Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cytokine ; 174: 156439, 2024 02.
Article in English | MEDLINE | ID: mdl-38134557

ABSTRACT

Neuregulin (NRG)-1 plays fundamental roles in several organ systems after binding to its receptors, ErbB2 and ErbB4. This study examines the role of NRG-1 in atopic dermatitis (AD), a chronic skin disease that causes dryness, pruritus, and inflammation. In mice administered Der p 38, the skin presents AD-like symptoms including filaggrin downregulation and infiltration of neutrophils and eosinophils. Noticeably, there is an increased expression of NRG-1, ErbB2, and ErbB4 in the skin. Upregulation of these proteins is significantly correlated to the clinical skin severity score. In human keratinocyte HaCaT cells, exposure to Der p 38 decreased filaggrin expression, and NRG-1 alone had no effect on the expression. However, co-treatment of Der p 38 with NRG-1 enhanced the filaggrin expression decreased by Der p 38. Pre-treatment with AG879 (an ErbB2 inhibitor) or ErbB4 siRNA blocked the recovery of filaggrin expression in the cells after co-treatment with Der p 38 and NRG-1. Der p 38 treatment enhanced the secretion of interleukin-6 (IL-6), IL-8, and monocyte chemoattractant protein-1 (MCP-1). Co-treatment of Der p 38 with NRG-1 lowered the cytokine secretion increased by Der p 38, although NRG-1 alone was not effective on cytokine alteration. Neutrophil apoptosis was not altered by NRG-1 or supernatants of cells treated with NRG-1, but the cell supernatants co-treated with Der p 38 and NRG-1 blocked the anti-apoptotic effects of Der p 38-treated supernatants on neutrophils, which was involved in the activation of caspase 9 and caspase 3. Taken together, we determined that NRG-1 has anti-inflammatory effects in AD triggered by Der p 38. These results will pave the way to understanding the functions of NRG-1 and in the future development of AD treatment.


Subject(s)
Dermatitis, Atopic , Mice , Animals , Humans , Dermatitis, Atopic/genetics , Filaggrin Proteins , Neuregulin-1/pharmacology , Neuregulin-1/metabolism , Neuregulin-1/therapeutic use , Keratinocytes/metabolism , Skin/metabolism , Cytokines/metabolism , Receptor, ErbB-4/metabolism , Receptor, ErbB-4/pharmacology , Anti-Inflammatory Agents/pharmacology
2.
Neurobiol Dis ; 177: 105982, 2023 02.
Article in English | MEDLINE | ID: mdl-36592864

ABSTRACT

Neuregulin-1 (NRG1) is an epidermal growth factor family member with essential roles in the developing and adult nervous systems. In recent years, establishing evidence has collectively suggested that NRG1 is a new modulator of central nervous system (CNS) injury and disease, with multifaceted roles in neuroprotection, remyelination, neuroinflammation, and other repair mechanisms. NRG1 signaling exerts its effects via the tyrosine kinase receptors ErbB2-ErbB4. The NRG1/ErbB network in CNS pathology and repair has evolved, primarily in recent years. In the present study, we demonstrated that a unilateral microinjection of CoCl2 into the ventral hippocampus (vHPC) induced hypoxic insult and led to anxiety-related behaviors and deficit sociability in mice. NRG1 treatment significantly alleviated the CoCl2-induced increase of hypoxic-related molecules and behavioral abnormalities. Furthermore, NRG1 reduced the CoCl2-induced neuroinflammation and neuronal deficits in the vHPC or primary hippocampal neurons in mice. Collectively, these results suggest that NRG1 ameliorates hypoxia by alleviating synaptic deficits and behavioral abnormalities of the CoCl2-induced vHPC hypoxic model.


Subject(s)
Neuregulin-1 , Neuroinflammatory Diseases , Mice , Animals , Neuregulin-1/metabolism , Hippocampus/metabolism , Social Behavior , Anxiety/drug therapy
3.
J Anat ; 241(6): 1477-1488, 2022 12.
Article in English | MEDLINE | ID: mdl-36073345

ABSTRACT

Although the level of neuroscience research is rapidly developing with the introduction of new technologies, the method of neuroanatomy education remains at the traditional level and requires improvement to meet the needs of educators and trainees. We developed a new three-dimensional (3D) printed device (human brain-cutting mold, HBCM) for creating human brain slices; moreover, we demonstrated a simple method for creating semi-permanent ultraviolet (UV) resin-mounted brain slice specimens for neuroanatomy education. We obtained brain slices of uniform thickness (3 mm) through the HBCM; the resultant brain slices were optimal for assessing morphological details of the human brain. Furthermore, we used an agar-embedding method for brain-slicing with the HBCM, which minimized geometrical distortions of the brain slices. Also, we prepared semi-permanent brain serial specimens using an acrylic brain slice frame and UV-curable resin, which was highly compatible with moist bio-specimens. During UV resin curing, neither air bubble formation nor color change occurred. The resultant UV resin-mounted brain slices produced definite coronal sections with high transparency and morphological accuracy. We also performed 3D modeling by stacking brain slice images that differentiated the cortical area and nine subcortical regions via manual segmentation. This method could be a reliable alternative for displaying high-quality human brain slices and would be helpful for students and trainee to understand anatomical orientation from 2D images to 3D structures. Also, this may present an innovative approach for preparing and preserving coronal sections of the normal or pathological human brain.


Subject(s)
Brain , Neuroanatomy , Humans , Brain/anatomy & histology , Imaging, Three-Dimensional
4.
Exp Mol Pathol ; 120: 104622, 2021 06.
Article in English | MEDLINE | ID: mdl-33684392

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by neuronal and synaptic loss. The cytoplasmic tail of amyloid precursor protein (APP) undergoes sequential cleavage at a specific intracellular caspase site to generate the cytoplasmic terminal 31 (CT31) fragment. The APP-CT31 fragment is a potent inducer of apoptosis. The cytotoxicity of APP-CT31 in SH-SY5Y cells was evaluated by the lactate dehydrogenase (LDH) assay. TUNEL staining was used to detect apoptotic signals in SH-SY5Y cells and primary cortical neurons. The expression of apoptosis-related proteins, such as p53, PUMA (p53 up-regulated modulator of apoptosis), and cleaved was investigated by immunofluorescence analysis and Western blotting. In this study, we investigated the neuroprotective effect of neuregulin 1 (NRG1) against cytotoxicity induced by APP-CT31. Our data showed that CT31 induced cytotoxicity and apoptosis in SH-SY5Y cells and primary cortical neurons. NRG1 attenuated the neurotoxicity induced by the expression of APP-CT31. We also showed that APP-CT31 altered the expression of p53 and cleaved caspase 3. However, treatment with NRG1 rescued the APP-CT31-induced upregulation of p53 and cleaved caspase 3 expression. The protective effect of NRG1 was abrogated by inhibition of the ErbB4 receptor and Akt. These results indicate an important role of ErbB4/Akt signaling in NRG1-mediated neuroprotection, suggesting that endogenous NRG1/ErbB4 signaling represents a valuable therapeutic target in AD.


Subject(s)
Amyloid beta-Protein Precursor/adverse effects , Neuregulin-1/metabolism , Neuroblastoma/prevention & control , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-4/metabolism , Apoptosis , Cell Proliferation , Humans , Neuregulin-1/genetics , Neuroblastoma/etiology , Neuroblastoma/pathology , Protein Domains , Proto-Oncogene Proteins c-akt/genetics , Receptor, ErbB-4/genetics , Tumor Cells, Cultured
5.
Int J Mol Sci ; 22(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34445142

ABSTRACT

It is difficult to treat allergic diseases including asthma completely because its pathogenesis remains unclear. House dust mite (HDM) is a critical allergen and Toll-like receptor (TLR) 4 is a member of the toll-like receptor family, which plays an important role in allergic diseases. The purpose of this study was to characterize a novel allergen, Der f 38 binding to TLR4, and unveil its role as an inducer of allergy. Der f 38 expression was detected in the body and feces of Dermatophagoides farinae (DF). Electron microscopy revealed that it was located in the granule layer, the epithelium layer, and microvilli of the posterior midgut. The skin prick test showed that 60% of allergic subjects were Der f 38-positive. Der f 38 enhanced surface 203c expression in basophils of Der f 38-positive allergic subjects. By analysis of the model structure of Der p 38, the expected epitope sites are exposed on the exterior side. In animal experiments, Der f 38 triggered an infiltration of inflammatory cells. Intranasal (IN) administration of Der f 38 increased neutrophils in the lung. Intraperitoneal (IP) and IN injections of Der f 38 induced both eosinophils and neutrophils. Increased total IgE level and histopathological features were found in BALB/c mice treated with Der f 38 by IP and IN injections. TLR4 knockout (KO) BALB/c mice exhibited less inflammation and IgE level in the sera compared to wild type (WT) mice. Der f 38 directly binds to TLR4 using biolayer interferometry. Der f 38 suppressed the apoptosis of neutrophils and eosinophils by downregulating proteins in the proapoptotic pathway including caspase 9, caspase 3, and BAX and upregulating proteins in the anti-apoptotic pathway including BCL-2 and MCL-1. These findings might shed light on the pathogenic mechanisms of allergy to HDM.


Subject(s)
Allergens/immunology , Antigens, Dermatophagoides/immunology , Arthropod Proteins/immunology , Dermatophagoides farinae/immunology , Hypersensitivity/immunology , Protein Binding/immunology , Toll-Like Receptor 4/immunology , Amino Acid Sequence , Animals , Epitopes/immunology , Female , Humans , Immunoglobulin E/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pyroglyphidae/metabolism , Skin Tests/methods
6.
J Pharmacol Sci ; 137(2): 146-153, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29914798

ABSTRACT

The amyloid precursor protein (APP) is a key molecule in Alzheimer's disease. The prevailing view is that APP is initially transported to the plasma membrane as a full-length protein. Its localization at the cell surface can trigger downstream signaling and APP cleavage. Our previous work has shown that Neuregulin 1 (NRG1) has neuroprotective effects in an Alzheimer's disease model. In the present study, we examine whether NRG1 signaling is involved in APP expression and non-amyloidogenic processing in neuronal cells. Here we show that NRG1 increased the cell surface expression of APP without changing the total amount of APP mRNA or protein expression in SH-SY5Y cells and in rat primary cortical neurons. In addition, NRG1 significantly increased the levels of the secreted form of APP, sAPPα, in the conditioned media but did not change the expression of ADAM10 on the cell surface or in the cell lysates. Furthermore, we found that the protein level of NRG1 was reduced in the hippocampus of Alzheimer's disease (AD) patients. Our results demonstrate that NRG1 increased APP expression on the cell surface and sAPPα secretion into the media of neuronal cell cultures. Taken together, these results suggest a role for NRG1 in non-amyloidogenic processing.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Neuregulin-1/physiology , Neurons/metabolism , Signal Transduction/physiology , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cell Membrane/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Gene Expression/genetics , Membrane Proteins/metabolism , Neuregulin-1/metabolism , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism
7.
J Neurosci ; 35(50): 16479-93, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26674872

ABSTRACT

Expression of neuregulin-2 (NRG2) is intense in a few regions of the adult brain where neurogenesis persists; however, little is understood about its role in developments of newborn neurons. To study the role of NRG2 in synaptogenesis at different developmental stages, newborn granule cells in rat hippocampal slice cultures were labeled with retrovirus encoding tetracycline-inducible microRNA targeting NRG2 and treated with doxycycline (Dox) at the fourth or seventh postinfection day (dpi). The developmental increase of GABAergic postsynaptic currents (GPSCs) was suppressed by the early Dox treatment (4 dpi), but not by late treatment (7 dpi). The late Dox treatment was used to study the effect of NRG2 depletion specific to excitatory synaptogenesis. The Dox effect on EPSCs emerged 4 d after the impairment in dendritic outgrowth became evident (10 dpi). Notably, Dox treatment abolished the developmental increases of AMPA-receptor mediated EPSCs and the AMPA/NMDA ratio, indicating impaired maturation of glutamatergic synapses. In contrast to GPSCs, Dox effects on EPSCs and dendritic growth were independent of ErbB4 and rescued by concurrent overexpression of NRG2 intracellular domain. These results suggest that forward signaling of NRG2 mediates GABAergic synaptogenesis and its reverse signaling contributes to dendritic outgrowth and maturation of glutamatergic synapses. SIGNIFICANCE STATEMENT: The hippocampal dentate gyrus is one of special brain regions where neurogenesis persists throughout adulthood. Synaptogenesis is a critical step for newborn neurons to be integrated into preexisting neural network. Because neuregulin-2 (NRG2), a growth factor, is intensely expressed in these regions, we investigated whether it plays a role in synaptogenesis and dendritic growth. We found that NRG2 has dual roles in the development of newborn neurons. For GABAergic synaptogenesis, the extracellular domain of NRG2 acts as a ligand for a receptor on GABAergic neurons. In contrast, its intracellular domain was essential for dendritic outgrowth and glutamatergic synapse maturation. These results imply that NRG2 may play a critical role in network integration of newborn neurons.


Subject(s)
Glutamates/physiology , Hippocampus/cytology , Hippocampus/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/physiology , Synapses/physiology , gamma-Aminobutyric Acid/physiology , Animals , Animals, Newborn , Dendrites/drug effects , Doxycycline/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Female , Gene Knockdown Techniques , Hippocampus/growth & development , Male , Rats , Rats, Sprague-Dawley , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Receptors, AMPA/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects
8.
J Biol Chem ; 290(33): 20233-44, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26092725

ABSTRACT

Neuregulin 1 (NRG1) is a trophic factor that is thought to have important roles in the regulating brain circuitry. Recent studies suggest that NRG1 regulates synaptic transmission, although the precise mechanisms remain unknown. Here we report that NRG1 influences glutamate uptake by increasing the protein level of excitatory amino acid carrier (EAAC1). Our data indicate that NRG1 induced the up-regulation of EAAC1 in primary cortical neurons with an increase in glutamate uptake. These in vitro results were corroborated in the prefrontal cortex (PFC) of mice given NRG1. The stimulatory effect of NRG1 was blocked by inhibition of the NRG1 receptor ErbB4. The suppressed expression of ErbB4 by siRNA led to a decrease in the expression of EAAC1. In addition, the ablation of ErbB4 in parvalbumin (PV)-positive neurons in PV-ErbB4(-/-) mice suppressed EAAC1 expression. Taken together, our results show that NRG1 signaling through ErbB4 modulates EAAC1. These findings link proposed effectors in schizophrenia: NRG1/ErbB4 signaling perturbation, EAAC1 deficit, and neurotransmission dysfunction.


Subject(s)
Excitatory Amino Acid Transporter 3/physiology , Glutamic Acid/metabolism , Neuregulin-1/physiology , Up-Regulation , Animals , Excitatory Amino Acid Transporter 3/metabolism , Rats , Rats, Sprague-Dawley
9.
J Neurosci Methods ; 402: 110029, 2024 02.
Article in English | MEDLINE | ID: mdl-38042304

ABSTRACT

BACKGROUND: The blood-brain barrier (BBB) is a specialized layer between blood vessels and tissue in the brain, which is comprised of a neuro-glia-vascular (NGV) unit, thus play a vital role in various brain diseases. NEW METHOD: We developed the in vitro NGV units by co-culturing brain microvascular endothelial cells (BMECs; bEnd.3) and primary neural stem cells extracted from subventricular zone of adult mice. This approach was designed to mimic the RNA profile conditions found in the microvessels of a mouse brain and confirmed through various comparative transcriptome analyses. RESULTS: Optimal NGV unit development was achieved by adjusting cell density-dependent co-culture ratios. Specifically, the morphogenic development and neuronal association of astrocyte endfeet were well observed in the contact region with BMECs in the NGV unit. Through transcriptome analysis, we compared co-cultured bEnd.3/NSCs with monocultured bEnd.3 or NSCs and additionally compared them with previously reported mouse brain vascular tissue to show that this NGV unit model is a suitable in vitro model for neurological disease such as Alzheimer's disease (AD). COMPARISON WITH EXISTING METHOD(S): This in vitro NGV unit was formed from neural stem cells and vascular cells in the brain of adult mice, not embryos. It is very useful for studying brain disease mechanisms by identifying proteins and genes associated with diseases progress. CONCLUSIONS: We suggest that this simple in vitro NGV model is appropriate to investigate the relationship between BBB changes and pathological factors in the fields of neurovascular biology and cerebrovascular diseases including AD.


Subject(s)
Neural Stem Cells , Animals , Mice , Alzheimer Disease/pathology , Blood-Brain Barrier/physiology , Brain , Coculture Techniques , Endothelial Cells/physiology , Gene Expression Profiling , Neuroglia/pathology
10.
Nat Commun ; 15(1): 2219, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38472255

ABSTRACT

Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.


Subject(s)
Neurodegenerative Diseases , Animals , Humans , Neurodegenerative Diseases/pathology , Microfluidics , Organoids/pathology , Lab-On-A-Chip Devices
11.
BMC Neurosci ; 14: 112, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-24093518

ABSTRACT

BACKGROUND: There is growing evidence that inflammatory processes of activated microglia could play an important role in the progression of nerve cell damage in neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease which harbor features of chronic microglial activation, though the precise mechanism is unknown. In this study, we presented in vivo and ex vivo experimental evidences indicating that activated microglia could exacerbate the survival of axotomized dopaminergic neurons and that appropriate inactivation of microglia could be neuroprotective. RESULTS: The transection of medial forebrain bundle (MFB) of a rat induced loss of dopaminergic neurons in a time-dependent manner and accompanied with microglial activation. Along with microglial activation, production of reactive oxygen species (ROS) was upregulated and TH/OX6/hydroethidine triple-immunofluorescence showed that the microglia mainly produced ROS. When the activated microglial cells that were isolated from the substantia nigra of the MFB axotomized animal, were transplanted into the substantia nigra of which MFB had been transected at 7 days ago, the survival rate of axotomized dopaminergic neurons was significantly reduced as compared with sham control. Meanwhile, when the microglial activation was attenuated by administration of tuftsin fragment 1-3 (microglia inhibitory factor) into the lateral ventricle using mini-osmotic pump, the survival rate of axotomized dopaminergic neurons was increased. CONCLUSION: The present study suggests that activated microglia could actively produce and secrete unfavorable toxic substances, such as ROS, which could accelerate dopaminergic neuronal cell loss. So, well-controlled blockade of microglial activation might be neuroprotective in some neuropathological conditions.


Subject(s)
Dopaminergic Neurons/pathology , Microglia/metabolism , Nerve Degeneration/pathology , Reactive Oxygen Species/metabolism , Animals , Axotomy , Blotting, Western , Down-Regulation , Immunohistochemistry , Male , Medial Forebrain Bundle/injuries , Rats , Rats, Wistar , Substantia Nigra/pathology
12.
Proc Natl Acad Sci U S A ; 107(3): 1211-6, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20080551

ABSTRACT

Neuregulin 1 (NRG1) is a trophic factor thought to play a role in neural development. Recent studies suggest that it may regulate neurotransmission, mechanisms of which remain elusive. Here we show that NRG1, via stimulating GABA release from interneurons, inhibits pyramidal neurons in the prefrontal cortex (PFC). Ablation of the NRG1 receptor ErbB4 in parvalbumin (PV)-positive interneurons prevented NRG1 from stimulating GABA release and from inhibiting pyramidal neurons. PV-ErbB4(-/-) mice exhibited schizophrenia-relevant phenotypes similar to those observed in NRG1 or ErbB4 null mutant mice, including hyperactivity, impaired working memory, and deficit in prepulse inhibition (PPI) that was ameliorated by diazepam, a GABA enhancer. These results indicate that NRG1 regulates the activity of pyramidal neurons by promoting GABA release from PV-positive interneurons, identifying a critical function of NRG1 in balancing brain activity. Because both NRG1 and ErbB4 are susceptibility genes of schizophrenia, our study provides insight into potential pathogenic mechanisms of schizophrenia and suggests that PV-ErbB4(-/-) mice may serve as a model in the study of this and relevant brain disorders.


Subject(s)
ErbB Receptors/physiology , Interneurons/metabolism , Neuregulin-1/physiology , Parvalbumins/metabolism , Pyramidal Cells/metabolism , Animals , ErbB Receptors/genetics , Interneurons/cytology , Memory , Mice , Mice, Knockout , Pyramidal Cells/cytology , Receptor, ErbB-4 , gamma-Aminobutyric Acid/metabolism
13.
Int J Neurosci ; 123(1): 42-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22937735

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is a soluble factor that is released from astrocytes, the most abundant type of glial cell in the brain. PAI-1 was initially identified as inhibiting two types of plasminogen activators, that is, tissue-type plasminogen and urokinase activators that are known to lead to the proteolytic degradation of the extracellular matrix. Recently, PAI-1 was reported to mediate the neuroprotective activity of transforming growth factor-ß1 against N-methyl-D-aspartate receptor-mediated excitotoxicity and to be involved in angiogenesis following ischemic stroke, independently of the effects via the inhibition of tissue-type plasminogen and urokinase-type plasminogen activators. In this study, we examined whether PAI-1 influences synaptogenesis and neurotoxicity induced by amyloid beta peptide(1-42) (Aß(1-42)) in rat primary hippocampal neurons. Using immunostaining, treatment with PAI-1 for 24 h was found to significantly upregulate synaptophysin, postsynaptic density-95, and the polysialylated form of neural cell adhesion molecule, compared to treatment with vehicle alone. In addition, PAI-1 has neuroprotective effects against Aß(1-42)-induced cytotoxicity in rat primary cultured hippocampal neurons. Taken together, our results suggest that PAI-1 has therapeutic potential in Alzheimer's disease by promoting synaptogenesis and by demonstrating neuroprotective effects against Aß(1-42)-oligomer-induced neurotoxicity in rat primary cultured hippocampal neurons.


Subject(s)
Amyloid beta-Peptides/toxicity , Hippocampus/cytology , Neurons/drug effects , Peptide Fragments/toxicity , Plasminogen Activator Inhibitor 1/pharmacology , Synapses/drug effects , Analysis of Variance , Animals , Cells, Cultured , Disks Large Homolog 4 Protein , Fetus , Humans , Intracellular Signaling Peptides and Proteins/metabolism , L-Lactate Dehydrogenase , Membrane Proteins/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurons/enzymology , Rats , Sialic Acids/metabolism , Synaptophysin/metabolism , Up-Regulation/drug effects
14.
Neurotoxicology ; 95: 205-217, 2023 03.
Article in English | MEDLINE | ID: mdl-36796651

ABSTRACT

Recently, several studies have demonstrated that low-dose radiation (LDR) therapy has positively impacts on the treatment of Alzheimer's disease (AD). LDR suppresses the production of pro-neuroinflammation molecules and improves cognitive function in AD. However, it is unclear whether direct exposure to LDR causes beneficial effects and what mechanism is involved in neuronal cells. In this study, we first determined the effect of high-dose radiation (HDR) alone on C6 cells and SH-SY5Y cells. We found that SH-SY5Y cells were more vulnerable than C6 cells to HDR. Moreover, in neuronal SH-SY5Y cells exposed to single or multiple LDR, N-type cells showed decreased cell viability with increasing radiation exposure time and frequency, but S-type cells were unaffected. Multiple LDR increased proapoptotic molecules such as p53, Bax and cleaved caspase-3, and decreased anti-apoptotic molecule (Bcl2). Multiple LDR also generated free radicals in neuronal SH-SY5Y cells. We detected a change in the expression of the neuronal cysteine transporter EAAC1. Pretreatment with N-acetylcysteine (NAC) rescued the increased in EAAC1 expression and the generation of ROS in neuronal SH-SY5Y cells after multiple LDR. Furthermore, we verified whether the increased in EAAC1 expression induces cell defense or cell death promotion signaling. We showed that transient overexpression of EAAC1 reduced the multiple LDR-induced p53 overexpression in neuronal SH-SY5Y cells. Our results indicate that neuronal cells can be injured by increased production of ROS not only by HDR but also by multiple LDR, which suggests that combination treatment with anti-free radical agents such as NAC may be useful in multiple LDR therapy.


Subject(s)
Acetylcysteine , Neuroblastoma , Humans , Acetylcysteine/pharmacology , Acetylcysteine/metabolism , Apoptosis , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Neuroblastoma/radiotherapy , Neuroblastoma/metabolism , Oxidative Stress , Cell Survival
15.
J Pharmacol Sci ; 119(1): 73-81, 2012.
Article in English | MEDLINE | ID: mdl-22739235

ABSTRACT

Neuregulin-1 (NRG1) plays important roles in the development and plasticity of the brain, and it is also reported to have potent neuroprotective properties. We previously reported that NRG1 has neuroprotective actions against Swedish amyloid precursor protein-induced neurotoxicity. In addition to the amyloid beta peptide, other metabolites of amyloid precursor protein (APP) such as the C-terminal fragments of APP (APP-CTs) have been reported to possess cytotoxic effects in neuronal cells. In this study, we investigated whether NRG1 exerts neuroprotective effects against APP-CTs and attempted to determine its neuroprotective mechanisms. NRG1 attenuated the neurotoxicities induced by the expression of APP-CTs in neuronal cells. NRG1 also reduced the accumulation of reactive oxygen species and attenuated mitochondrial membrane potential loss induced by APP-CTs. In addition, NRG1 upregulated the expression of the anti-apoptotic protein Bcl-2. This effect was blocked by the inhibition of ErbB4, a key NRG1 receptor. Taken together, these results demonstrate the neuroprotective potential of NRG1 in Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , ErbB Receptors/metabolism , Neuregulin-1/metabolism , Neuroprotective Agents/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apoptosis Regulatory Proteins/metabolism , Brain/metabolism , Cell Death/physiology , Humans , Membrane Potential, Mitochondrial/physiology , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Receptor, ErbB-4 , Superoxide Dismutase/metabolism , Up-Regulation
16.
Korean J Med Educ ; 34(3): 223-229, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36070992

ABSTRACT

PURPOSE: This study investigated students' perceptions of non-face-to-face theory classes and face-to-face laboratory classes conducted in anatomy courses at medical schools during the coronavirus disease 2019 pandemic. METHODS: This study utilized a questionnaire to assess self-reported academic achievement level, satisfaction with non-face-to-face theory classes, satisfaction with face-to-face laboratory classes, and self-directed learning level, and conducted difference verification and regression analysis for 51 students who took anatomy courses from the fall semester of 2020 to the spring semester of 2021. RESULTS: The group with a high self-reported academic achievement level was more satisfied with the non-face-to-face theory classes than the group with a low self-reported academic achievement level. The group with a high self-reported academic achievement level had a higher self-directed learning level than the group with a low self-reported academic achievement level. In addition, it was found that the higher the self-directed learning level, the higher the satisfaction with non-face-to-face theory classes. CONCLUSION: These results suggest that to enhance satisfaction with non-face-to-face theory classes in an anatomy course, a favorable class environment that can increase the self-directed learning level is needed. In particular, careful concern is required when designing non-face-to-face classes for students with a low self-reported academic achievement.


Subject(s)
Academic Success , COVID-19 , Students, Medical , Humans , Pandemics , Perception
17.
Transl Psychiatry ; 12(1): 324, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35945212

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder that exhibits neurobehavioral deficits characterized by abnormalities in social interactions, deficits in communication as well as restricted interests, and repetitive behaviors. The basal ganglia is one of the brain regions implicated as dysfunctional in ASD. In particular, the defects in corticostriatal function have been reported to be involved in the pathogenesis of ASD. Surface deformation of the striatum in the brains of patients with ASD and their correlation with behavioral symptoms was reported in magnetic resonance imaging (MRI) studies. We demonstrated that prenatal valproic acid (VPA) exposure induced synaptic and molecular changes and decreased neuronal activity in the striatum. Using RNA sequencing (RNA-Seq), we analyzed transcriptome alterations in striatal tissues from 10-week-old prenatally VPA-exposed BALB/c male mice. Among the upregulated genes, Nurr1 was significantly upregulated in striatal tissues from prenatally VPA-exposed mice. Viral knockdown of Nurr1 by shRNA significantly rescued the reduction in dendritic spine density and the number of mature dendritic spines in the striatum and markedly improved social deficits in prenatally VPA-exposed mice. In addition, treatment with amodiaquine, which is a known ligand for Nurr1, mimicked the social deficits and synaptic abnormalities in saline-exposed mice as observed in prenatally VPA-exposed mice. Furthermore, PatDp+/- mice, a commonly used ASD genetic mouse model, also showed increased levels of Nurr1 in the striatum. Taken together, these results suggest that the increase in Nurr1 expression in the striatum is a mechanism related to the changes in synaptic deficits and behavioral phenotypes of the VPA-induced ASD mouse model.


Subject(s)
Autism Spectrum Disorder , Prenatal Exposure Delayed Effects , Animals , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/genetics , Behavior, Animal , Disease Models, Animal , Female , Humans , Male , Mice , Pregnancy , Social Behavior , Transcriptome , Valproic Acid/adverse effects
18.
Neuron ; 54(4): 583-97, 2007 May 24.
Article in English | MEDLINE | ID: mdl-17521571

ABSTRACT

Neuregulin-1 (NRG1) signaling participates in numerous neurodevelopmental processes. Through linkage analysis, nrg1 has been associated with schizophrenia, although its pathophysiological role is not understood. The prevailing models of schizophrenia invoke hypofunction of the glutamatergic synapse and defects in early development of hippocampal-cortical circuitry. Here, we show that the erbB4 receptor, as a postsynaptic target of NRG1, plays a key role in activity-dependent maturation and plasticity of excitatory synaptic structure and function. Synaptic activity leads to the activation and recruitment of erbB4 into the synapse. Overexpressed erbB4 selectively enhances AMPA synaptic currents and increases dendritic spine size. Preventing NRG1/erbB4 signaling destabilizes synaptic AMPA receptors and leads to loss of synaptic NMDA currents and spines. Our results indicate that normal activity-driven glutamatergic synapse development is impaired by genetic deficits in NRG1/erbB4 signaling leading to glutamatergic hypofunction. These findings link proposed effectors in schizophrenia: NRG1/erbB4 signaling perturbation, neurodevelopmental deficit, and glutamatergic hypofunction.


Subject(s)
ErbB Receptors/physiology , Glutamic Acid/metabolism , Neuronal Plasticity/physiology , Neurons/cytology , Synapses/physiology , Animals , Animals, Newborn , Drug Interactions , Enzyme Inhibitors/pharmacology , ErbB Receptors/genetics , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Hippocampus/cytology , In Vitro Techniques , Microscopy, Confocal/methods , N-Methylaspartate/pharmacology , Neuregulin-1/genetics , Neuregulin-1/metabolism , Patch-Clamp Techniques/methods , Quinazolines , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Receptor, ErbB-4 , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Synaptic Transmission/radiation effects , Transfection/methods , Tyrphostins/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
19.
Neuron ; 54(4): 599-610, 2007 May 24.
Article in English | MEDLINE | ID: mdl-17521572

ABSTRACT

Neuregulin-1 (NRG1), a regulator of neural development, has been shown to regulate neurotransmission at excitatory synapses. Although ErbB4, a key NRG1 receptor, is expressed in glutamic acid decarboxylase (GAD)-positive neurons, little is known about its role in GABAergic transmission. We show that ErbB4 is localized at GABAergic terminals of the prefrontal cortex. Our data indicate a role of NRG1, both endogenous and exogenous, in regulation of GABAergic transmission. This effect was blocked by inhibition or mutation of ErbB4, suggesting the involvement of ErbB4. Together, these results indicate that NRG1 regulates GABAergic transmission via presynaptic ErbB4 receptors, identifying a novel function of NRG1. Because both NRG1 and ErbB4 have emerged as susceptibility genes of schizophrenia, these observations may suggest a mechanism for abnormal GABAergic neurotransmission in this disorder.


Subject(s)
Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Neurons/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , In Situ Hybridization , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/radiation effects , Neurons/cytology , Neurons/physiology , Neurons/radiation effects , Patch-Clamp Techniques/methods , Potassium Chloride/pharmacology , Presynaptic Terminals/metabolism , Rats , Receptor, ErbB-4 , Transfection/methods
20.
Cell Death Discov ; 6: 73, 2020.
Article in English | MEDLINE | ID: mdl-32818073

ABSTRACT

Neonatal maternal separation (NMS), as an early-life stress (ELS), is a risk factor to develop emotional disorders. However, the exact mechanisms remain to be defined. In the present study, we investigated the mechanisms involved in developing emotional disorders caused by NMS. First, we confirmed that NMS provoked impulsive behavior, orienting and nonselective attention-deficit, abnormal grooming, and depressive-like behaviors in adolescence. Excitatory amino acid carrier 1 (EAAC1) is an excitatory amino acid transporter expressed specifically by neurons and is the route for the neuronal uptake of glutamate/aspartate/cysteine. Compared with that in the normal control group, EAAC1 expression was remarkably reduced in the ventral hippocampus and cerebral cortex in the NMS group. Additionally, EAAC1 expression was reduced in parvalbumin-positive hippocampal GABAergic neurons in the NMS group. We also found that EAAC1-knockout (EAAC1-/-) mice exhibited impulsive-like, nonselective attention-deficit, and depressive-like behaviors compared with WT mice in adolescence, characteristics similar to those of the NMS behavior phenotype. Taken together, our results revealed that ELS induced a reduction in EAAC1 expression, suggesting that reduced EAAC1 expression is involved in the pathophysiology of attention-deficit and depressive behaviors in adolescence caused by NMS.

SELECTION OF CITATIONS
SEARCH DETAIL