Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Life Sci ; 346: 122618, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38614306

ABSTRACT

AIMS: This study was designed to investigate the role of growth arrest and DNA damage-inducible ß (GADD45B) in modulating fear memory acquisition and elucidate its underlying mechanisms. MAIN METHODS: Adeno-associated virus (AAV) that knockdown or overexpression GADD45B were injected into ventral hippocampal CA1 (vCA1) by stereotactic, and verified by fluorescence and Western blot. The contextual fear conditioning paradigm was employed to examine the involvement of GADD45B in modulating aversive memory acquisition. The Y-maze and novel location recognition (NLR) tests were used to examine non-aversive cognition. The synaptic plasticity and electrophysiological properties of neurons were measured by slice patch clamp. KEY FINDINGS: Knockdown of GADD45B in the vCA1 significantly enhanced fear memory acquisition, accompanied by an upregulation of long-term potentiation (LTP) expression and intrinsic excitability of vCA1 pyramidal neurons (PNs). Conversely, overexpression of GADD45B produced the opposite effects. Notably, silencing the activity of vCA1 neurons abolished the impact of GADD45B knockdown on fear memory development. Moreover, mice with vCA1 GADD45B overexpression exhibited impaired spatial cognition, whereas mice with GADD45B knockdown did not display such impairment. SIGNIFICANCE: These results provided compelling evidence for the crucial involvement of GADD45B in the formation of aversive memory and spatial cognition.


Subject(s)
CA1 Region, Hippocampal , Fear , GADD45 Proteins , Mice, Inbred C57BL , Animals , Male , Fear/physiology , Mice , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Cognition/physiology , Memory/physiology , Long-Term Potentiation/physiology , Maze Learning/physiology , Neuronal Plasticity/physiology , Antigens, Differentiation/metabolism , Antigens, Differentiation/genetics , Gene Knockdown Techniques
2.
Mol Nutr Food Res ; 68(7): e2300669, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38491393

ABSTRACT

Alzheimer's disease (AD) is the most prevailing form of dementia, with long-term high-fat diet (HFD) consumption being a pivotal contributor to AD pathogenesis. As microglial dysfunction is a crucial factor in the AD onset, it becomes imperative to explore the effects of HFD on microglial function and AD pathogenesis. In the present study, 3xTg-AD model mice at the age of 9-month are subjected to random allocation, with one group receiving a standard diet (ND) and the other an HFD for 3 months. Subsequently, transcriptomic profiling of microglia unveils that HFD alters fatty acid metabolism and mediates T cell infiltration. Within the hippocampus, microglia exhibit aberrant morphology and lipid accretion in response to the HFD, evidenced by conspicuously enlarged microglial cell bodies and accumulation of lipid droplets. These lipid-droplet-accumulating microglia exhibit diminished migratory capacity and compromise plaque consolidation, thereby exacerbating the accumulation of ß-amyloid. Noteworthy, the HFD induces T cell infiltration, thereby aggravating neuroinflammation and Tau phosphorylation. Morris water maze test reveals that HFD-consuming mice display marked impairment in memory performance. In summary, this study demonstrates that prolonged HFD consumption exacerbates amyloid deposition, tau pathology, and cognitive deficits, which is associated with the accumulation of lipid droplets within microglia.


Subject(s)
Alzheimer Disease , Diet, High-Fat , Animals , Mice , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Lipids , Mice, Transgenic , Microglia , tau Proteins/metabolism
3.
Front Neurosci ; 18: 1349409, 2024.
Article in English | MEDLINE | ID: mdl-38332860

ABSTRACT

The growth arrest and DNA damage inducible protein 45 (GADD45) family comprises stress-induced nuclear proteins that interact with DNA demethylases to facilitate DNA demethylation, thereby regulating diverse cellular processes including oxidative stress, DNA damage repair, apoptosis, proliferation, differentiation, inflammation, and neuroplasticity by modulating the expression patterns of specific genes. Widely expressed in the central nervous system, the GADD45 family plays a pivotal role in various neurological disorders, rendering it a potential therapeutic target for central nervous system diseases. This review presented a comprehensive overview of the expression patterns and potential mechanisms of action associated with each member of GADD45 family (GADD45α, GADD45ß, and GADD45γ) in neurodevelopmental, neurodegenerative, and neuropsychiatric disorders, while also explored strategies to harness these mechanisms for intervention and treatment. Future research should prioritize the development of effective modulators targeting the GADD45 family for clinical trials aimed at treating central nervous system diseases.

4.
Front Behav Neurosci ; 15: 743484, 2021.
Article in English | MEDLINE | ID: mdl-34744654

ABSTRACT

Zona incerta (ZI), a largely inhibitory subthalamic region connected with many brain areas, has been suggested to serve as an integrative node for modulation of behaviors and physiological states, such as fear memory conditioning and aversion responses. It is, however, unclear whether ZI regulated the repeated social defeat stress (RSDS)-induced social conditioned place aversion (CPA) and post-traumatic stress disorder (PTSD)-like behaviors. In this study, the function of ZI was silenced via bilateral injection of tetanus toxin light chain (Tet-tox), a neurotoxin that completely blocks the evoked synaptic transmissions, expressing adeno-associated viruses (AAVs). We found ZI silencing: (1) significantly blocked the expression of RSDS-induced social CPA with no effect on the innate preference; (2) significantly enhanced the anxiety level in mice experienced RSDS with no effect on the locomotion activity; (3) altered the PTSD-associated behaviors, including the promotion of spatial cognitive impairment and the preventions of PPI deficit and social avoidance behavior. These effects were not observed on non-stressed mice. In summary, our results suggest the important role of ZI in modulating RSDS-induced social CPA and PTSD-like behaviors.

5.
Neurotoxicology ; 78: 163-169, 2020 05.
Article in English | MEDLINE | ID: mdl-32203791

ABSTRACT

Methamphetamine (METH) has been reported to induce endoplasmic reticulum (ER) stress and neuronal apoptosis in the central nervous system (CNS) during the development of addiction. Thioredoxin-1 (Trx-1) is a redox regulating protein and plays an important role in inhibiting apoptosis and protects neurons from cytotoxicity through ER and mitochondria-mediated pathways. Our previous study has been reported that Trx-1 protects mice from METH-induced rewarding effect. However, whether Trx-1 plays the role in resisting METH injury is still unclear. Here, we aim to investigate whether Trx-1 participates in the regulation of METH-induced CNS injury via ER stress and mitochondria-mediated pathways. Our study first repeated the conditioned place preference expression induced by METH. Then we detected and found that METH increased the expression of N-methyl-d-asparate (NMDA) receptor subunit 2B (NR2B) and the level of glutamate (Glu) in the ventral tegmental area (VTA) and nucleus accumbens (NAc), while Trx-1 overexpression suppressed the increases. We further examined ER stress-related proteins and mitochondrial apoptosis pathway in the VTA and NAc, and found that METH increased the expressions of glucose regulated protein 78 (GRP78), C/EBP homologous protein (CHOP), and Bax, as same time decreased the expressions of procaspase12, Bcl-2, and procaspase3, while Trx-1 overexpression blocked these changes. These results indicate that Trx-1 blocks METH-induced injury by suppressing ER stress and mitochondria-mediated apoptosis in the VTA and NAc via targeting glutamatergic system.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Brain/metabolism , Central Nervous System Stimulants/toxicity , Endoplasmic Reticulum Stress/drug effects , Methamphetamine/toxicity , Thioredoxins/metabolism , Animals , Endoplasmic Reticulum Chaperone BiP , Male , Mice, Inbred C57BL , Mice, Transgenic , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
6.
Int J Neurosci ; 129(1): 30-35, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29936883

ABSTRACT

AIM OF THE STUDY: Parkinson's disease (PD) is a neurodegenerative disorder. It is caused by the degeneration of dopaminergic neurons and the dopamine (DA) deletion in the substantia nigra pars compacta (SNpc). Morphine elevates the level of dopamine in the mesolimbic dopamine system and plays a role in alleviating PD symptoms. However, the molecular mechanism is still unclear. The aim of the study is to investigate the mechanism on morphine alleviating PD symptoms. MATERIALS AND METHODS: The viability of PC12 cells was measured by using MTT assay. The expressions of tyrosine hydroxylase (TH), thioredoxin-1 (Trx-1), CyclinD1 and Cyclin-dependent kinase5 (Cdk5) were detected by Western Blot. RESULTS: In present study, we found that morphine increased the cell viability in PC12 cells. 1-methyl-4-phenylpyridi-nium (MPP+) reduced the cell viability and TH expression, which were reversed by morphine. MPP+ decreased the expressions of Trx-1, CyclinD1, Cdk5, which were restored by morphine. Moreover, the role of morphine in restoring the expressions of Trx-1, CyclinD1 and Cdk5 decreased by MPP+ was abolished by LY294002, phosphatidylinositol-3-kinase (PI3K)/Akt inhibitor. CONCLUSIONS: These results suggest that morphine reverses effects induced by MPP þ through activating PI3K/Akt pathway.


Subject(s)
1-Methyl-4-phenylpyridinium/administration & dosage , Morphine/administration & dosage , Parkinson Disease/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Survival/drug effects , PC12 Cells , Rats , Signal Transduction/drug effects , Tyrosine 3-Monooxygenase/metabolism
7.
Neuropharmacology ; 143: 63-70, 2018 12.
Article in English | MEDLINE | ID: mdl-30240785

ABSTRACT

Morphine is widely used for clinical pain management and induces the dependence. Addiction to morphine is a major public health issue. Geranylgeranylacetone (GGA) is widely used in clinic for treating ulcer. GGA induces expression of thioredoxin-1 (Trx-1) extensively. Trx-1 is a redox regulating protein and plays protecting roles in nervous system. GGA prevents mice against morphine-induced hyperlocomotion, rewarding effect, and withdrawal syndrome. However, whether GGA blocks morphine-conditioned place preference (CPP) reinstatement is still unknown. In the present study, we found that GGA administration blocked the reinstatement of morphine-CPP. The expressions of Trx-1, N-methyl d-aspartate receptor 2B subunit (NR2B), phosphorylated Ca2+/calmodulin-dependent protein kinase II (p-CaMKII), phosphorylated extracellular signaling regulated kinases (p-ERK), and phosphorylated cAMP-response element binding protein (p-CREB) were induced in nucleus accumbens (NAc) and hippocampus by morphine or GGA, whereas these proteins were not changed by morphine in GGA-treated mice. Our results indicate that GGA may prevent the reinstatement of morphine-CPP through strengthening the expression of Trx-1 and regulating NR2B/ERK pathway. Thus, we suggest that GGA may be a promising therapeutic candidate for morphine-induced relapse.


Subject(s)
Diterpenes/pharmacology , Morphine Dependence/drug therapy , Psychotropic Drugs/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Morphine/pharmacology , Morphine Dependence/metabolism , Narcotics/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phosphorylation/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Recurrence , Spatial Behavior/drug effects , Spatial Behavior/physiology , Thioredoxins/metabolism
8.
J Psychopharmacol ; 32(9): 1037-1046, 2018 09.
Article in English | MEDLINE | ID: mdl-30136629

ABSTRACT

BACKGROUND: Drug addiction is characterized by compulsive drug use and relapse. Thioredoxin-1 is emerging as an important modulator involved in the cellular protective response against a variety of toxic stressors. Previous study has reported that thioredoxin-1 overexpression prevents the acquisition of methamphetamine-conditioned place preference. Here, we aimed to investigate the effect of thioredoxin-1 on methamphetamine-conditioned place preference extinction and the possible mechanism. METHODS: (a) An extinction procedure in mice was employed to investigate the effect of thioredoxin-1 on the extinction of methamphetamine-conditioned place preference. After the acquisition of methamphetamine-conditioned place preference, mice underwent the following procedures: the injection of thioredoxin-1 small interfering RNA in the ventral tegmental area followed by the post-conditioned place preference test, four days of extinction training followed by four days of recovery after surgery. (b) The levels of thioredoxin-1, dopamine D1 receptor, tyrosine hydroxylase, phosphorylated extracellular regulated kinase, and phosphorylated cyclic adenosine monophosphate response element binding protein were examined by using Western blot analysis. RESULTS: Thioredoxin-1 downregulation in the ventral tegmental area delayed methamphetamine-conditioned place preference extinction. The expression of thioredoxin-1 was decreased in the ventral tegmental area of mice in control and negative groups after methamphetamine-conditioned place preference extinction, but not in the thioredoxin-1 siRNA group. The levels of dopamine D1 receptor, tyrosine hydroxylase, phosphorylated extracellular regulated kinase, and phosphorylated cyclic adenosine monophosphate response element binding protein were decreased in the ventral tegmental area, nucleus accumbens, and prefrontal cortex of mice in the control and negative groups after methamphetamine-conditioned place preference extinction, but were inversely increased in thioredoxin-1 siRNA group. CONCLUSIONS: The results suggest that downregulation of thioredoxin-1 in the ventral tegmental area may delay methamphetamine-conditioned place preference extinction by regulating the mesocorticolimbic dopaminergic signaling pathway.


Subject(s)
Down-Regulation , Extinction, Psychological/drug effects , Methamphetamine/pharmacology , Thioredoxins/antagonists & inhibitors , Thioredoxins/metabolism , Ventral Tegmental Area/metabolism , Animals , Conditioning, Classical/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Mice , Microinjections , Mitogen-Activated Protein Kinase 3/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phosphorylation , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , RNA, Small Interfering/pharmacology , Receptors, Dopamine D1/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/drug effects
9.
Neuropharmacology ; 139: 117-123, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29981334

ABSTRACT

Relapse of drug abuse after abstinence is a major challenge to the treatment of addicts. Thioredoxin-1 (Trx-1) is an important regulator of neuroprotection, and inhibits morphine-induced hyperlocomotion, reward and withdrawal signs, as well as blocks methamphetamine (METH)-induced conditioned place preference (CPP). The nucleus accumbens (NAc) is essential for relapse like behavior in reinstatement animal models. In the present study, we aimed to investigate the role of Trx-1 in the NAc in METH-primed reinstatement by using a reinstatement procedure in mice. Adeno-associated virus vectors expressing shRNA-mTrx-1 (AAV-shRNA-mTrx-1) were bilaterally microinjected into the NAc after METH-CPP extinction. The results showed that Trx-1 downregulation in the NAc promoted the reinstatement of METH-CPP. We also examined the expression of N-methyl-D-asparate (NMDA) receptor 2B subunit (GluN2b), the levels of phosphorylated extracellular signal-regulated kinase (p-ERK) and phosphorylated cAMP-response element binding protein (p-CREB) in the NAc by western blot analysis, and found that the GluN2b expression, p-ERK and p-CREB levels were increased in the NAc in response to low-dose METH in AAV-shRNA-mTrx-1 mice, but were not changed in control and AAV-vehicle mice. These data indicate that the increased GluN2b expression, and p-ERK and p-CREB levels in the NAc of AAV-shRNA-mTrx-1 mice may be responsible for the METH-primed reinstatement. Thus, we suggest that downregulation of Trx-1 in the NAc may make mice more sensitive to METH reinstatement.


Subject(s)
Amphetamine-Related Disorders/metabolism , Central Nervous System Stimulants/pharmacology , Down-Regulation , Methamphetamine/pharmacology , Nucleus Accumbens/metabolism , Thioredoxins/metabolism , Animals , Conditioning, Psychological/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Extinction, Psychological/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Phosphorylation , Receptors, N-Methyl-D-Aspartate/metabolism , Spatial Behavior/drug effects , Spatial Behavior/physiology
10.
Front Neurol ; 9: 309, 2018.
Article in English | MEDLINE | ID: mdl-29770121

ABSTRACT

Morphine is one kind of opioid, which is currently the most effective widely utilized pain relieving pharmaceutical. Long-term administration of morphine leads to dependence and addiction. Thioredoxin-1 (Trx-1) is an important redox regulating protein and works as a neurotrophic cofactor. Our previous study showed that geranylgeranylaceton, an inducer of Trx-1 protected mice from rewarding effects induced by morphine. However, whether overexpression of Trx-1 can block morphine-induced conditioned place preference (CPP) in mice is still unknown. In this study, we first examined whether overexpression of Trx-1 affects the CPP after morphine training and further examined the dopamine (DA) and γ-aminobutyric acid (GABA) systems involved in rewarding effects. Our results showed that morphine-induced CPP was blocked in Trx-1 overexpression transgenic (TG) mice. Trx-1 expression was induced by morphine in the ventral tegmental area (VTA) and nucleus accumbens (NAc) in wild-type (WT) mice, which was not induced in Trx-1 TG mice. The DA level and expressions of tyrosine hydroxylase (TH) and D1 were induced by morphine in WT mice, which were not induced in Trx-1 TG mice. The GABA level and expression of GABABR were decreased by morphine, which were restored in Trx-1 TG mice. Therefore, Trx-1 may play a role in blocking CPP induced by morphine through regulating the expressions of D1, TH, and GABABR in the VTA and NAc.

11.
Behav Brain Res ; 337: 280-286, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-28782589

ABSTRACT

Methamphetamine (METH) is a highly addictive drug of abuse which induces behavioral sensitization and rewarding effects. Thioredoxin-1 (Trx-1) is a redox protein and plays roles in regulating activity of transcription factor, such as cAMP responsive element-binding protein (CREB), AP-1, p53, is emerging as an important modulator of neuronal function. It has been reported that Trx-1 is involved in morphine dependence. In this study, we examined the rewarding effect after METH administration by conditioned place preference (CPP) of mice, and detected the levels of dopamine and the activity of cAMP responsive element-binding protein (CREB), the expressions of ΔFosB and cyclin-dependent kinase 5 (CDK5) in the ventral tegmental area (VTA) and nucleus accumbens (NAc) in mice. Our results showed that the expression of METH-CPP was occluded in Trx-1 overexpression transgenic (TG) mice. The increase of dopamine level induced by METH was not further higher in Trx-1 TG mice. METH decreased the expression of Trx-1 which was restored in TG mice. The activity of CREB and the expressions of ΔFosB and CDK5 were increased by METH in wile-type mice, which were not further increased in TG mice. These results suggest that overexpression of Trx-1 may occlude the CPP induced by METH through regulating the activity of CREB and the expression of ΔFosB.


Subject(s)
Central Nervous System Stimulants/pharmacology , Conditioning, Operant/drug effects , Methamphetamine/pharmacology , Reward , Thioredoxins/metabolism , Animals , Antioxidants/pharmacology , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Conditioning, Operant/physiology , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Cystine/analogs & derivatives , Cystine/pharmacology , Dopamine/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Thioredoxins/genetics , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
12.
Cancer Biomark ; 20(4): 527-537, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-28800315

ABSTRACT

miR-99a is down-regulated in esophageal squamous cell carcinoma (ESCC), however the role and underlying mechanism are still unknown. We aim to explore the role and mechanism of miR-99a down-regulation in ESCC. The expression of miR-99a in ESCC tissues and cell lines was detected by Human miRNA Microarrays and Real-time PCR. The effects of miR-99a on cell proliferation, migration and invasion were determined by Cell Counting Kit-8 (CCK-8) assay, transwell migration and invasion assay. Target gene of miR-99a were analyzed by target prediction software and validated by Real-time PCR and Western blotting assay. Our microarray results and four Gene Expression Omnibus (GEO) datasets showed lower expression level of miR-99a in ESCC tissues. Overexpression of miR-99a using mimics significantly suppressed cell proliferation, and decreased expressions of CCND1, CCNA2 and CCNE1. We also found that enhanced miR-99a significantly inhibited migration, invasion and epithelial-mesenchymal transition (EMT) of ESCC cells, and down-regulated EMT associated transcription factor Slug, and MMPs including MMP2, MMP7 and MMP13. TargetScan predicted insulin-like growth factor 1 receptor (IGF1R) as the cadidate target gene of miR-99a, and western blotting confirmed the negative correlation between miR-99a and IGF1R. Importantly, we further found that knockdown of IGF1R also significantly inhibited the proliferation, migration, invasion and slug-induced EMT of ESCC cells, and reduced the cell cycle regulatory proteins and MMPs. In conclusion, our findings suggested that loss of miR-99a in ESCC promoted the tumor cell proliferation, migration, invasion and slug-induced EMT through activating IGF1R signaling pathway.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Receptors, Somatomedin/metabolism , Signal Transduction , 3' Untranslated Regions , Biomarkers , Carcinoma, Squamous Cell/pathology , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Computational Biology/methods , Epithelial-Mesenchymal Transition/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Humans , RNA Interference , Receptor, IGF Type 1
13.
Shock ; 47(6): 753-758, 2017 06.
Article in English | MEDLINE | ID: mdl-28505020

ABSTRACT

Sepsis is a serious public health issue and the leading cause of death in critically ill patients in intensive care units. Thioredoxin-1 (Trx-1) is a protein of regulating redox, as well as a modulator of inflammation and apoptosis. Our previous study reported that Trx-1 decreased endoplasmic reticulum-mediated inflammation involved in lung in a model of experimental sepsis. However, its effect on mitochondrial-mediated apoptosis in spleen has not been reported. We studied whether Trx-1 could prevent spleen cells apoptosis in sepsis. In the present study, we showed that the apoptosis in spleen was decreased in sepsis induced by cecal ligation and puncture (CLP) in Trx-1 overexpression transgenic (Tg) mice compared with wild-type mice. Colony forming units in the peritoneal cavity and the level of procalcitonin in plasma were significantly decreased in Trx-1 Tg mice 12 h after CLP. The expressions of c-jun-N-terminal kinase, Bax, caspase-9, and caspase-3 were increased in spleen, which were suppressed in Trx-1 Tg mice. However, the decreased Bcl-2 expression in sepsis was recovered in Trx-1 Tg mice. Our results suggest that overexpression of Trx-1 provides protection against sepsis through suppressing mitochondria-induced apoptosis pathway in spleen. This study may provide a new target for clinical intervention, as well potential strategies for treatment of sepsis.


Subject(s)
Mitochondria/metabolism , Sepsis/metabolism , Spleen/cytology , Thioredoxins/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Caspase 3/metabolism , Caspase 9/metabolism , Male , Mice , Mice, Transgenic , Sepsis/genetics , Spleen/metabolism , Thioredoxins/genetics
14.
Shock ; 46(1): 67-74, 2016 07.
Article in English | MEDLINE | ID: mdl-27299588

ABSTRACT

Sepsis is the main cause of death in critically ill patients, pathogenesis of which is still unclear. The nuclear factor κB (NF-κB) inflammatory signal pathway mediated by endoplasmic reticulum stress is involved in sepsis. Thioredoxin-1 (Trx-1) is an important protein of regulating oxidative stress. It plays a crucial role in the anti-oxidation, anti-apoptosis, and anti-inflammation. However, the role and the mechanism of Trx-1 in sepsis have not been extensively studied. In the present study, we showed that the survival was longer in sepsis induced by cecal ligation and puncture in Trx-1 overexpression transgenic (Tg) mice compared with wild-type mice. Wet/dry lung weight ratio was decreased in Trx-1 Tg mice. The levels of TNF-α and IL-1ß in plasma and lung tissue were inhibited in Tg mice. The expressions of glucose-regulated protein 78, inositol-requiring enzyme 1α (IRE1α), tumor necrosis factor receptor-associated factor 2, C/EBP homologous protein, NF-κB, and inhibitors of NF-κBα were increased in lung tissue. More importantly, the overexpression of Trx-1 in transgenic mice suppressed NF-κB inflammatory signal pathway by inhibiting the activation of molecules involved in ER stress. Our results suggest that Trx-1 may play protective role in extending survival in sepsis by regulating inflammatory response through suppressing ER stress.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Inflammation/metabolism , Sepsis/metabolism , Sepsis/mortality , Thioredoxins/metabolism , Animals , Cecum/injuries , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Heat-Shock Proteins/metabolism , Humans , Inflammation/genetics , Interleukin-1beta/metabolism , Ligation/adverse effects , Lung/metabolism , Male , Mice , Mice, Transgenic , NF-kappa B/metabolism , Sepsis/genetics , Thioredoxins/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Nanotechnology ; 21(47): 475206, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-21030760

ABSTRACT

The resistive switching properties of Cu-doped-HfO(2)-based resistive-random-access-memory (ReRAM) devices are investigated under proton-based irradiations with different high-range total doses of 1.5, 3 and 5 Giga-rad[Si]. The measurement results obtained immediately after irradiation demonstrate that the proton-based total dose will introduce significant variations in the operation voltages and resistance values. These effects are enhanced almost linearly when the dose increases from 1.5 to 5 Giga-rad[Si]. Furthermore, five days after irradiation, the electrical properties of the device rebound, resulting in reduced operation voltages and resistance values. This is consistent with the time-dependent super-recovery behavior observed previously in CMOS gate oxide. These results can be explained by the proton irradiation effect on the electron/hole trap density inside HfO(2) and its impact on ReRAM device metallic filament formation-and-rupture, which is based on electrolyte theory.

SELECTION OF CITATIONS
SEARCH DETAIL
...