ABSTRACT
Upregulation of genes and coexpression networks related to immune function and inflammation have been repeatedly reported in the brain of individuals with schizophrenia. However, a causal relationship between the abnormal immune/inflammation-related gene expression and schizophrenia has not been determined. We conducted co-expression networks using publicly available RNA-seq data from prefrontal cortex (PFC) and hippocampus (HP) of 64 individuals with schizophrenia and 64 unaffected controls from the SMRI tissue collections. We identified proinflammatory cytokine, transmembrane tumor necrosis factor-α (tmTNFα), as a potential regulator in the module of co-expressed genes that we find related to the immune/inflammation response in endothelial cells (ECs) and/or microglia of the brain of individuals with schizophrenia. The immune/inflammation-related modules associated with schizophrenia and the TNF signaling pathway that regulate the network were replicated in an independent cohort of brain samples from 68 individuals with schizophrenia and 135 unaffected controls. To investigate the association between the overexpression of tmTNFα in brain ECs and schizophrenia-like behaviors, we induced short-term overexpression of the uncleavable form of (uc)-tmTNFα in ECs of mouse brain for 7 weeks. We found schizophrenia-relevant behavioral deficits in these mice, including cognitive impairment, abnormal sensorimotor gating, and sensitization to methamphetamine (METH) induced locomotor activity and METH-induced neurotransmitter levels. These uc-tmTNFα effects were mediated by TNF receptor2 (TNFR2) and induced activation of TNFR2 signaling in astrocytes and neurons. A neuronal module including neurotransmitter signaling pathways was down-regulated in the brain of mice by the short-term overexpression of the gene, while an immune/inflammation-related module was up-regulated in the brain of mice after long-term expression of 22 weeks. Our results indicate that tmTNFα may play a direct role in regulating neurotransmitter signaling pathways that contribute to the clinical features of schizophrenia.
Subject(s)
Methamphetamine , Schizophrenia , Mice , Animals , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Schizophrenia/metabolism , Endothelial Cells/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Brain/metabolism , Inflammation/geneticsABSTRACT
The simultaneous abuse of alcohol-cocaine is known to cause stronger and more unpredictable cellular damage in the liver, heart, and brain. However, the mechanistic crosstalk between cocaine and alcohol in liver injury remains unclear. The findings revealed cocaine-induced liver injury and inflammation in both marmosets and mice. Of note, co-administration of cocaine and ethanol in mice causes more severe liver damage than individual treatment. The metabolomic analysis confirmed that hippuric acid (HA) is the most abundant metabolite in marmoset serum after cocaine consumption and that is formed in primary marmoset hepatocytes. HA, a metabolite of cocaine, increases mitochondrial DNA leakage and subsequently increases the production of proinflammatory factors via STING signaling in Kupffer cells (KCs). In addition, conditioned media of cocaine-treated KC induced hepatocellular necrosis via alcohol-induced TNFR1. Finally, disruption of STING signaling in vivo ameliorated co-administration of alcohol- and cocaine-induced liver damage and inflammation. These findings postulate intervention of HA-STING-TNFR1 axis as a novel strategy for treatment of alcohol- and cocaine-induced excessive liver damage.
Subject(s)
Cocaine , DNA, Mitochondrial , Hippurates , Liver Diseases, Alcoholic , Membrane Proteins , Signal Transduction , Animals , Cocaine/pharmacology , Cocaine/toxicity , Signal Transduction/drug effects , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/drug effects , Mice , Hippurates/metabolism , Male , Membrane Proteins/metabolism , Hepatocytes/metabolism , Hepatocytes/drug effects , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Ethanol/toxicity , Mice, Inbred C57BL , Cocaine-Related Disorders/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolismABSTRACT
Several clinical studies reported that the elevated expression of Chitinase-3-like 1 (CHI3L1) was observed in patients suffering from a wide range of diseases: cancer, metabolic, and neurological diseases. However, the role of CHI3L1 in AD is still unclear. Our previous study demonstrated that 2-({3-[2-(1-Cyclohexen-1-yl)ethyl]-6,7-dimethoxy-4-oxo-3,4-dihydro-2-quinazolinyl}culfanyl)-N-(4-ethylphenyl)butanamide, a CHI3L1 inhibiting compound, alleviates memory and cognitive impairment and inhibits neuroinflammation in AD mouse models. In this study, we studied the detailed correlation of CHI3L1 and AD using serum from AD patients and using CHI3L1 knockout (KO) mice with Aß infusion (300 pmol/day, 14 days). Serum levels of CHI3L1 were significantly elevated in patients with AD compared to normal subjects, and receiver operating characteristic (ROC) analysis data based on serum analysis suggested that CHI3L1 could be a significant diagnostic reference for AD. To reveal the role of CHI3L1 in AD, we investigated the CHI3L1 deficiency effect on memory impairment in Aß-infused mice and microglial BV-2 cells. In CHI3L1 KO mice, Aß infusion resulted in lower levels of memory dysfunction and neuroinflammation compared to that of WT mice. CHI3L1 deficiency selectively inhibited phosphorylation of ERK and IκB as well as inhibition of neuroinflammation-related factors in vivo and in vitro. On the other hand, treatment with recombinant CHI3L1 increased neuroinflammation-related factors and promoted phosphorylation of IκB except for ERK in vitro. Web-based gene network analysis and our results showed that CHI3L1 is closely correlated with PTX3. Moreover, in AD patients, we found that serum levels of PTX3 were correlated with serum levels of CHI3L1 by Spearman correlation analysis. These results suggest that CHI3L1 deficiency could inhibit AD development by blocking the ERK-dependent PTX3 pathway.
Subject(s)
Alzheimer Disease , Chitinase-3-Like Protein 1 , Humans , Mice , Quinazolines/administration & dosage , Disease Models, Animal , Alzheimer Disease/blood , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/pathology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Microglia/metabolism , Microglia/pathology , Serum Amyloid P-Component/metabolism , C-Reactive Protein/metabolism , Chitinase-3-Like Protein 1/blood , Chitinase-3-Like Protein 1/genetics , Biomarkers/bloodABSTRACT
Abuse of new psychoactive substances is an emerging social problem. Several phenethylamines are internationally controlled substances as they are likely to be abused and have adverse effects. Phenethylamine analog 2-(4-iodo-2,5-dimethoxyphenyl)-N-(2-methoxybenzyl)ethanamine (25I-NBOMe) has been reported as one of the most commonly abused psychoactive substance. However, the cardiotoxicity of this compound has not been extensively evaluated. Thus, in this study, we investigated the adverse cardiovascular effects of 25I-NBOMe, related to p21 (CDC42/RAC)-activated kinase 1 (PAK1). The cardiotoxicity of 25I-NBOMe was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, live/dead cytotoxicity assay, PAK1/CDC42 kinase assay, and in vivo electrocardiography (ECG). Also, we analyzed the expression level of PAK1, which is known to play key roles in the cardiovascular system. In the MTT assay, cell viability of 25I-NBOMe-treated H9c2 cells or primary cardiomyocytes of ICR mice decreased in a concentration-dependent manner. Results from the in vitro cytotoxicity assay in cardiomyocytes showed that 25I-NBOMe decreased the viability of H9c2 rat cardiomyocytes, and TC50 of 25I-NBOMe was found to be 70.4 µM. We also observed that 25I-NBOMe reduced PAK1 activity in vitro. Surface ECG measurement revealed that intravenous injection of 25I-NBOMe (doses of 1.0 and 3.0 mg/kg, corresponding to serum concentrations of 18.1 and 28.6 ng/mL, respectively) prolonged the QTc interval in SD rats. Furthermore, treatment with 25I-NBOMe downregulated the expression of PAK1 in the hearts of SD rats and H9c2 cells. In summary, our findings indicate that PAK1-related adverse effects of 25I-NBOMe can cause toxicity to cardiomyocytes and induce an abnormal ECG pattern in animals.
Subject(s)
Dimethoxyphenylethylamine , Rodentia , Animals , Dimethoxyphenylethylamine/analogs & derivatives , Dimethoxyphenylethylamine/pharmacology , Mice , Mice, Inbred ICR , Myocytes, Cardiac , Phenethylamines/toxicity , Rats , Rats, Sprague-DawleyABSTRACT
Our previous big data analyses reported a strong association between CHI3L1 expression and lung tumor development. In this present study, we investigated whether a CHI3L1-inhibiting natural compound, ebractenoid F, inhibits lung cancer cell growth and migration and induces apoptosis. Ebractenoid F concentration-dependently (0, 17, 35, 70 µM) and significantly inhibited the proliferation and migration of A549 and H460 lung cancer cells and induced apoptosis. In the mechanism study, we found that ebractenoid F bound to CHI3L1 and suppressed CHI3L1-associated AKT signaling. Combined treatment with an AKT inhibitor, LY294002, and ebractenoid F synergistically decreased the expression of CHI3L1. Moreover, the combination treatment further inhibited the growth and migration of lung cancer cells and further induced apoptosis, as well as the expression levels of apoptosis-related proteins. Thus, our data demonstrate that ebractenoid F may serve as a potential anti-lung cancer compound targeting CHI3L1-associated AKT signaling.
Subject(s)
Lung Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor , Cell Proliferation , Lung Neoplasms/metabolism , Apoptosis , Chitinase-3-Like Protein 1ABSTRACT
Acupuncture affects the central nervous system via the regulation of neurotransmitter transmission. We previously showed that Shemen (HT7) acupoint stimulation decreased cocaine-induced dopamine release in the nucleus accumbens. Here, we used the intracranial self-stimulation (ICSS) paradigm to evaluate whether HT stimulation regulates the brain reward function of rats. We found that HT stimulation triggered a rightward shift of the frequency-rate curve and elevated the ICSS thresholds. However, HT7 stimulation did not affect the threshold-lowering effects produced by cocaine. These results indicate that HT7 points only effectively regulates the ICSS thresholds of the medial forebrain bundle in drug-naïve rats.
Subject(s)
Acupuncture Therapy/methods , Cocaine/administration & dosage , Electric Stimulation/methods , Medial Forebrain Bundle/physiology , Reward , Self Stimulation/physiology , Anesthetics, Local/administration & dosage , Animals , Male , Medial Forebrain Bundle/drug effects , Rats , Rats, Sprague-Dawley , Self Stimulation/drug effectsABSTRACT
Interaction of cannabinoid receptor type 1 (CB1) and GABAergic neuronal activity is involved in drug abuse-related behavior. However, its role in drug-dependent Pavlovian conditioning is not well understood. In this study, we aimed to evaluate the effects of a CB1 agonist, JWH-210, on the development of conditioned place preference (CPP)-induced by methamphetamine (METH). Pretreatment with a synthetic cannabinoid, JWH-210 (CB1 agonist), increased METH-induced CPP score and METH-induced dopamine release in acute striatal slices. Interestingly, CB1 was expressed in glutamate decarboxylase 67 (GAD67) positive cells, and overexpression of CB1 increased GAD67 expression, while CB1 knockdown reduced GAD67 expression in vivo and in vitro. GAD67 is known as an enzyme involved in the synthesis of GABA. CB1 knockdown in the mice striatum increased METH-induced CPP. When GAD67 decreased in the mice striatum, mRNA level of CB1 did not change, suggesting that CB1 can regulate GAD67 expression. GAD67 knockdown in the mouse striatum augmented apomorphine (dopamine receptor D2 agonist)-induced climbing behavior and METH-induced CPP score. Moreover, in the human brain, mRNA level of GAD67 was found to be decreased in drug users. Therefore, we suggest that CB1 potentiates METH-induced CPP through inhibitory GABAergic regulation of dopaminergic neuronal activity.
Subject(s)
Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Gene Expression Regulation, Enzymologic , Glutamate Decarboxylase/biosynthesis , Receptor, Cannabinoid, CB1/metabolism , Substance-Related Disorders/metabolism , Animals , Apomorphine/pharmacology , Gene Knockdown Techniques , Glutamate Decarboxylase/genetics , Humans , Indoles/pharmacology , Male , Methamphetamine/pharmacology , Mice , Naphthalenes/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/geneticsABSTRACT
BACKGROUND: Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders characterized by gradual memory loss and neuropsychiatric symptoms. We have previously demonstrated that the 2-({3-[2-(1-cyclohexene-1-yl)ethyl]-6,7-dimethoxy-4-oxo-3,4-dihydro-2-quinazolinyl}sulfanyl)-N-(4-ethylphenyl)butanamide (K284-6111), the inhibitor of CHI3L1, has the inhibitory effect on memory impairment in Αß infusion mouse model and on LPS-induced neuroinflammation in the murine BV-2 microglia and primary cultured astrocyte. METHODS: In the present study, we investigated the inhibitory effect of K284-6111 on memory dysfunction and neuroinflammation in Tg2576 transgenic mice, and a more detailed correlation of CHI3L1 and AD. To investigate the effects of K284-6111 on memory dysfunction, we administered K284-6111 (3 mg/kg, p.o.) daily for 4 weeks to Tg2576 mice, followed by behavioral tests of water maze test, probe test, and passive avoidance test. RESULTS: Administration of K284-6111 alleviated memory impairment in Tg2576 mice and had the effect of reducing the accumulation of Aß and neuroinflammatory responses in the mouse brain. K284-6111 treatment also selectively inactivated ERK and NF-κB pathways, which were activated when CHI3L1 was overexpressed, in the mouse brain and in BV-2 cells. Web-based gene network analysis and our results of gene expression level in BV-2 cells showed that CHI3L1 is closely correlated with PTX3. Our result revealed that knockdown of PTX3 has an inhibitory effect on the production of inflammatory proteins and cytokines, and on the phosphorylation of ERK and IκBα. CONCLUSION: These results suggest that K284-6111 could improve memory dysfunction by alleviating neuroinflammation through inhibiting CHI3L1 enhancing ERK-dependent PTX3 pathway.
Subject(s)
C-Reactive Protein/deficiency , Chitinase-3-Like Protein 1/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Memory Disorders/drug therapy , Nerve Tissue Proteins/deficiency , Quinazolines/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , C-Reactive Protein/genetics , Cell Line , Chitinase-3-Like Protein 1/metabolism , Gene Knockdown Techniques/methods , Inflammation Mediators/metabolism , MAP Kinase Signaling System/physiology , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Quinazolines/pharmacologyABSTRACT
BACKGROUND: Epilepsy is a chronic neurological disorder characterized by the recurrence of seizures. One-third of patients with epilepsy may not respond to antiseizure drugs. PURPOSE: We aimed to examine whether D-limonene, a cyclic monoterpene, exhibited any antiseizure activity in the pentylenetetrazole (PTZ)-induced kindling mouse model and in vitro. METHODS: PTZ kindling mouse model was established by administering PTZ (30 mg/kg) intraperitoneally to mice once every 48 h. We performed immunoblot blots, immunohistochemistry (IHC), and high-performance liquid chromatography (HPLC) analysis after the behavioral study. RESULTS: An acute injection of PTZ (60 mg/kg) induced seizure in mice, while pretreatment with D-limonene inhibited PTZ-induced seizure. Repeated administration of PTZ (30 mg/kg) increased the seizure score gradually in mice, which was reduced in D-limonene (10 mg/kg)-pretreated group. In addition, D-limonene treatment increased glutamate decarboxylase-67 (GAD-67) expression in the hippocampus. Axonal sprouting of hippocampal neurons after kindling was inhibited by D-limonene pretreatment. Moreover, D-limonene reduced the expression levels of Neuronal PAS Domain Protein 4 (Npas4)-induced by PTZ. Furthermore, the adenosine A2A antagonist SCH58261 and ZM241385 inhibited anticonvulsant activity and gamma-aminobutyric acid (GABA)ergic neurotransmission-induced by D-limonene. CONCLUSION: These results suggest that D-limonene exhibits anticonvulsant activity through modulation of adenosine A2A receptors on GABAergic neuronal function.
Subject(s)
GABAergic Neurons/metabolism , Limonene/pharmacology , Pentylenetetrazole/adverse effects , Receptor, Adenosine A2A/metabolism , Seizures/etiology , Seizures/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Convulsants/administration & dosage , Convulsants/adverse effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Neuronal Plasticity/drug effects , Pentylenetetrazole/administration & dosage , Phosphorylation , Rats , Seizures/physiopathologyABSTRACT
PURPOSE: To investigate the relationship between αB-crystallin levels in the vitreous of patients with rhegmatogenous retinal detachment (RRD) and correlate these levels with the presence of proliferative vitreoretinopathy (PVR) and to the extent of RRD. METHODS: Prospective, cross-sectional study. Vitreous samples were collected from twenty-six patients with RRD at the surgical repair. PVR was evaluated using the Updated Proliferative Vitreoretinopathy Grade Classification system. RRD area was measured using ultra-widefield retinal photographs. The concentration of αB-crystallin in the vitreous was measured using an enzyme-linked immunosorbent assay kit. RESULTS: The average concentration of αB-crystallin was significantly higher in the PVR Grade B and C group (97.83 ± 69.47) than in the PVR (-) and Grade A group (39.43 ± 13.53 ng/mL, P = 0.005). The level of αB-crystallin was also significantly higher in patients with large RD area (RD area > 1/4, 85.52 ± 66.07 ng/mL) than in patients with small RD area (RD area ≤ 1/4, 39.20 ± 13.02 ng/mL, P = 0.016). CONCLUSIONS: The level of αB-crystallin was significantly increased in the vitreous of RRD patients with high-grade PVR and large RD area. This finding suggests that αB-crystallin may function as an endogenous mediator in fibrotic processes in RRD patients.
Subject(s)
Crystallins , Retinal Detachment , Vitreoretinopathy, Proliferative , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Humans , Prospective Studies , Vitreoretinopathy, Proliferative/complications , Vitreoretinopathy, Proliferative/diagnosis , Vitreous BodyABSTRACT
The aim of this study was to investigate whether the glutathione peroxidase-1 gene (GPx-1) affects cocaine-induced conditioned place preference (CPP) using a mouse model. Cocaine-induced CPP was accompanied by an increase in the level of σ-1 receptor in the nucleus accumbens (NAc). This phenomenon was more pronounced in the GPx-1 gene knockout (GPx-1 KO) than in wild type (WT) mice. In contrast, the CPP and expression of σ-1 receptor were much less pronounced in GPx-1-overexpressing transgenic (GPx-1 TG) mice than non-transgenic (non-TG) mice. Treatment of the mice with BD1047, a σ-1 receptor antagonist, significantly attenuated both cocaine-induced CPP and c-Fos-immunoreactivity (c-Fos-IR) in WT and GPx-1 KO mice, although the effects were more evident in the latter group. Despite the protective effects of BD1047 on cocaine-induced CPP and c-Fos in non-TG mice, there were no additional protective effects in cocaine-treated GPx-1 TG mice, indicating that the σ-1 receptor is a critical target for GPx-1-mediated psychoprotective activity. Overall, our results suggest that GPx-1 attenuates cocaine-induced CPP via inhibition of σ-1 receptor expression.
Subject(s)
Behavior, Animal/drug effects , Cocaine/pharmacology , Conditioning, Psychological/drug effects , Gene Expression Regulation/drug effects , Glutathione Peroxidase/genetics , Receptors, sigma/genetics , Animals , Gene Knockout Techniques , Glutathione Peroxidase/deficiency , Mice , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Glutathione Peroxidase GPX1 , Sigma-1 ReceptorABSTRACT
Neuroinflammation is implicated in dopaminergic neurodegeneration. We have previously demonstrated that (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol (MMPP), a selective signal transducer and activator of transcription 3 (STAT3) inhibitor, has anti-inflammatory properties in several inflammatory disease models. We investigated whether MMPP could protect against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic cell loss and behavioral impairment. Imprinting control region (ICR) mice (8 weeks old, n = 10 per group) were administered MMPP (5 mg/kg) in drinking water for 1 month, and injected with MPTP (15 mg/kg, four times with 2 h intervals) during the last 7 days of treatment. MMPP decreased MPTP-induced behavioral impairments in rotarod, pole, and gait tests. We also showed that MMPP ameliorated dopamine depletion in the striatum and inflammatory marker elevation in primary cultured neurons by high-performance liquid chromatography and immunohistochemical analysis. Increased activation of STAT3, p38, and monoamine oxidase B (MAO-B) were observed in the substantia nigra and striatum after MPTP injection, effects that were attenuated by MMPP treatment. Furthermore, MMPP inhibited STAT3 activity and expression of neuroinflammatory proteins, including ionized calcium binding adaptor molecule 1 (Iba1), inducible nitric oxide synthase (iNOS), and glial fibrillary acidic protein (GFAP) in 1-methyl-4-phenylpyridinium (MPP+; 0.5 mM)-treated primary cultured cells. However, mitogen-activated protein kinase (MAPK) inhibitors augmented the activity of MMPP. Collectively, our results suggest that MMPP may be an anti-inflammatory agent that attenuates dopaminergic neurodegeneration and neuroinflammation through MAO-B and MAPK pathway-dependent inhibition of STAT3 activation.
Subject(s)
Dopaminergic Neurons/metabolism , MPTP Poisoning/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cells, Cultured , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine/metabolism , Dopaminergic Neurons/pathology , Glial Fibrillary Acidic Protein/metabolism , Inflammation , MPTP Poisoning/pathology , Male , Mice , Mice, Inbred ICR , Monoamine Oxidase/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolismABSTRACT
Estrogen is well known to have a preventative effect in Alzheimer's disease (AD) pathology. Several studies have demonstrated that nuclear factor kappa-B (NF-ĸB) can contribute to the effects of estrogen on the development of AD. We investigated whether NF-ĸB affects amyloid-beta (Aß)-induced memory impairment in an estrogen-lacking condition. In the present study, nine-week-old Institute cancer research (ICR) mice were ovariectomized to block estrogen stimulation. Ten weeks after the ovariectomization, mice were administered with Aß (300â¯pmol) via intracerebroventricular (ICV) infusion for 2â¯weeks. Memory impairment, neuroinflammatory protein expression, and amyloidogenic pathways were then measured. Ovariectomized mice demonstrated severe memory impairment, Aß accumulation, neprilysin downregulation, and activation of NF-ĸB signaling compared to sham-control mice. In vitro experiments demonstrated that ß-estradiol (10⯵M) inhibited Aß (1⯵M)-induced neuroinflammation in microglial BV-2 cells and prevented Aß-induced cell death in primary cultured neuronal cells. As in in vivo experiments, NF-ĸB activation was significantly upregulated in in vitro experiments. Furthermore ß-estradiol treatment inhibited NF-ĸB activation in both of microglial BV-2 cells and cultured neuronal cells. These findings suggest that estrogen may protect against memory impairment through the regulation of Aß accumulation and neurogenic inflammation by inhibiting NF-κB activity.
Subject(s)
Amyloid beta-Peptides/metabolism , Estrogens/physiology , Memory Disorders/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/physiology , Animals , Astrocytes/metabolism , Cyclooxygenase 2/metabolism , Estradiol/pharmacology , Estrogens/deficiency , Estrogens/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Memory Disorders/physiopathology , Mice , Mice, Inbred ICR , Microglia/metabolism , NF-kappa B/metabolism , Neuroimmunomodulation/immunology , Nitric Oxide Synthase Type II/metabolism , Ovariectomy/methods , Primary Cell Culture , Signal Transduction/drug effectsABSTRACT
Ultraviolet (UV) irradiation damages skin and produces symptoms of photoaging, such as thickening, rough texture, wrinkles, and pigmentation. However, the cellular and molecular mechanisms underlying photoaging induced by chronic UV irradiation are not yet fully understood. Matrix metalloproteinases (MMPs) have been reported to be involved in the response to UV irradiation. In this study, we examined the effects of the sunscreen agent Octylmethoxycinnamate (OMC) on photoaging of the skin induced by chronic UV exposure in hairless albino Crl:SKH1-Hrhr (SKH-1) mice. We demonstrated that the expression of MMPs was elevated by UV irradiation, whereas the topical application of OMC inhibited the upregulation of MMPs. Furthermore, UV-induced wrinkle formation was decreased by OMC treatment. These results suggest that OMC is a potential agent for the prevention and treatment of skin photoaging.
Subject(s)
Cinnamates/pharmacology , Matrix Metalloproteinases/metabolism , Skin Aging/drug effects , Skin/drug effects , Animals , Collagen/metabolism , Elastin/metabolism , Female , Mice, Hairless , Skin/metabolism , Skin/radiation effects , Skin Aging/radiation effects , Sunscreening Agents/pharmacology , Ultraviolet RaysABSTRACT
A number of some chalcone derivatives possess promising biological properties including anti-inflammation, anti-oxidant, and anti-tumor activity. Although it has been shown that some derivatives of chalcone induce apoptosis in different kinds of cancer cells, the involved mechanism of action is not well defined. The purpose of this study is to investigate the primary target of a benzylideneacetophenone derivative (JC3), which is a synthetic compound derived from the chalcone family, in human cancer, using prostate cancer cells as a working model. Herein, we show that JC3 inhibits proteasomal activity as indicated by both in vitro and in cell-based assays. Especially, the JC3-dimer was more potent than monomer in the aspect of proteasome inhibition, which induced apoptosis significantly in the prostate cancer cells. Owing to the critical roles of the proteasome in the biology of human tumor progression, invasion, and metastasis, these findings give an important clue for the development of novel anti-tumor agents.
Subject(s)
Antineoplastic Agents/pharmacology , Chalcone/pharmacology , Propiophenones/pharmacology , Prostatic Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Chalcone/chemistry , Dimerization , Humans , Male , Propiophenones/chemistry , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/pharmacologyABSTRACT
Neuronal Per Arnt Sim domain protein 4 (NPAS4), a brain-specific basic helix-loop-helix transcription factor, has recently been shown to regulate the development of the GABAergic inhibitory synapses and transcription program for contextual memory formation in the hippocampus. We previously reported that chronic social isolation or restriction stress in mice resulted in an impairment in memory and emotional behavior, which was associated with a decrease in Npas4 mRNA levels. In this study, we investigated the role of NPAS4 in neuronal function in vitro and in vivo. Differentiation medium-induced neurite outgrowth was inhibited in Npas4 knockdown Neuro2a cells, whereas overexpression of NPAS4 accelerated the neurite outgrowth in Neuro2a cells. Furthermore, depolarization-induced neurite outgrowth was abolished in Npas4 KO hippocampal neurons. NPAS4 overexpression increased cyclin-dependent kinase 5 (CDK5)-dependent synapsin I phosphorylation in Neuro2a cells and primary cultured hippocampal neurons. A CDK5 inhibitor, roscovitine, inhibited the neurite outgrowth and the increase in phosphorylated synapsin I (p-SYN I) levels in Npas4-overexpressed Neuro2a cells. Interaction of NPAS4 with promoters of Cdk5 and NeuN genes was demonstrated by a chromatin immunoprecipitation assay. In an in vivo study, pentylenetetrazole-induced convulsions in mice resulted in an increase in NPAS4 and p-SYN I levels in the prefrontal cortex of wild-type mice, although no changes in p-SYN I levels were observed in Npas4 knock-out mice. These results suggest that NPAS4 plays an important role in the structural and functional plasticity of neurons.
Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cyclin-Dependent Kinase 5/metabolism , Gene Expression Regulation , Synapsins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin Immunoprecipitation , Hippocampus/metabolism , Mice , Mice, Knockout , Models, Biological , Neurites/metabolism , Neurons/metabolism , Phosphorylation , Protein Structure, Tertiary , RNA, Messenger/metabolismABSTRACT
CONTEXT: Scutellaria baicalensis Georgi (Lamiaceae) has been used as a traditional herbal preparation for the treatment of neuropsychiatric disorders in Asian countries for centuries. OBJECTIVE: To evaluate the effects of S. baicalensis on morphine-induced drug dependence in rats. MATERIALS AND METHODS: In order to evaluate the effect of S. baicalensis and baicalin on morphine-induced dependence-like behavior, a water extract of S. baicalensis [500 mg/kg, intraperitoneally (i.p.)] or baicalin (50 mg/kg, i.p., a flavonoid found in S. baicalensis) was administered prior to morphine injection [5 and 2.5 mg/kg, respectively, subcutaneously (s.c.)] to rats for 8 and 4 d, respectively. Morphine-induced conditioned place preference was assessed by measuring the time spent in a drug-paired chamber. The effect of S. baicalensis on dopamine receptor supersensitivity (locomotor activity) and dopamine agonist-induced climbing behavior due to a single apomorphine treatment (2 mg/kg, s.c.) was also measured. RESULTS: At 50 mg/kg, a water extract of S. baicalensis decreased morphine (5 mg/kg)-induced conditioned place preference by 86% in rats. Apomorphine (2 mg/kg)-induced locomotor activity (dopamine receptor supersensitivity) in rats and climbing behavior in mice were attenuated after pretreatment with 500 mg/kg of S. baicalensis water extract by 41% and 56%, respectively. In addition, baicalin-reduced morphine-induced conditioned places preference by 86% in rats at 50 mg/kg. DISCUSSION AND CONCLUSION: These results suggest that S. baicalensis can ameliorate drug addiction-related behavior through functional regulation of dopamine receptors.
Subject(s)
Conditioning, Operant/drug effects , Morphine Dependence/prevention & control , Morphine/pharmacology , Plant Extracts/pharmacology , Plant Roots , Animals , Conditioning, Operant/physiology , Male , Mice , Mice, Inbred ICR , Morphine Dependence/metabolism , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Dopamine/metabolism , Scutellaria baicalensis , Water/pharmacologyABSTRACT
[This corrects the article DOI: 10.1016/j.jgr.2021.02.001.][This corrects the article DOI: 10.1016/j.jgr.2022.12.004.].
ABSTRACT
Recently, the abuse of synthetic cathinones is increasing among young people. α-Pyrrolidinobutiothiophenone (α-PBT), a synthetic cathinone, is a designer drug that is freely traded online with no legal restrictions. Moreover, there is currently no scientific basis for legal regulation. Here, we examined the addictive properties of α-PBT using a drug discrimination (DD) task. We also investigated the role of α-PBT in brain stimulation reward (BSR) using an intracranial self-stimulation (ICSS) paradigm in rats. Initially, the rats were trained to discriminate between cocaine and saline. After the discrimination training criteria were met, we determined the dose-effect curves of cocaine and conducted generalization tests with α-PBT and α-pyrrolidinopentiothiophenone (α-PVT) using a cumulative dosing protocol. In a separate set of studies, we examined the dopaminergic mechanisms underlying the function of α-PBT as an interoceptive stimulus (17.8 mg/kg) by intraperitoneally injecting either the dopamine (DA) D1 antagonist SCH23390 (0.06 and 0.12 mg/kg) or the D2 antagonist eticlopride (0.05 and 0.1 mg/kg) 15 min before DD testing. Brain reward function was measured using an ICSS procedure to examine the effects of α-PBT on ICSS threshold under the frequency-rate procedure. Our results showed that α-PBT functioned as a discriminative cue similar to cocaine in rats. More importantly, SCH23390 abolished the effects of α-PBT as an interoceptive stimulus in a dose-dependent manner in rats trained to press a lever to receive cocaine. Similarly, eticlopride dose-dependently attenuated the effect of α-PBT used as a discriminative cue. Additionally, cumulative α-PBT administration dose-dependently lowered ICSS thresholds compared with those in saline-treated rats. Furthermore, α-PBT-induced potentiation of BSR was abolished by pretreatment with both SCH23390 and eticlopride. Taken together, our results suggest that α-PBT can function as a cocaine-like discriminative cue via the activation of D1 and D2 receptors. α-PBT also appears to influence BSR by reducing the brain reward threshold via changes in D1 and D2 receptors. The present study suggests that α-PBT could have addictive properties through DA D1 and D2 receptors and thus poses a threat to humans.
Subject(s)
Cocaine , Self Stimulation , Animals , Male , Self Stimulation/drug effects , Rats , Cocaine/pharmacology , Rats, Sprague-Dawley , Pyrrolidines/pharmacology , Reward , Dose-Response Relationship, Drug , Thiophenes/pharmacology , Benzazepines/pharmacology , Designer Drugs/pharmacology , Discrimination, Psychological/drug effects , Brain/drug effects , Brain/metabolismABSTRACT
IMPORTANCE: Glutamic acid decarboxylase 67 (GAD67) is a gamma-aminobutyric acid (GABA) synthesis enzyme associated with the function of other neurotransmitter receptors, such as the N-methyl-D-aspartate (NMDA) receptor and cannabinoid receptor 1. However, the role of GAD67 in the development of different abused drug-induced reward behaviors remains unknown. In order to elucidate the mechanisms of substance use disorder, it is crucial to study changes in biomarkers within the brain's reward circuit induced by drug use. OBJECTIVE: The study was designed to examine the effects of the downregulation of GAD67 expression in the dorsal striatum on reward behavior development. METHODS: We evaluated the effects of GAD67 knockdown on depression-like behavior and anxiety using the forced swim test and elevated plus maze test in a mouse model. We further determined the effects of GAD67 knockdown on ketamine- and JWH-018-induced conditioned place preference (CPP). RESULTS: Knockdown of GAD67 in the dorsal striatum of mice increased depression-like behavior, but it decreased anxiety. Moreover, the CPP score on the NMDA receptor antagonist ketamine was increased by GAD67 knockdown, whereas the administration of JWH-018, a cannabinoid receptor agonist, did not affect the CPP score in the GAD67 knockdown mice group compared with the control group. CONCLUSIONS AND RELEVANCE: These results suggest that striatal GAD67 reduces GABAergic neuronal activity and may cause ketamine-induced NMDA receptor inhibition. Consequently, GAD67 downregulation induces vulnerability to the drug reward behavior of ketamine.