Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Phys Chem A ; 127(34): 7148-7155, 2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37595363

ABSTRACT

In understanding the mechanism of aggregation-induced emission (AIE), the multilevel ONIOM framework has been demonstrated as one of the efficient tools that can capture the essential mechanistic information by choosing a single fluorophore as the quantum mechanics (QM) model and putting all surrounding molecules in the low-level region. Recently, the ionic styryl-pyridine salt (namely, SPH) has been reported as a new class of AIEgen with a high fluorescence yield. In the SPH crystal, a pair of ionic SPH molecules are closely stacked with each other in an antiparallel, head-to-tail pattern, thus the choice of QM models (an individual or dimeric structure) becomes critical in the ONIOM study. Herein we report the AIE mechanism of the ionic SPH at the QM ((TD)-CAM-B3LYP) and ONIOM(QM:MM) levels. As usual, the fluorescence quenching of SPH in tetrahydrofuran (THF) solution is attributed to a nonradiative relaxation via the central C═C bond rotation, with a rather low barrier of 2.7 kcal/mol. In crystals, either with a monomer or dimer model, the fluorescence quenching channel is found to be restricted due to the obvious C═C rotation barriers. Compared with the monomer model, the dimer model, by treating the orbital interaction of the two SPH molecules at the QM level, provides significantly increased barriers and a red-shifted emission wavelength that better matches the experimental value. In addition, the calculated exciton coupling in the fluorescence emission state can be discovered only by a dimer model. The findings here emphasize not only the importance of choosing a proper model in the ONIOM study of AIE but also expanding our understanding of novel AIE systems.

2.
Neurochem Res ; 43(12): 2446-2459, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30382449

ABSTRACT

Perfluorooctanesulfonate (PFOS) may cause neurotoxicity through the initiation of oxidative stress. In the current study, we investigated the role of anti-oxidant nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in PFOS-induced neurotoxicity. We found that human neuroblastoma SH-SY5Y cells exhibited significant apoptotic cell death following PFOS exposure, and this process was accompanied with apparent accumulation of reactive oxidative species (ROS). In addition, we revealed that PFOS exposure caused marked activation of Nrf2 pathway and the expression of Nrf2 transcription target heme oxygenase-1. We further found that pre-treatment with ROS scavenger N-acetyl-L-cysteine (NAC) dramatically ameliorated PFOS-induced ROS production and Nrf2 signaling. In keeping with these findings, western blot and Cell Counter Kit-8 analyses revealed that pre-incubation with NAC suppressed PFOS-induced expression of pro-apoptotic proteins and impairment of neuronal viability. Moreover, antagonizing Nrf2 pathway with Nrf2 inhibitor brusatol resulted in increased ROS production and enhanced PFOS-induced expression of apoptosis related proteins. Finally, we showed that PFOS exposure altered mitochondrial transmembrane potential and disrupted normal mitochondrial morphology in SH-SY5Y cells. Whereas treatment with NAC ameliorated PFOS-induced mitochondrial disorders, co-incubation with brusatol augmented PFOS-induced mitochondrial deficits, consequently contributing to neuronal apoptosis. These results manifest that Nrf2 pathway plays a protective role in PFOS-induced neurotoxicity, providing new insights into the prevention and treatment of PFOS-related toxicities.


Subject(s)
Alkanesulfonic Acids/toxicity , Apoptosis/physiology , Fluorocarbons/toxicity , NF-E2-Related Factor 2/metabolism , Neuroprotection/physiology , Oxidative Stress/physiology , Signal Transduction/physiology , Apoptosis/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Neuroprotection/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects
3.
Neurotoxicology ; 66: 32-42, 2018 05.
Article in English | MEDLINE | ID: mdl-29526747

ABSTRACT

Perfluorooctanesulfonate (PFOS)-containing compounds are widely used in all aspects of industrial and consumer products. Recent studies indicated that PFOS is ubiquitous in environments and is considered to be a new type of persistent organic pollutant (POP). This has raised concerns regarding its adverse effects on human health. The nervous system is regarded as a sensitive target of environmental contaminants, including PFOS. Previous findings showed that PFOS can induce neurobehavioral deficits. However, the molecular mechanism underlying PFOS neurotoxicity remains obscure. Astrocyte activation and the resulting pro-inflammatory cytokine release play an integral role in protecting neurons from neurotoxin-mediated damage. If uncontrolled, sustained astrocyte activation may cause the secretion of excessive levels of pro-inflammatory cytokines that exacerbate the initial damage. In this study, we showed that PFOS could promote excessive secretion of tumor necrosis factor-α (TNF-α) in dose- and time-dependent manners in astrocytes. Furthermore, PFOS exposure could induce the phosphorylation of Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3). This suggests that the JAK2/STAT3 signal transduction pathway is involved in PFOS-mediated astrocyte activation and secretion of TNF-α. Indeed, targeted blockage of the JAK2/STAT3 pathway prevented the phosphorylation of JAK and STAT3, and it also caused abnormal expression of TNF-α. Finally, we demonstrated that SH-SY5Y neuronal cells underwent rapid apoptosis via a TNF-α-dependent mechanism after exposure to PFOS-treated astrocyte-conditioned medium. In summary, our findings reveal that PFOS mediates a rapid activation of JAK2/STAT3 signal transduction in C6 astrocytes, which plays a pivotal role in the initiation of PFOS-mediated neurotoxicity.


Subject(s)
Alkanesulfonic Acids/toxicity , Encephalitis/chemically induced , Encephalitis/metabolism , Fluorocarbons/toxicity , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Humans , Neurons/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects
4.
Neurotoxicol Teratol ; 67: 65-75, 2018.
Article in English | MEDLINE | ID: mdl-29577981

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a persistent and bioaccumulative compound that has been widely used in various fields of life and industrial productions, because of its special chemical and physical properties. Numerous studies have indicated significant neurotoxic effect of PFOS, especially on neurons and microglia. However, the influence of PFOS on astrocyte physiology remains unclear. In this study, we showed that PFOS triggered reactive astrocytosis in time- and dose-dependent manners. The low-doses of PFOS increased the cell number and the expression of glial fibrillary acidic protein (GFAP), a well-known hallmark of reactive astrocytes, in C6 astrocyte cells. ELISA and RT-PCR analysis showed that PFOS promoted the expression and secretion of Interleukin-1 beta (IL-1ß) in dose- and time-dependent manners. Furthermore, PFOS exposure could induce the phosphorylation and degradation of IκBα, and the translocation of NF-κB p65 from the cytoplasm to the nucleus in C6 glioma cell line. Thus, the NF-кB signaling pathway can be activated after PFOS exposure. In addition, pretreatment with AKT inhibitor LY294002 could obviously attenuate PFOS-induced NF-κB activation and IL-1ß secretion. Taken together, these results indicated that PFOS could facilitate reactive astrocytosis and the secretion of pro-inflammatory cytokines through AKT-dependent NF-κB signaling pathway.


Subject(s)
Alkanesulfonic Acids/toxicity , Astrocytes/metabolism , Fluorocarbons/toxicity , Interleukin-1beta/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Alkanesulfonic Acids/antagonists & inhibitors , Animals , Cell Count , Cells, Cultured , Chromones/pharmacology , Dose-Response Relationship, Drug , Fluorocarbons/antagonists & inhibitors , Glial Fibrillary Acidic Protein/metabolism , Morpholines/pharmacology , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Rats
5.
Neurochem Int ; 108: 40-51, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28232070

ABSTRACT

Manganese (Mn) is a widely distributed trace element that is essential for normal brain function and development. However, chronic exposure to excessive Mn has been known to lead to neuronal loss and manganism, a disease with debilitating motor and cognitive deficits, whose clinical syndrome resembling idiopathic Parkinson's disease (IPD). However, the precise molecular mechanism underlying Mn neurotoxicity remains largely unclear. Accumulating evidence indicates that abnormal mitochondrial functionality is an early and causal event in Mn-induced neurodegeneration and apoptosis. Here, we investigated whether Mitofusin 2 (Mfn2), a highly conserved dynamin-related protein (DRP), played a role in the regulation of Mn-induced neuronal apoptosis. We revealed that Mfn2 was significantly dysregulated in rat striatum and PC12 neuronal-like cells following Mn exposure. Western blot analysis revealed that the expression of Mfn2 was remarkably decreased following different concentrations of Mn exposure. Immunohistochemistry analysis confirmed a remarkable downregulation of Mfn2 in rat striatum after Mn exposure. Immunofluorescent staining showed that Mfn2 was expressed predominantly in neurons, and neuronal loss of Mfn2 was associated with the expression of active caspase-3 following Mn exposure. Importantly, overexpression of Mfn2 apparently attenuated Mn-induced neuronal apoptosis. Notably, treatment with caspase-3 inhibitor Ac-DEVD-CH could not rescue Mn-induced downregulation of Mfn2, suggesting that Mn-induced mfn2 occurs prior to neuronal apoptosis. Taken together, these results indicated that down-regulated expression of Mfn2 might contribute to the pathological processes underlying Mn neurotoxicity.


Subject(s)
Apoptosis/physiology , Corpus Striatum/metabolism , Down-Regulation/physiology , Manganese/toxicity , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Neurons/metabolism , Animals , Apoptosis/drug effects , Corpus Striatum/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , GTP Phosphohydrolases , Male , Membrane Proteins/antagonists & inhibitors , Mitochondrial Proteins/antagonists & inhibitors , Neurons/drug effects , PC12 Cells , Random Allocation , Rats , Rats, Sprague-Dawley
6.
Regul Toxicol Pharmacol ; 81: 480-488, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27664319

ABSTRACT

Arsenic is a widely distributed toxic metalloid in around the world. Inorganic arsenic species are deemed to affect astrocytes functions and to cause neuron apoptosis. Microglia are the key cell type involved in innate immune responses in CNS, and microglia activation has been linked to inflammation and neurotoxicity. In this study, using ELISA and reverse transcriptase PCR (RT-PCR), we showed that Arsenic trioxide up-regulated the expression and secretion of IL-6 in a dose-dependent manner and a time-dependent manner in cultured HAPI microglia cells. These pro-inflammatory responses were inhibited by the Akt blocker, LY294002. Further, Arsenic trioxide exposure could induce phospho rylationand degradation of IкBα, and the translocation of NF-κB p65 from the cytosol to the nucleus in this HAPI microglia cell line. Thus, the NF-кB signaling pathway can be activated after Arsenic trioxide treatment. Besides, Akt blocker LY294002 also obviously attenuated NF-кB activation and transnuclear induced by Arsenic trioxide. In concert with these results, we highlighted that the secretion of pro-inflammatory cytokine and NF-кB activation induced by Arsenic trioxide can be mediated by elevation of p-Akt in HAPI microglia cells.


Subject(s)
Arsenic/toxicity , Inflammation/metabolism , Interleukin-6/metabolism , Microglia/drug effects , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cells, Cultured , Inflammation/immunology , Inflammation/pathology , Interleukin-6/immunology , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Rats , Signal Transduction/drug effects
7.
Environ Toxicol Pharmacol ; 46: 9-16, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27414741

ABSTRACT

Perfluorooctane sulfonate (PFOS), the most extensively studied member of perfluoroalkyl and polyfluoroalkyl substances (PFASs), has been thought to be toxic to the central nervous system (CNS) of mammals. However, the neurotoxic effects of PFOS remain largely unknown. In this study, the effect of PFOS on microglial apoptosis was examined. The results showed that PFOS could significantly reduce the cell viability and mediate cell apoptosis in HAPI microglia, which was closely accompanied with ROS production and p53 overexpression. Moreover, p53 interference significantly ameliorated PFOS-triggered cytotoxic effects in HAPI microglia, including the downregulation of cleaved PARP and cleaved caspase 3. Interestingly, NAC, a ROS inhibitor, inhibited p53 expression, and decreased the apoptosis of HAPI microglia. Taken together, these findings suggest that upregulated production of ROS plays a vital role in PFOS-mediated apoptosis in HAPI microglia via the modulation of p53 signaling.


Subject(s)
Alkanesulfonic Acids/toxicity , Apoptosis/drug effects , Fluorocarbons/toxicity , Microglia/drug effects , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Environmental Pollutants/toxicity , Enzyme Activation/drug effects , Microglia/metabolism , Microglia/pathology , Poly(ADP-ribose) Polymerases/metabolism , Rats , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics
8.
Toxicol Appl Pharmacol ; 303: 79-89, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27174766

ABSTRACT

Arsenic is a widely distributed toxic metalloid all over the world. Inorganic arsenic species are supposed to affect astrocytic functions and to cause neuron apoptosis in CNS. Microglias are the key cell type involved in innate immune responses in CNS, and microglia activation has been linked to inflammation and neurotoxicity. In this study, using ELISA, we showed that Arsenic trioxide up-regulated the expression and secretion of IL-1ß in a dose-dependent manner and a time-dependent manner in cultured HAPI microglia cells. The secretion of IL-1ß caused the apoptosis of SH-SY5Y. These pro-inflammatory responses were inhibited by the STAT3 blocker, AG490 and P38/JNK MAPK blockers SB202190, SP600125. Further, Arsenic trioxide exposure could induce phosphorylation and activation of STAT3, and the translocation of STAT3 from the cytosol to the nucleus in this HAPI microglia cell line. Thus, the STAT3 signaling pathway can be activated after Arsenic trioxide treatment. However, P38/JNK MAPK blockers SB202190, SP600125 also obviously attenuated STAT3 activation and transnuclear transport induced by Arsenic trioxide. In concert with these results, we highlighted that the secretion of IL-1ß and STAT3 activation induced by Arsenic trioxide can be mediated by elevation of P38/JNK MAPK in HAPI microglia cells and then induced the toxicity of neurons.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Microglia/drug effects , Neurons/drug effects , Oxides/toxicity , STAT3 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Anthracenes/pharmacology , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals , Cell Line , Cell Line, Tumor , Humans , Imidazoles/pharmacology , Inflammation , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Microglia/metabolism , Neurons/metabolism , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rats , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
J Appl Toxicol ; 36(12): 1591-1598, 2016 12.
Article in English | MEDLINE | ID: mdl-27018151

ABSTRACT

The neurotoxic effects of perfluorooctane sulfonate (PFOS) have attracted significant research attention in recent years. In the present study, we investigated the impact of PFOS exposure on the physiology of neural stem cells (NSCs) in vitro. We showed that PFOS exposure markedly attenuated the proliferation of C17.2 neural stem cells in both dose- and time-dependent manners. Additionally, we found that PFOS decreased Ser9 phosphorylation of glycogen synthase kinase-3ß (pSer9-GSK-3ß), leading to the activation of GSK-3ß and resultant downregulation of cellular ß-catenin. Furthermore, blockage of GSK-3ß with lithium chloride significantly attenuated both the PFOS-induced downregulation of GSK-3ß/ß-catenin and the proliferative impairment of C17.2 cells. Notably, the expression of various downstream targets was altered accordingly, such as c-myc, cyclin D1 and survivin. In conclusion, the present study demonstrated that PFOS decreased the proliferation of C17.2 cells via the negative modulation of the GSK-3ß/ß-catenin pathway. We present the potential mechanisms underlying the PFOS-induced toxic effects on NSCs to provide novel insights into the neurotoxic mechanism of PFOS. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Alkanesulfonic Acids/toxicity , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Glycogen Synthase Kinase 3 beta/metabolism , Neural Stem Cells/drug effects , beta Catenin/metabolism , Animals , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta/genetics , Mice , Neural Stem Cells/metabolism , Signal Transduction/drug effects , Time Factors , beta Catenin/genetics
10.
Pathol Res Pract ; 212(5): 365-71, 2016 May.
Article in English | MEDLINE | ID: mdl-26997491

ABSTRACT

Glyoxylate reductase/hydroxypyruvate reductase (GRHPR), which exists mainly in the liver, is a D-2-hydroxy-acid dehydrogenase that plays a critical role in the formation of primary hyperoxaluria type 2 (PH2). Here, we investigated GRHPR expression and its potential role in both human Crohn's disease (CD) and experimental colitis. Murine experimental colitis models were established by administration of trinitrobenzenesulphonic acid (TNBS). As shown by Western blot, significant up-regulation of GRHPR was found in TNBS-treated mice as compared with normal controls. Immunohistochemistry (IHC) also showed increased GRHPR expression, and the molecule was located in intestinal epithelial cells (IECs). This phenomenon also occurred in patients with Crohn's disease. Besides, in an in vitro study, human IEC line HT-29 cells cultured with tumor necrosis factor α (TNF-α) were used to evaluate the changes in expression of GRHPR. Moreover, overexpression of GRHPR was accompanied by active caspase-3 and cleaved poly ADP-ribose polymerase (PARP) accumulation. Furthermore, knock-down GRHPR could inhibit the accumulation of active caspase-3 and cleaved PARP as shown by Western blot in TNF-α treated HT-29 cells. Flow cytometry assay indicated that interference of GRHPR led to increasing apoptosis of IECs. These data suggested that GRHPR might exert its pro-apoptosis function in IECs. Thus, GRHPR might play an important role in regulating IECs apoptosis, and might be a potential therapeutic target for CD.


Subject(s)
Alcohol Oxidoreductases/metabolism , Apoptosis/physiology , Colitis/metabolism , Intestinal Mucosa/metabolism , Up-Regulation , Animals , Caspase 3/metabolism , Cell Line, Tumor , Colitis/chemically induced , Colitis/pathology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Intestinal Mucosa/pathology , Intestines/pathology , Mice , Trinitrobenzenesulfonic Acid
11.
J Appl Toxicol ; 36(11): 1409-17, 2016 11.
Article in English | MEDLINE | ID: mdl-26988466

ABSTRACT

The widespread environmental contaminant, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), is considered one of the most toxic dioxin-like compounds. Although epidemiological studies have shown that TCDD exposure is linked to some neurological and neurophysiological disorders, the underlying mechanism of TCDD-mediated neurotoxicity has remained unclear. Astrocytes are the most abundant cells in the nervous systems, and are recognized as the important mediators of normal brain functions as well as neurological, neurodevelopmental and neurodegenerative brain diseases. In this study, we investigated the role of TCDD in regulating the expression of glutamate transporter GLT-1 in astrocytes. TCDD, at concentrations of 0.1-100 nm, had no significantly harmful effect on the viability of C6 glioma cells. However, the expression of GLT-1 in C6 glioma cells was downregulated in a dose- and time-dependent manner. TCDD also caused activation of protein kinase C (PKC), as TCDD induced translocation of the PKC from the cytoplasm or perinuclear to the membrane. The translocation of PKC was inhibited by one Ca(2+) blocker, nifedipine, suggesting that the effects are triggered by the initial elevated intracellular concentration of free Ca(2+) . Finally, we showed that inhibition of the PKC activity reverses the TCDD-triggered reduction of GLT-1. In summary, our results suggested that TCDD exposure could downregulate the expression of GLT-1 in C6 via Ca(2+) /PKC pathway. The downregulation of GLT-1 might participate in TCDD-mediated neurotoxicity. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Astrocytes/drug effects , Calcium/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Polychlorinated Dibenzodioxins/toxicity , Protein Kinase C/metabolism , Animals , Astrocytes/metabolism , Calcium Signaling , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Rats , Signal Transduction , Time Factors
12.
Environ Toxicol ; 31(9): 1068-79, 2016 Sep.
Article in English | MEDLINE | ID: mdl-25865073

ABSTRACT

The environmental toxicant TCDD may elicit cytotoxic effects by inducing reactive oxygen species (ROS) generation. Autophagy is one of the first lines of defense against oxidative stress damage. Herein, we investigated whether autophagy played a regulatory role in TCDD-induced neurotoxicity. Here, we showed that TCDD exposure caused marked autophagy in SH-SY5Y cells, whose dose range was close to that inducing apoptosis. Electron microscopic and Western blot analyses revealed that TCDD induced autophagy at a starting dose of approximate 100 nM. Interestingly, 100-200 nM TCDD exposure resulted in obviously decreased cell viability and evident apoptotic phenotype. Furthermore, the levels of pro-apoptotic molecules, Bax and cleaved-PARP, increased significantly, whereas Bcl2 declined after exposed to 100 nM TCDD. In addition, the apoptosis was verified using flow cytometrical analysis. These data strongly suggested that TCDD induced both autophagy and apoptosis at a similar dose range in SH-SY5Y cells. Interestingly, pretreatment with ROS scavenger, N-acetyl-cysteine (NAC), could effectively block both TCDD-induced apoptosis and autophagy. More surprisingly, inhibition of autophagy with 3-methyladenine (3MA), remarkably augmented TCDD-induced apoptosis. The findings implicated that the onset of autophagy might serve as a protective mechanism to ameliorate ROS-triggered cytotoxic effects in human SH-SY5Y neuronal cells under TCDD exposure. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1068-1079, 2016.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Polychlorinated Dibenzodioxins/toxicity , Protective Agents/pharmacology , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Oxidative Stress/drug effects , Phenotype , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism
13.
Toxicol Appl Pharmacol ; 288(2): 143-51, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26086160

ABSTRACT

Perfluorooctane sulfonate (PFOS), an emerging persistent contaminant that is commonly encountered during daily life, has been shown to exert toxic effects on the central nervous system (CNS). However, the molecular mechanisms underlying the neurotoxicity of PFOS remain largely unknown. It has been widely acknowledged that the inflammatory mediators released by hyper-activated microglia play vital roles in the pathogenesis of various neurological diseases. In the present study, we examined the impact of PFOS exposure on microglial activation and the release of proinflammatory mediators, including nitric oxide (NO) and reactive oxidative species (ROS). We found that PFOS exposure led to concentration-dependent NO and ROS production by rat HAPI microglia. We also discovered that there was rapid activation of the ERK/JNK MAPK signaling pathway in the HAPI microglia following PFOS treatment. Moreover, the PFOS-induced iNOS expression and NO production were attenuated after the inhibition of ERK or JNK MAPK by their corresponding inhibitors, PD98059 and SP600125. Interestingly, NAC, a ROS inhibitor, blocked iNOS expression, NO production, and activation of ERK and JNK MAPKs, which suggested that PFOS-mediated microglial NO production occurs via a ROS/ERK/JNK MAPK signaling pathway. Finally, by exposing SH-SY5Y cells to PFOS-treated microglia-conditioned medium, we demonstrated that NO was responsible for PFOS-mediated neuronal apoptosis.


Subject(s)
Alkanesulfonic Acids/toxicity , Environmental Pollutants/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorocarbons/toxicity , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Microglia/drug effects , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Microglia/enzymology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Paracrine Communication/drug effects , Protein Kinase Inhibitors/pharmacology , Rats , Time Factors
14.
Neurochem Res ; 40(6): 1220-31, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25998883

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been recently shown to elicit inflammatory response in a number of cell-types. However, whether TCDD could provoke inflammation in astrocytes, the most abundant glial cells in central nervous system (CNS), remains virtually unknown. In the present study, we showed that TCDD exposure could induce evident astrocyte activation both in vivo and in vitro. Further, we found that TGF-ß-activated kinase 1 (TAK1), a critical regulator of NF-κB signaling, was rapidly phosphorylated in the process of TCDD-induced reactive astroglia. Exposure to TCDD led to rapid TAK1 and NF-κB p65 phosphorylation, as well as IKBα degradation. Moreover, blockage of TAK1 using siRNA oligos or TAK1 inhibitor 5Z-7-oxozeaenol significantly attenuated TCDD-induced astrocyte activation as well as the release of TNF-α. Finally, we showed that the conditioned medium of TCDD-treated astrocytes promoted the apoptosis of PC12 neuronal cells, which could be blocked with the pre-treatment of TAK1 inhibitor. Taken together, these findings suggested that TCDD could promote the inflammatory activation of astrocytes through modulating TAK1-NF-κB cascade, implicating that reactive astrocytes might contribute to TCDD-induced adverse effects on CNS system.


Subject(s)
Astrocytes/drug effects , Environmental Pollutants/toxicity , MAP Kinase Kinase Kinases/drug effects , NF-kappa B/drug effects , Neurons/drug effects , Polychlorinated Dibenzodioxins/toxicity , Animals , Cell Death/drug effects , Cells, Cultured , Culture Media, Conditioned , Female , I-kappa B Proteins/drug effects , I-kappa B Proteins/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , PC12 Cells , Phosphorylation , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Transcription Factor RelA/metabolism
15.
Int Immunopharmacol ; 28(1): 52-60, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26004316

ABSTRACT

Perfluorooctane sulfonate (PFOS), a ubiquitous pollutant widely found in the environment and biota, can cause numerous adverse effects on human health. In recent years, PFOS's toxic effects on the central nervous system (CNS) have been shown. However, we still have a lot to study in the underlying molecular mechanism of PFOS's neurotoxicity. Microglia, the innate immune cells of CNS, are critically implicated in various neurological diseases caused by pro-inflammatory mediators. In our research, we found that HAPI microglia secreted tumor necrosis factor-alpha (TNF-α) after PFOS exposure in time-dependent and dose-dependent way. We also discovered that intracellular concentration of free Ca(2+) ([Ca(2+)]i) significantly increased after PFOS treatments. It was noteworthy here the secretion of TNF-α mediated by PFOS was blocked by Ca(2+) inhibitor and protein kinase C (PKC) inhibitor. Besides these, we had learned as well that PFOS brought about the up-regulation of phosphorylated nuclear factor kappa B (NF-кB) p65 expression and accelerated degradation of NF-κB inhibitor alpha (IкBα), however, these effects could be attenuated or blocked by Ca(2+) inhibitor and PKC inhibitor. Finally, through treating SH-SY5Y cells with PFOS-treated microglial conditioned medium, we demonstrated that TNF-α mediated neuronal apoptosis. To sum up, our research had shown, for the first time, that the distinct TNF-α secretion brought by PFOS in HAPI microglia, was achieved through the Ca(2+)-dependent PKC-NF-кB signaling, subsequently participating in neuronal loss.


Subject(s)
Alkanesulfonic Acids/toxicity , Calcium Signaling/drug effects , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Macrophage Activation/drug effects , Microglia/drug effects , Microglia/metabolism , NF-kappa B/drug effects , Protein Kinase C/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Cell Line , Dose-Response Relationship, Drug , Humans , I-kappa B Kinase/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Transcription Factor RelA/biosynthesis , Up-Regulation/drug effects
16.
Biochem Biophys Res Commun ; 463(1-2): 116-22, 2015.
Article in English | MEDLINE | ID: mdl-26002461

ABSTRACT

Elevated free fatty acids (FFAs) are fundamental to the pathogenesis of hepatic insulin resistance. However, the molecular mechanisms of insulin resistance remain not completely understood. Transcriptional dysregulation, post-transcriptional modifications and protein degradation contribute to the pathogenesis of insulin resistance. Poly(C) binding proteins (PCBPs) are RNA-binding proteins that are involved in post-transcriptional control pathways. However, there are little studies about the roles of PCBPs in insulin resistance. PCBP2 is the member of the RNA-binding proteins and is thought to participate in regulating hypoxia inducible factor-1 (HIF-1α) and signal transducers and activators of transcription (STAT) pathway which are involved in regulating insulin signaling pathway. Here, we investigated the influence of PCBP2 on hepatic insulin resistance. We showed that the protein and mRNA levels of PCBP2 were down-regulated under insulin-resistant conditions. In addition, we showed that over-expression of PCBP2 ameliorates palmitate (PA)-induced insulin resistance, which was indicated by elevated phosphorylation of protein kinase B (AKT) and glycogen synthase kinase 3ß (GSK3ß). We also found that over-expression of PCBP2 inhibits HIF1α and STAT3 pathway. Furthermore, glucose uptake was found to display a similar tendency with the phosphorylation of Akt. The expressions of phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase), two key gluconeogenic enzymes, were down-regulated following Over-expression of PCBP2. Accordingly, PA-induced intracellular lipid accumulation was suppressed in over-expression of PCBP2 HepG2 cells. In addition, we found that over-expression of PCBP2 inhibits HIF1α and STAT3 pathway. Our results demonstrate that PCBP2 was involved in hepatic insulin sensitivity might via HIF-1α and STAT3 pathway in HepG2 cells.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Resistance/physiology , RNA-Binding Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Diet, High-Fat/adverse effects , Down-Regulation , Gluconeogenesis/drug effects , Glucose/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hep G2 Cells , Humans , Insulin Resistance/genetics , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Palmitic Acid/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Signal Transduction
17.
Brain Res ; 1615: 61-70, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-25912432

ABSTRACT

Zinc is an essential nutrient that is important for normal brain development. Zinc deficiency has been linked to aberrant neurological development and functioning. However, the molecular mechanisms underlying Zinc deficiency-induced neurological disorders remain largely elusive. In the present study, we showed that the proliferation of C17.2 neural stem cells (NSCs) was evidently impaired after exposed to low levels of Zinc chelator, N,N,N',N'-tetrakis-(2-pyridylmethy) ethylenediamine (TPEN). In addition, we found that TPEN-induced proliferative deficit of NSCs was related with significant downregulation of Wnt/ß-catenin signaling. Zinc deficiency impaired the proliferation of neural stem cells in dose- and time-dependent manners. Western blot revealed that the levels of p-Ser9-glycogensynthase kinase-3ß (p-GSK-3ß) and ß-catenin were remarkably downregulated during TPEN-induced C17.2 proliferative impairment. Moreover, immunofluorescent analysis indicated that the level of nuclear ß-catenin was apparently decreased following TPEN exposure. Furthermore, application with GSK-3ß inhibitor lithium chloride (LiCl) reversed TPEN-induced downregulation of ß-catenin and impairment of cell proliferation. Flow cytometry analysis also showed that TPEN-induced impairment of NSC proliferation could be reversed by LiCl. Taken together, these findings suggested that the disturbance of canonical Wnt/ß-catenin signaling pathway partially accounted for Zinc deficiency-induced proliferative impairment of NSCs.


Subject(s)
Cell Proliferation , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Wnt Signaling Pathway , Zinc/deficiency , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Down-Regulation , Ethylenediamines , Mice , Neural Stem Cells/drug effects , Signal Transduction/drug effects , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
18.
Biochem Biophys Res Commun ; 457(4): 578-84, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25600807

ABSTRACT

Excess serum free fatty acids (FFAs) are fundamental to the pathogenesis of insulin resistance. Chronic endoplasmic reticulum (ER) stress is a major contributor to obesity-induced insulin resistance in the liver. With high-fat feeding (HFD), FFAs can activate chronic endoplasmic reticulum (ER) stress in target tissues, initiating negative crosstalk between FFAs and insulin signaling. However, the molecular link between insulin resistance and ER stress remains to be identified. We here reported that translocating chain-associated membrane protein 1 (TRAM1), an ER-resident membrane protein, was involved in the onset of insulin resistance in hepatocytes. TRAM1 was significantly up-regulated in insulin-resistant liver tissues and palmitate (PA)-treated HepG2 cells. In addition, we showed that depletion of TRAM1 led to hyperactivation of CHOP and GRP78, and the activation of downstream JNK pathway. Given the fact that the activation of ER stress played a facilitating role in insulin resistance, the phosphorylation of Akt and GSK-3ß was also analyzed. We found that depletion of TRAM1 markedly attenuated the phosphorylation of Akt and GSK-3ß in the cells. Moreover, application with JNK inhibitor SP600125 reversed the effect of TRAM1 interference on Akt phosphorylation. The accumulation of lipid droplets and expression of two key gluconeogenic enzymes, PEPCK and G6Pase, were also determined and found to display a similar tendency with the phosphorylation of Akt. Glucose uptake assay indicated that knocking down TRAM1 augmented PA-induced down-regulation of glucose uptake, and inhibition of JNK using SP600125 could block the effect of TRAM1 on glucose uptake. These data implicated that TRAM1 might protect HepG2 cells against PA-induced insulin resistance through alleviating ER stress.


Subject(s)
Endoplasmic Reticulum Stress , Hep G2 Cells/metabolism , Insulin Resistance , MAP Kinase Signaling System , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Palmitates/metabolism , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Chaperone BiP , Glucose/metabolism , Humans
19.
Cell Mol Neurobiol ; 35(4): 463-71, 2015 May.
Article in English | MEDLINE | ID: mdl-25407628

ABSTRACT

RNA-binding motif protein, X-linked (RBMX) is a 43 kDa nuclear protein in the RBM family and functions on alternative splicing of RNA. The gene encoding RBMX is located on chromosome Xq26. To investigate whether RBMX is involved in retinal neuron apoptosis, we performed a light-induced retinal damage model in adult rats. Western blotting analysis showed RBMX gradually increased, reached a peak at 12 h and then declined during the following days. The association of RBMX in retinal ganglion cells (RGCs) with light exposure was found by immunofluorescence staining. The injury-induced expression of RBMX was detected in active caspase-3 and TUNEL positive cells. We also examined the expression profiles of active caspase-3, bcl-2 and Bax, whose changes were correlated with the expression profiles of RBMX. To summarize, we uncovered the dynamic changes of RBMX in the light-induced retinal damage model for the first time. RBMX might play a significant role in the degenerative process of RGCs after light-induced damage in the retina.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Light , Retina/metabolism , Retina/radiation effects , Animals , Apoptosis/radiation effects , Female , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Male , Protein Transport/radiation effects , Rats, Sprague-Dawley , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Staining and Labeling , bcl-2-Associated X Protein/metabolism
20.
J Appl Toxicol ; 35(7): 851-60, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25382668

ABSTRACT

2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD) is a ubiquitous environmental contaminant that could exert significant neurotoxicity in the human nervous system. Nevertheless, the molecular mechanism underlying TCDD-mediated neurotoxicity has not been clarified clearly. Herein, we investigated the potential role of TCDD in facilitating premature senescence in astrocytes and the underlying molecular mechanisms. Using the senescence-associated ß-galactosidase (SA-ß-Gal) assay, we demonstrated that TCDD exposure triggered significant premature senescence of astrocyte cells, which was accompanied by a marked activation of the Wingless and int (WNT)/ß-catenin signaling pathway. In addition, TCDD altered the expression of senescence marker proteins, such as p16, p21 and GFAP, which together have been reported to be upregulated in aging astrocytes, in both dose- and time-dependent manners. Further, TCDD led to cell-cycle arrest, F-actin reorganization and the accumulation of cellular reactive oxygen species (ROS). Moreover, the ROS scavenger N-acetylcysteine (NAC) markedly attenuated TCDD-induced ROS production, cellular oxidative damage and astrocyte senescence. Notably, the application of XAV939, an inhibitor of WNT/ß-catenin signaling pathway, ameliorated the effect of TCDD on cellular ß-catenin level, ROS production, cellular oxidative damage and premature senescence in astrocytes. In summary, our findings indicated that TCDD might induce astrocyte senescence via WNT/ß-catenin and ROS-dependent mechanisms.


Subject(s)
Astrocytes/drug effects , Cellular Senescence/drug effects , Dioxins/pharmacology , Reactive Oxygen Species/metabolism , Wnt Signaling Pathway/drug effects , Animals , Blotting, Western , Cell Cycle/drug effects , DNA Damage/drug effects , Dioxins/toxicity , Fluorescent Antibody Technique , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...