Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nano Lett ; 2024 May 24.
Article in English | MEDLINE | ID: mdl-38785400

ABSTRACT

The gallium-doped hafnium oxide (Ga-HfO2) films with different Ga doping concentrations were prepared by adjusting the HfO2/Ga2O3 atomic layer deposition cycle ratio for high-speed and low-voltage operation in HfO2-based ferroelectric memory. The Ga-HfO2 ferroelectric films reveal a finely modulated coercive field (Ec) from 1.1 (HfO2/Ga2O3 = 32:1) to an exceptionally low 0.6 MV/cm (HfO2/Ga2O3 = 11:1). This modulation arises from the competition between domain nucleation and propagation speed during polarization switching, influenced by the intrinsic domain density and phase dispersion in the film with specific Ga doping concentrations. Higher Ec samples exhibit a nucleation-dominant switching mechanism, while lower Ec samples undergo a transition from a nucleation-dominant to a propagation-dominant reversal mechanism as the electric field increases. This work introduces Ga as a viable dopant for low Ec and offers insights into material design strategies for HfO2-based ferroelectric memory applications.

2.
Aging Cell ; : e14187, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38716507

ABSTRACT

Behavioral changes or neuropsychiatric symptoms (NPSs) are common features in dementia and are associated with accelerated cognitive impairment and earlier deaths. However, how NPSs are intertwined with cognitive decline remains elusive. In this study, we identify that the basolateral amygdala (BLA) is a key brain region that is associated with mood disorders and memory decline in the AD course. During the process from pre- to post-onset in AD, the dysfunction of parvalbumin (PV) interneurons and pyramidal neurons in the amygdala leads to hyperactivity of pyramidal neurons in the basal state and insensitivity to external stimuli. We further demonstrate that serotonin (5-HT) receptors in distinct neurons synergistically regulate the BLA microcircuit of AD rather than 5-HT levels, in which both restrained inhibitory inputs by excessive 5-HT1AR signaling in PV interneurons and depolarized pyramidal neurons via upregulated 5-HT2AR contribute to aberrant neuronal hyperactivity. Downregulation of these two 5-HT receptors simultaneously enables neurons to resist ß-amyloid peptides (Aß) neurotoxicity and ameliorates the mood and cognitive defects. Therefore, our study reveals a crucial role of 5-HT receptors for regulating neuronal homeostasis in AD pathogenesis, and this would provide early intervention and potential targets for AD cognitive decline.

3.
Zool Res ; 45(3): 679-690, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38766749

ABSTRACT

General anesthesia is widely applied in clinical practice. However, the precise mechanism of loss of consciousness induced by general anesthetics remains unknown. Here, we measured the dynamics of five neurotransmitters, including γ-aminobutyric acid, glutamate, norepinephrine, acetylcholine, and dopamine, in the medial prefrontal cortex and primary visual cortex of C57BL/6 mice through in vivo fiber photometry and genetically encoded neurotransmitter sensors under anesthesia to reveal the mechanism of general anesthesia from a neurotransmitter perspective. Results revealed that the concentrations of γ-aminobutyric acid, glutamate, norepinephrine, and acetylcholine increased in the cortex during propofol-induced loss of consciousness. Dopamine levels did not change following the hypnotic dose of propofol but increased significantly following surgical doses of propofol anesthesia. Notably, the concentrations of the five neurotransmitters generally decreased during sevoflurane-induced loss of consciousness. Furthermore, the neurotransmitter dynamic networks were not synchronized in the non-anesthesia groups but were highly synchronized in the anesthetic groups. These findings suggest that neurotransmitter dynamic network synchronization may cause anesthetic-induced loss of consciousness.


Subject(s)
Anesthetics, Inhalation , Mice, Inbred C57BL , Neurotransmitter Agents , Propofol , Sevoflurane , Sevoflurane/pharmacology , Animals , Propofol/pharmacology , Neurotransmitter Agents/metabolism , Mice , Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/pharmacology , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
4.
Nat Commun ; 15(1): 3221, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622129

ABSTRACT

The hippocampus creates a cognitive map of the external environment by encoding spatial and self-motion-related information. However, it is unclear whether hippocampal neurons could also incorporate internal cognitive states reflecting an animal's exploratory intention, which is not driven by rewards or unexpected sensory stimuli. In this study, a subgroup of CA1 neurons was found to encode both spatial information and animals' investigatory intentions in male mice. These neurons became active before the initiation of exploration behaviors at specific locations and were nearly silent when the same fields were traversed without exploration. Interestingly, this neuronal activity could not be explained by object features, rewards, or mismatches in environmental cues. Inhibition of the lateral entorhinal cortex decreased the activity of these cells during exploration. Our findings demonstrate that hippocampal neurons may bridge external and internal signals, indicating a potential connection between spatial representation and intentional states in the construction of internal navigation systems.


Subject(s)
Intention , Spatial Navigation , Male , Mice , Animals , Space Perception/physiology , Hippocampus/physiology , Entorhinal Cortex , Cues , Spatial Navigation/physiology
5.
Cancer Sci ; 114(7): 2882-2894, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37137487

ABSTRACT

ANP32B, a member of the acidic leucine-rich nuclear phosphoprotein 32 kDa (ANP32) family of proteins, is critical for normal development because its constitutive knockout mice are perinatal lethal. It is also shown that ANP32B acts as a tumor-promoting gene in some kinds of cancer such as breast cancer and chronic myelogenous leukemia. Herein, we observe that ANP32B is lowly expressed in B-cell acute lymphoblastic leukemia (B-ALL) patients, which correlates with poor prognosis. Furthermore, we utilized the N-myc or BCR-ABLp190 -induced B-ALL mouse model to investigate the role of ANP32B in B-ALL development. Intriguingly, conditional deletion of Anp32b in hematopoietic cells significantly promotes leukemogenesis in two B-ALL mouse models. Mechanistically, ANP32B interacts with purine rich box-1 (PU.1) and enhances the transcriptional activity of PU.1 in B-ALL cells. Overexpression of PU.1 dramatically suppresses B-ALL progression, and highly expressed PU.1 significantly reverses the accelerated leukemogenesis in Anp32b-deficient mice. Collectively, our findings identify ANP32B as a suppressor gene and provide novel insight into B-ALL pathogenesis.


Subject(s)
Burkitt Lymphoma , Leukemia, Myeloid , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Mice , Nuclear Proteins/genetics , Mice, Knockout , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Fusion Proteins, bcr-abl , Nerve Tissue Proteins/metabolism , Cell Cycle Proteins/metabolism
6.
Neuron ; 111(10): 1626-1636.e6, 2023 05 17.
Article in English | MEDLINE | ID: mdl-36917979

ABSTRACT

Lasker's award-winning drug propofol is widely used in general anesthesia. The recreational use of propofol is reported to produce a well-rested feeling and euphoric state; yet, the neural mechanisms underlying such pleasant effects remain unelucidated. Here, we report that propofol actively and directly binds to the dopamine transporter (DAT), but not the serotonin transporter (SERT), which contributes to the rapid relief of anhedonia. Then, we predict the binding mode of propofol by molecular docking and mutation of critical binding residues on the DAT. Fiber photometry recording on awake freely moving mice and [18F] FP-CIT-PET scanning further establishes that propofol administration evokes rapid and lasting dopamine accumulation in nucleus accumbens (NAc). The enhanced dopaminergic tone drives biased activation of dopamine-receptor-1-expressing medium spiny neurons (D1-MSNs) in NAc and reverses anhedonia in chronically stressed animals. Collectively, these findings suggest the therapeutic potential of propofol against anhedonia, which warrants future clinical investigations.


Subject(s)
Dopamine , Propofol , Mice , Animals , Dopamine/metabolism , Propofol/pharmacology , Propofol/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Molecular Docking Simulation , Receptors, Dopamine D1/metabolism , Nucleus Accumbens/physiology , Anhedonia , Mice, Inbred C57BL
7.
J Hematol Oncol ; 16(1): 9, 2023 02 11.
Article in English | MEDLINE | ID: mdl-36774506

ABSTRACT

BACKGROUND: Selectively targeting leukemia stem cells (LSCs) is a promising approach in treating acute myeloid leukemia (AML), for which identification of such therapeutic targets is critical. Increasing lines of evidence indicate that FBXO22 plays a critical role in solid tumor development and therapy response. However, its potential roles in leukemogenesis remain largely unknown. METHODS: We established a mixed lineage leukemia (MLL)-AF9-induced AML model with hematopoietic cell-specific FBXO22 knockout mice to elucidate the role of FBXO22 in AML progression and LSCs regulation, including self-renewal, cell cycle, apoptosis and survival analysis. Immunoprecipitation combined with liquid chromatography-tandem mass spectrometry analysis, Western blotting and rescue experiments were performed to study the mechanisms underlying the oncogenic role of FBXO22. RESULTS: FBXO22 was highly expressed in AML, especially in MLL-rearranged (MLLr) AML. Upon FBXO22 knockdown, human MLLr leukemia cells presented markedly increased apoptosis. Although conditional deletion of Fbxo22 in hematopoietic cells did not significantly affect the function of hematopoietic stem cells, MLL-AF9-induced leukemogenesis was dramatically abrogated upon Fbxo22 deletion, together with remarkably reduced LSCs after serial transplantations. Mechanistically, FBXO22 promoted degradation of BACH1 in MLLr AML cells, and overexpression of BACH1 suppressed MLLr AML progression. In line with this, heterozygous deletion of BACH1 significantly reversed delayed leukemogenesis in Fbxo22-deficient mice. CONCLUSIONS: FBXO22 promotes MLLr AML progression by targeting BACH1 and targeting FBXO22 might be an ideal strategy to eradicate LSCs without influencing normal hematopoiesis.


Subject(s)
Basic-Leucine Zipper Transcription Factors , F-Box Proteins , Leukemia, Myeloid, Acute , Receptors, Cytoplasmic and Nuclear , Animals , Humans , Mice , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Cycle , F-Box Proteins/genetics , F-Box Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/pathology , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplastic Stem Cells/pathology , Receptors, Cytoplasmic and Nuclear/metabolism
8.
Neuropharmacology ; 220: 109259, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36126726

ABSTRACT

Somatic symptom disorder (SSD), which occurs in about 5-7 percent of the adult population, involves heightened physical and emotional sensitivity to pain. However, its neural mechanism remains elusive and thus hinders effective clinical intervention. In this study, we employed chronic restraint stress (CRS)-induced hyperalgesia as a mouse model to investigate the neural mechanism underlying SSD and its pharmacological treatment. We found that CRS induced hyperactivity of anterior cingulate cortex (ACC), whereas chemogenetic inhibition of such hyperactivity could prevent CRS-induced hyperalgesia. Systematic application and ACC local infusion of fluoxetine alleviated CRS-induced hyperalgesia. Moreover, we found that fluoxetine exerted its anti-hyperalgesic effects through inhibiting the hyperactivity of ACC and upregulating 5-HT1A receptors. Our study thus uncovers the functional role of 5-HT signaling in modulating pain sensation and provides a neural basis for developing precise clinical intervention for SSD.


Subject(s)
Fluoxetine , Hyperalgesia , Animals , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Gyrus Cinguli , Hyperalgesia/drug therapy , Mice , Pain/drug therapy , Serotonin
9.
Blood ; 138(24): 2485-2498, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34359074

ABSTRACT

Proper regulation of p53 signaling is critical for the maintenance of hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs). The hematopoietic cell-specific mechanisms regulating p53 activity remain largely unknown. Here, we demonstrate that conditional deletion of acidic leucine-rich nuclear phosphoprotein 32B (ANP32B) in hematopoietic cells impairs repopulation capacity and postinjury regeneration of HSCs. Mechanistically, ANP32B forms a repressive complex with p53 and thus inhibits the transcriptional activity of p53 in hematopoietic cells, and p53 deletion rescues the functional defect in Anp32b-deficient HSCs. Of great interest, ANP32B is highly expressed in leukemic cells from patients with chronic myelogenous leukemia (CML). Anp32b deletion enhances p53 transcriptional activity to impair LSC function in a murine CML model and exhibits synergistic therapeutic effects with tyrosine kinase inhibitors in inhibiting CML propagation. In summary, our findings provide a novel strategy to enhance p53 activity in LSCs by inhibiting ANP32B and identify ANP32B as a potential therapeutic target in treating CML.


Subject(s)
Cell Cycle Proteins/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Tumor Suppressor Protein p53/genetics
10.
Mol Psychiatry ; 26(8): 3956-3969, 2021 08.
Article in English | MEDLINE | ID: mdl-31772302

ABSTRACT

Social interaction and communication are evolutionary conserved behaviours that are developed in mammals to establish partner cognition. Deficit in sociability has been represented in human patients and animal models of neurodevelopmental disorders, which are connected with genetic variants of synaptic glutamate receptors and associated PDZ-binding proteins. However, it remains elusive how these key proteins are specialized in the cellular level for the initial social behaviour during postnatal developmental stage. Here we identify a hippocampal CA3 specifically expressed PDZ scaffold protein Lnx1 required for initial social behaviour. Through gene targeting we find that Lnx1 deficiency led to a hippocampal subregional disorder in neuronal activity and social memory impairments for partner discrimination observed in juvenile mice which also show cognitive defects in adult stage. We further demonstrate that Lnx1 deletion causes NMDA receptor (NMDAR) hypofunction and this is attributable to decreased GluN2B expression in PSD compartment and disruption of the Lnx1-NMDAR-EphB2 complex. Specific restoration of Lnx1 or EphB2 protein in the CA3 area of Lnx1-/- mice rescues the defective synaptic function and social memory. These findings thus reveal crucial roles of postsynaptic NMDAR multiprotein complex that regulates the formation of initial social memory during the adolescent period.


Subject(s)
CA3 Region, Hippocampal/physiology , Memory , Receptors, N-Methyl-D-Aspartate , Social Behavior , Ubiquitin-Protein Ligases , Animals , Memory Disorders/genetics , Mice , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism
11.
Transl Neurodegener ; 9(1): 18, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32398165

ABSTRACT

BACKGROUND: Neuropsychiatric symptoms (NPS) such as depression, anxiety, apathy, and irritability occur in prodromal phases of clinical Alzheimer's disease (AD), which might be an increased risk for later developing AD. Here we treated young APP/PS1 AD model mice prophylactically with serotonin-selective re-uptake inhibitor (SSRI) paroxetine and investigated the protective role of anti-depressant agent in emotional abnormalities and cognitive defects during disease progress. METHODS: To investigate the protective role of paroxetine in emotional abnormalities and cognitive defects during disease progress, we performed emotional behaviors of 3 months old APP/PS1 mouse following oral administration of paroxetine prophylactically starting at 1 month of age. Next, we tested the cognitive, biochemical and pathological, effects of long term administration of paroxetine at 6 months old. RESULTS: Our results showed that AD mice displayed emotional dysfunction in the early stage. Prophylactic administration of paroxetine ameliorated the initial emotional abnormalities and preserved the eventual memory function in AD mice. CONCLUSION: Our data indicate that prophylactic administration of paroxetine ameliorates the emotional dysfunction and memory deficit in AD mice. These neuroprotective effects are attributable to functional restoration of glutamate receptor (GluN2A) in AD mice.


Subject(s)
Affective Symptoms/drug therapy , Alzheimer Disease/drug therapy , Memory Disorders/drug therapy , Neuroprotective Agents/therapeutic use , Paroxetine/therapeutic use , Prodromal Symptoms , Affective Symptoms/genetics , Affective Symptoms/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Mice, Transgenic , Neuroprotective Agents/metabolism , Paroxetine/metabolism , Presenilin-1/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Selective Serotonin Reuptake Inhibitors/metabolism , Selective Serotonin Reuptake Inhibitors/therapeutic use , Time Factors
12.
Cell Death Dis ; 10(7): 486, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217475

ABSTRACT

Liver kinase B1 (LKB1) regulates both cell growth and energy metabolism. Inactivated mutations of LKB1, observed in 20-30% of nonsmall cell lung cancers (NSCLC), contribute significantly to lung cancer malignancy progression. However, the upstream signalings regulating LKB1 activity remain incompletely understood. Here, we present evidence that FBXO22 interacts with and promotes polyubiquitination of LKB1. More intriguingly, FBXO22 mediates Lys-63-linked LKB1 polyubiquitination and inhibits kinase activity of LKB1. Furthermore, over-expression of FBXO22 promotes NSCLC cell growth through inhibiting LKB1-AMPK-mTOR signaling in vitro and in vivo. Clinically, FBXO22 is highly expressed in human lung adenocarcinoma and high FBXO22 expression predicts significant poor prognosis. Our study provides new insights into the upstream regulation of LKB1 activation and identifies FBXO22 as a potential therapeutic target for lung cancer treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , F-Box Proteins/metabolism , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , A549 Cells , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Colony-Forming Units Assay , F-Box Proteins/genetics , Female , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Lung Neoplasms/genetics , Mice , Mice, Nude , Protein Serine-Threonine Kinases/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Ubiquitination/genetics , Ubiquitination/physiology
13.
J Cell Biol ; 217(11): 4007-4024, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30185604

ABSTRACT

Neuronal connections are initiated by axon targeting to form synapses. However, how the maturation of axon terminals is modulated through interacting with postsynaptic elements remains elusive. In this study, we find that ligand of Numb protein X 1 (Lnx1), a postsynaptic PDZ protein expressed in hippocampal CA3 pyramidal neurons, is essential for mossy fiber (MF) axon targeting during the postnatal period. Lnx1 deletion causes defective synaptic arrangement that leads to aberrant presynaptic terminals. We further identify EphB receptors as novel Lnx1-binding proteins to form a multiprotein complex that is stabilized on the CA3 neuron membrane through preventing proteasome activity. EphB1 and EphB2 are independently required to transduce distinct signals controlling MF pruning and targeting for precise DG-CA3 synapse formation. Furthermore, constitutively active EphB2 kinase rescues structure of the wired MF terminals in Lnx1 mutant mice. Our data thus define a retrograde trans-synaptic regulation required for integration of post- and presynaptic structure that participates in building hippocampal neural circuits during the adolescence period.


Subject(s)
Axons/metabolism , CA3 Region, Hippocampal/metabolism , Mossy Fibers, Hippocampal/metabolism , Pyramidal Cells/metabolism , Synapses/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Mice , Mice, Knockout , Presynaptic Terminals/metabolism , Receptor, EphB1/genetics , Receptor, EphB1/metabolism , Receptor, EphB2/genetics , Receptor, EphB2/metabolism , Synapses/genetics , Ubiquitin-Protein Ligases/genetics
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(3): 296-304, 2018 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-29643035

ABSTRACT

OBJECTIVE: To analyze the correlation between the expressions of vascular endothelial growth factor (VEGF) and transient receptor potential canonical 6 (TRPC6) and their role in podocyte injury in rats with diabetic nephropathy. METHODS: Forty SD rats with diabetic nephropathy induced by intraperitoneal injection of 65 mg/kg streptozotocin were randomized equally into 5 groups, including a diabetic nephropathy model group and 4 treatment groups, with 8 normal SD rats as the normal control group. In the 4 treatment groups, the rats received intraperitoneal injections with SU5416 at 5 mg/kg or 10 mg/kg twice a week or with LY294002 at 1 mg/kg or 2 mg/kg once daily for 8 weeks. Blood glucose, serum creatinine, blood urea nitrogen, and 24-h urinary protein levels of the rats were detected at different time points, and the pathologies in the renal tissue were observed using HE staining, PAS staining and immunohistochemistry. The expressions of VEGF, nephrin, and TRPC6 at mRNA and protein levels were detected using RT-PCR and Western blotting. RESULTS: Compared with normal control rats, the diabetic rats showed significantly increased fasting blood glucose, serum creatinine, blood urea nitrogen and 24-h urinary protein levels with decreased expressions of nephrin mRNA and protein (P<0.05) and increased expressions of VEGF and TRPC6 (P<0.05). Compared with the untreated diabetic rats, the rats with SU5416 treatment showed increased 24-h urinary protein, urea nitrogen, and nephrin expression and decreased TRPC6 expression without significant changes in fasting blood glucose, serum creatinine, or VEGF expression. The rats treated with LY294002 showed decreased 24-h urinary protein and TRPC6 expression without significant changes in fasting blood glucose, serum creatinine, urea nitrogen, or expressions of nephrin and VEGF. CONCLUSION: The regulatory effect of VEGF on TRPC6 can be blocked by inhibiting VEGFR-2 or blocking PI3K/Akt signaling pathway.


Subject(s)
Diabetic Nephropathies/pathology , Podocytes/pathology , TRPC Cation Channels/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Chromones/pharmacology , Diabetes Mellitus, Experimental , Diabetic Nephropathies/metabolism , Indoles/pharmacology , Kidney/physiopathology , Membrane Proteins/metabolism , Morpholines/pharmacology , Pyrroles/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley
15.
Sci Rep ; 7: 40614, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28094295

ABSTRACT

Traumatic brain injury (TBI) is a principal cause of death and disability worldwide, which is a major public health problem. Death caused by TBI accounts for a third of all damage related illnesses, which 75% TBI occurred in low and middle income countries. With the increasing use of motor vehicles, the incidence of TBI has been at a high level. The abnormal brain functions of TBI patients often show the acute and long-term neurological dysfunction, which mainly associated with the pathological process of malignant brain edema and neuroinflammation in the brain. Owing to the neuroinflammation lasts for months or even years after TBI, which is a pivotal causative factor that give rise to neurodegenerative disease at late stage of TBI. Studies have shown that platelet activating factor (PAF) inducing inflammatory reaction after TBI could not be ignored. The morphological and behavioral abnormalities after TBI in wild type mice are rescued by general knockout of PAFR gene that neuroinflammation responses and cognitive ability are improved. Our results thus define a key inflammatory molecule PAF that participates in the neuroinflammation and helps bring about cerebral dysfunction during the TBI acute phase.


Subject(s)
Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/physiopathology , Inflammation/genetics , Platelet Membrane Glycoproteins/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Astrocytes/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Targeting , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/ultrastructure , Inflammation Mediators/metabolism , Male , Memory , Mice , Mice, Knockout , Neuroglia/metabolism , Spatial Learning
16.
J Neurosci ; 36(39): 10151-62, 2016 09 28.
Article in English | MEDLINE | ID: mdl-27683910

ABSTRACT

UNLABELLED: The amygdala serves as emotional center to mediate innate fear behaviors that are reflected through neuronal responses to environmental aversive cues. However, the molecular mechanism underlying the initial neuron responses is poorly understood. In this study, we monitored the innate defensive responses to aversive stimuli of either elevated plus maze or predator odor in juvenile mice and found that glutamatergic neurons were activated in amygdala. Loss of EphB2, a receptor tyrosine kinase expressed in amygdala neurons, suppressed the reactions and led to defects in spine morphogenesis and fear behaviors. We further found a coupling of spinogenesis with these threat cues induced neuron activation in developing amygdala that was controlled by EphB2. A constitutively active form of EphB2 was sufficient to rescue the behavioral and morphological defects caused by ablation of ephrin-B3, a brain-enriched ligand to EphB2. These data suggest that kinase-dependent EphB2 intracellular signaling plays a major role for innate fear responses during the critical developing period, in which spinogenesis in amygdala glutamatergic neurons was involved. SIGNIFICANCE STATEMENT: Generation of innate fear responses to threat as an evolutionally conserved brain feature relies on development of functional neural circuit in amygdala, but the molecular mechanism remains largely unknown. We here identify that EphB2 receptor tyrosine kinase, which is specifically expressed in glutamatergic neurons, is required for the innate fear responses in the neonatal brain. We further reveal that EphB2 mediates coordination of spinogenesis and neuron activation in amygdala during the critical period for the innate fear. EphB2 catalytic activity plays a major role for the behavior upon EphB-ephrin-B3 binding and transnucleus neuronal connections. Our work thus indicates an essential synaptic molecular signaling within amygdala that controls synapse development and helps bring about innate fear emotions in the postnatal developing brain.


Subject(s)
Amygdala/physiology , Fear/physiology , Glutamates/metabolism , Instinct , Neurogenesis/physiology , Neurons/physiology , Receptor, EphB2/metabolism , Aging/physiology , Animals , Defense Mechanisms , Gene Expression Regulation, Developmental/physiology , Mice , Mice, Knockout , Mice, Transgenic , Signal Transduction/physiology
17.
ACS Appl Mater Interfaces ; 8(34): 22309-15, 2016 Aug 31.
Article in English | MEDLINE | ID: mdl-27514235

ABSTRACT

Highly oriented multilayered BaTiO3-(Ba,Ca)TiO3-CaTiO3 thin films were fabricated on Nb-doped (001) SrTiO3 (Nb:STO) substrates by pulsed laser deposition. The configurations of multilayered BaTiO3-(Ba,Ca)TiO3-CaTiO3 thin films are designed with the thickness ratio of 1:1:1 and 2:1:1 and total thickness ∼300 nm. Microstructural characterization by X-ray diffraction indicates that the as-deposited thin films are highly c-axis oriented and large in-plane strain is determined in BaTiO3 and CaTiO3 layers. Piezoresponse force microscopy (PFM) studies reveal an intense in-plane polarization component, whereas the out-of-plane shows inferior phase contrast. The optimized combination is found to be the BaTiO3-(Ba0.85Ca0.15)TiO3-CaTiO3 structure with combination ratio 2:1:1, which displays the largest domain switching amplitude under DC electric field, the largest room-temperature dielectric constant ∼646, a small dielectric loss of 0.03, and the largest dielectric tunability of ∼50% at 400 kV/cm. These results suggest that the enhanced dielectric and tunability performance are greatly associated with the large in-plane polarization component and domain switching.

18.
Nat Commun ; 7: 11096, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27008987

ABSTRACT

Innate emotion response to environmental stimuli is a fundamental brain function that is controlled by specific neural circuits. Dysfunction of early emotional circuits may lead to neurodevelopmental disorders such as autism and schizophrenia. However, how the functional circuits are formed to prime initial emotional behaviours remain elusive. We reveal here using gene-targeted mutations an essential role for ephrin-B3 ligand-like activity in the development of innate fear in the neonatal brain. We further demonstrate that ephrin-B3 controls axon targeting and coordinates spinogenesis and neuronal activity within the amygdala. The morphological and behavioural abnormalities in ephrin-B3 mutant mice are rescued by conditional knock-in of wild-type ephrin-B3 during the critical period when axon targeting and fear responses are initiated. Our results thus define a key axonal molecule that participates in the wiring of amygdala circuits and helps bring about fear emotion during the important adolescence period.


Subject(s)
Amygdala/metabolism , Axons/metabolism , Ephrin-B3/metabolism , Fear/physiology , Instinct , Neurogenesis , Animals , Cell Nucleus/metabolism , Ephrin-B3/genetics , Mice , Mutation/genetics , Synapses/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...