , Male , Mice, Knockout , Mice, Inbred C57BL , Long-Term Potentiation/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Aging/drug effects
17.
Nutrients ; 16(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38794753

ABSTRACT

Recent studies have indicated that fucoidan has the potential to improve cognitive impairment. The objective of this study was to demonstrate the protective effect and possible mechanisms of fucoidan in D-galactose (D-gal)-induced cognitive dysfunction. Sprague Dawley rats were injected with D-galactose (200 mg/kg, sc) and administrated with fucoidan (100 mg/kg or 200 mg/kg, ig) for 8 weeks. Our results suggested that fucoidan significantly ameliorated cognitive impairment in D-gal-exposed rats and reversed histopathological changes in the hippocampus. Fucoidan reduced D-gal-induced oxidative stress, declined the inflammation level and improved mitochondrial dysfunction in hippocampal. Fucoidan promoted mitochondrial biogenesis by regulating the PGC-1α/NRF1/TFAM pathway, thereby improving D-gal-induced mitochondrial dysfunction. The regulation effect of fucoidan on PGC-1α is linked to the upstream protein of APN/AMPK/SIRT1. Additionally, the neuroprotective action of fucoidan could be related to maintaining intestinal flora homeostasis with up-regulation of Bacteroidota, Muribaculaceae and Akkermansia and down-regulation of Firmicutes. In summary, fucoidan may be a natural, promising candidate active ingredient for age-related cognitive impairment interventions.


Subject(s)
Cognitive Dysfunction , Galactose , Gastrointestinal Microbiome , Hippocampus , Homeostasis , Mitochondria , Organelle Biogenesis , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Polysaccharides , Rats, Sprague-Dawley , Polysaccharides/pharmacology , Animals , Gastrointestinal Microbiome/drug effects , Cognitive Dysfunction/drug therapy , Homeostasis/drug effects , Male , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Neuroprotective Agents/pharmacology , Sirtuin 1/metabolism , Disease Models, Animal , Transcription Factors
18.
J Neuroimmune Pharmacol ; 19(1): 23, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38775885

ABSTRACT

Hyperbilirubinemia is one of the most common occurrence in newborns and is toxic to the brain, resulting in neurological sequelae such as auditory impairment, with potential to evolve to chronic bilirubin encephalopathy and long-term cognitive impairment in adults. In the early postnatal period, neurogenesis is rigorous and neuroinflammation is detrimental to the brain. What are the alterations in neurogenesis and the underlying mechanisms of bilirubin encephalopathy during the early postnatal period? This study found that, there were a reduction in the number of neuronal stem/progenitor cells, an increase in microglia in the dentate gyrus (DG) and an inflammatory state in the hippocampus, characterized by increased levels of IL-6, TNF-α, and IL-1ß, as well as a decreased level of IL-10 in a rat model of bilirubin encephalopathy (BE). Furthermore, there was a significant decrease in the number of newborn neurons and the expression of neuronal differentiation-associated genes (NeuroD and Ascl1) in the BE group. Additionally, cognitive impairment was observed in this group. The administration of minocycline, an inhibitor of microglial activation, resulted in a reduction of inflammation in the hippocampus, an enhancement of neurogenesis, an increase in the expression of neuron-related genes (NeuroD and Ascl1), and an improvement in cognitive function in the BE group. These results demonstrate that microglia play a critical role in reduced neurogenesis and impaired brain function resulting from bilirubin encephalopathy model, which could inspire the development of novel pharmaceutical and therapeutic strategies.


Subject(s)
Hippocampus , Kernicterus , Microglia , Minocycline , Neurogenesis , Animals , Neurogenesis/drug effects , Neurogenesis/physiology , Microglia/drug effects , Microglia/metabolism , Rats , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Minocycline/pharmacology , Disease Models, Animal , Rats, Sprague-Dawley , Inflammation/metabolism , Inflammation/pathology , Neuroinflammatory Diseases/drug therapy
19.
CNS Neurosci Ther ; 30(5): e14719, 2024 May.
Article in English | MEDLINE | ID: mdl-38783536

ABSTRACT

BACKGROUND: Methamphetamine (METH) is a psychostimulant substance with highly addictive and neurotoxic effects, but no ideal treatment option exists to improve METH-induced neurocognitive deficits. Recently, mesenchymal stem cells (MSCs)-derived exosomes have raised many hopes for treating neurodegenerative sequela of brain disorders. This study aimed to determine the therapeutic potential of MSCs-derived exosomes on cognitive function and neurogenesis of METH-addicted rodents. METHODS: Male BALB/c mice were subjected to chronic METH addiction, followed by intravenous administration of bone marrow MSCs-derived exosomes. Then, the spatial memory and recognition memory of animals were assessed by the Barnes maze and the novel object recognition test (NORT). The neurogenesis-related factors, including NeuN and DCX, and the expression of Iba-1, a microglial activation marker, were assessed in the hippocampus by immunofluorescence staining. Also, the expression of inflammatory cytokines, including TNF-α and NF-κB, were evaluated by western blotting. RESULTS: The results showed that BMSCs-exosomes improved the time spent in the target quadrant and correct-to-wrong relative time in the Barnes maze. Also, NORT's discrimination index (DI) and recognition index (RI) were improved following exosome therapy. Additionally, exosome therapy significantly increased the expression of NeuN and DCX in the hippocampus while decreasing the expression of inflammatory cytokines, including TNF-α and NF-κB. Besides, BMSC-exosomes down-regulated the expression of Iba-1. CONCLUSION: Our findings indicate that BMSC-exosomes mitigated METH-caused cognitive dysfunction by improving neurogenesis and inhibiting neuroinflammation in the hippocampus.


Subject(s)
Amphetamine-Related Disorders , Doublecortin Protein , Exosomes , Hippocampus , Mesenchymal Stem Cells , Methamphetamine , Mice, Inbred BALB C , Neurogenesis , Animals , Exosomes/metabolism , Male , Neurogenesis/drug effects , Neurogenesis/physiology , Mice , Methamphetamine/toxicity , Amphetamine-Related Disorders/therapy , Amphetamine-Related Disorders/psychology , Amphetamine-Related Disorders/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Cognition/drug effects , Cognition/physiology , Maze Learning/drug effects , Maze Learning/physiology , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Nerve Tissue Proteins/metabolism , Central Nervous System Stimulants/toxicity , Spatial Memory/drug effects , Spatial Memory/physiology , Microfilament Proteins/metabolism , Mesenchymal Stem Cell Transplantation/methods , Calcium-Binding Proteins , DNA-Binding Proteins
20.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791161

ABSTRACT

Adult neurogenesis in the dentate gyrus (DG) is impaired during Alzheimer's disease (AD) progression. Curcumin has been reported to reduce cell apoptosis and stimulate neurogenesis. This study aimed to investigate the influence of curcumin on adult neurogenesis in AD mice and its potential mechanism. Two-month-old male C57BL/6J mice were injected with soluble ß-amyloid (Aß1-42) using lateral ventricle stereolocalization to establish AD models. An immunofluorescence assay, including bromodeoxyuridine (BrdU), doublecortin (DCX), and neuron-specific nuclear antigen (NeuN), was used to detect hippocampal neurogenesis. Western blot and an enzyme-linked immunosorbent assay (ELISA) were used to test the expression of related proteins and the secretion of brain-derived neurotrophic factor (BDNF). A Morris water maze was used to detect the cognitive function of the mice. Our results showed that curcumin administration (100 mg/kg) rescued the impaired neurogenesis of Aß1-42 mice, shown as enhanced BrdU+/DCX+ and BrdU+/NeuN+ cells in DG. In addition, curcumin regulated the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt) -mediated glycogen synthase kinase-3ß (GSK3ß) /Wingless/Integrated (Wnt)/ß-catenin pathway and cyclic adenosine monophosphate response element-binding protein (CREB)/BDNF in Aß1-42 mice. Inhibiting Wnt/ß-catenin and depriving BDNF could reverse both the upregulated neurogenesis and cognitive function of curcumin-treated Aß1-42 mice. In conclusion, our study indicates that curcumin, through targeting PI3K/Akt, regulates GSK3ß/Wnt/ß-catenin and CREB/BDNF pathways, improving the adult neurogenesis of AD mice.


Subject(s)
Alzheimer Disease , Brain-Derived Neurotrophic Factor , Curcumin , Disease Models, Animal , Doublecortin Protein , Mice, Inbred C57BL , Neurogenesis , Wnt Signaling Pathway , beta Catenin , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Neurogenesis/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Curcumin/pharmacology , Mice , Male , Wnt Signaling Pathway/drug effects , Doublecortin Protein/metabolism , beta Catenin/metabolism , Amyloid beta-Peptides/metabolism , Up-Regulation/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL