Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sensors (Basel) ; 24(12)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38931498

ABSTRACT

In recent years, with the increasing demand for high-quality images in various fields, more and more attention has been focused on noise removal techniques for image processing. The effective elimination of unwanted noise plays a crucial role in improving image quality. To meet this challenge, many noise removal methods have been proposed, among which the diffusion model has become one of the focuses of many researchers. In order to make the restored image closer to the real image and retain more features of the image, this paper proposes a DIR-SDE method with reference to the diffusion models of IR-SDE and IDM, which improve the feature retention of the image in the de-raining process, and then improve the realism of the image for the image de-raining task. In this study, IR-SDE was used as the base structure of the diffusion model, IR-SDE was improved, and DINO-ViT was combined to enhance the image features. During the diffusion process, the image features were extracted using DINO-ViT, and these features were fused with the original images to enhance the learning effect of the model. The model was also trained and validated with the Rain100H dataset. Compared with the IR-SDE method, it improved 0.003 in the SSIM, 0.003 in the LPIPS, and 1.23 in the FID. The experimental results show that the diffusion model proposed in this study can effectively improve the image restoration performance.

2.
Toxicol In Vitro ; 86: 105483, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36252918

ABSTRACT

Chlorpyrifos (CPF) is one of the most abundant and widely used organophosphate pesticides for agricultural, industrial, and household purposes in the world. Epidemiological studies have reported that CPF can induce neurotoxic impairments in mammalian, which is linked to an important risk factor for development of neurodegenerative diseases (NDs). However, limited information is available on CPF-induced neurotoxicity, with the underlying exact mechanism remains unclear. In this study, CPF exposure (10-400 µM) significantly reduced Neuro-2a cell viability and induced apoptotic events, including the increase in caspase-3 activity, apoptotic cell population, and cleavage of caspase-3/-7 and PARP. Exposure of Neuro-2a cells to CPF also triggered CHOP activation. Transfection with CHOP-specific siRNA markedly suppressed the expression of CHOP, and attenuated cytotoxicity and apoptotic events in CPF-exposed Neuro-2a cells. Furthermore, CPF exposure obviously evoked the phosphorylation of Akt as well as ROS generation in a time-dependent manner. Pretreatment with LY294002 (an Akt inhibitor) effectively attenuated the CPF-induced Akt phosphorylation, CHOP activation, and apoptotic events, but not that ROS production. Of note, buffering the ROS generation with antioxidant N-acetylcysteine effectively prevented the CPF-induced ROS generation, CHOP activation, and apoptotic events, but not that the Akt phosphorylation. Collectively, these findings indicate that CPF exposure exerts neuronal cytotoxicity via the independent pathways of ROS generation and Akt activation downstream-regulated CHOP-triggered apoptosis, ultimately leading to neuronal cell death.


Subject(s)
Chlorpyrifos , Animals , Chlorpyrifos/toxicity , Reactive Oxygen Species/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Caspase 3/metabolism , Oxidative Stress , Cell Death , Apoptosis , Mammals/metabolism
3.
Int J Mol Sci ; 23(22)2022 Nov 10.
Article in English | MEDLINE | ID: mdl-36430348

ABSTRACT

Cancers of the oral cavity can develop in the anatomic area extending from the lip, gum, tongue, mouth, and to the palate. Histologically, about 85-90% of oral cavity cancers are of the type squamous cells carcinomas (SCCs). The incidence of oral tongue SCC is higher in the tongue than any other anatomic area of the oral cavity. Here, we investigated the therapeutic effects and molecular mechanisms of docetaxel, which is a paclitaxel antitumor agent, on the cell growth of a human tongue SCC-derived SAS cell line. The results showed that docetaxel (10-300 nM) induced cytotoxicity and caspase-3 activity in SAS cells. Moreover, docetaxel (100 nM) promoted the expression of apoptosis-related signaling molecules, including the cleavages of caspase-3, caspase-7, and poly (ADP-ribose) polymerase (PARP). In mitochondria, docetaxel (100 nM) decreased the mitochondrial membrane potential (MMP) and Bcl-2 mRNA and protein expression and increased cytosolic cytochrome c protein expression and Bax mRNA and protein expression. In terms of mitogen-activated protein kinase (MAPK) and adenosine monophosphate-activated protein kinase (AMPK) signaling, docetaxel increased the expression of phosphorylated (p)-extracellular signal-regulated kinase (ERK), p-c-Jun N-terminal kinase (JNK), and p-AMPKα protein expression but not p-p38 protein expression. Moreover, the increase in caspase-3/-7 activity and Bax protein expression and decreased Bcl-2 protein expression and MMP depolarization observed in docetaxel-treated SAS cells could be reversed by treatment with either SP600125 (a JNK inhibitor), PD98059 (an MEK1/2 (mitogen-activated protein kinase kinase 1/2) inhibitor), or compound c (an AMPK inhibitor). The docetaxel-induced increases in p-JNK, p-ERK, and p-AMPKα protein expression could also be reversed by treatment with either SP600125, PD98059, or compound c. These results indicate that docetaxel induces human tongue SCC cell apoptosis via interdependent MAPK-JNK, MAPK-ERK1/2, and AMPKα signaling pathways. Our results show that docetaxel could possibly exert a potent pharmacological effect on human oral tongue SCC cell growth.


Subject(s)
Carcinoma, Squamous Cell , Tongue Neoplasms , Humans , Extracellular Signal-Regulated MAP Kinases/metabolism , Docetaxel/pharmacology , Caspase 3/metabolism , AMP-Activated Protein Kinases , Carcinoma, Squamous Cell/drug therapy , Tongue Neoplasms/drug therapy , Apoptosis , Proto-Oncogene Proteins c-bcl-2 , Epithelial Cells/metabolism , Tongue/metabolism , RNA, Messenger
4.
Int J Mol Sci ; 23(9)2022 Apr 23.
Article in English | MEDLINE | ID: mdl-35563057

ABSTRACT

Ketamine-associated cystitis is characterized by suburothelial inflammation and urothelial cell death. Norketamine (NK), the main metabolite of ketamine, is abundant in urine following ketamine exposure. NK has been speculated to exert toxic effects in urothelial cells, similarly to ketamine. However, the molecular mechanisms contributing to NK-induced urothelial cytotoxicity are almost unclear. Here, we aimed to investigate the toxic effects of NK and the potential mechanisms underlying NK-induced urothelial cell injury. In this study, NK exposure significantly reduced cell viability and induced apoptosis in human urinary bladder epithelial-derived RT4 cells that NK (0.01-0.5 mM) exhibited greater cytotoxicity than ketamine (0.1-3 mM). Signals of mitochondrial dysfunction, including mitochondrial membrane potential (MMP) loss and cytosolic cytochrome c release, were found to be involved in NK-induced cell apoptosis and death. NK exposure of cells also triggered the expression of endoplasmic reticulum (ER) stress-related proteins including GRP78, CHOP, XBP-1, ATF-4 and -6, caspase-12, PERK, eIF-2α, and IRE-1. Pretreatment with 4-phenylbutyric acid (an ER stress inhibitor) markedly prevented the expression of ER stress-related proteins and apoptotic events in NK-exposed cells. Additionally, NK exposure significantly activated JNK, ERK1/2, and p38 signaling and increased intracellular calcium concentrations ([Ca2+]i). Pretreatment of cells with both PD98059 (an ERK1/2 inhibitor) and BAPTA/AM (a cell-permeable Ca2+ chelator), but not SP600125 (a JNK inhibitor) and SB203580 (a p38 inhibitor), effectively suppressed NK-induced mitochondrial dysfunction, ER stress-related signals, and apoptotic events. The elevation of [Ca2+]i in NK-exposed cells could be obviously inhibited by BAPTA/AM, but not PD98059. Taken together, these findings suggest that NK exposure exerts urothelial cytotoxicity via a [Ca2+]i-regulated ERK1/2 activation, which is involved in downstream mediation of the mitochondria-dependent and ER stress-triggered apoptotic pathway, consequently resulting in urothelial cell death. Our findings suggest that regulating [Ca2+]i/ERK signaling pathways may be a promising strategy for treatment of NK-induced urothelial cystitis.


Subject(s)
Cystitis , Ketamine , Apoptosis , Endoplasmic Reticulum Stress , Female , Humans , Ketamine/analogs & derivatives , Ketamine/pharmacology , MAP Kinase Signaling System , Male , Mitochondria/metabolism
5.
Neurotoxicology ; 85: 133-144, 2021 07.
Article in English | MEDLINE | ID: mdl-34038756

ABSTRACT

Inorganic arsenic (As3+), a well-known worldwide industrial and environmental pollutant, has been linked to neurodegenerative disorders (NDs). Autophagy plays an important role in controlling neuronal cell survival/death. However, limited information is available regarding the toxicological mechanism at the interplay between autophagy and As3+-induced neurotoxicity. The present study found that As3+ exposure induced a concomitant activation of apoptosis and autophagy in Neuro-2a cells, which was accompanied with the increase of phosphatidylserine exposure on outer membrane leaflets and apoptotic cell population, and the activation of caspase-3, -7, and PARP as well as the elevation of protein expressions of LC3-II, Atg-5, and Beclin-1, and the accumulation of autophagosome. Pretreatment of cells with autophagy inhibitor 3-MA, but not that of Z-VAD-FMK (a pan-caspase inhibitor), effectively prevented the As3+-induced autophagic and apoptotic responses, indicating that As3+-triggered autophagy was contributing to neuronal cell apoptosis. Furthermore, As3+ exposure evoked the dephosphorylation of Akt. Pretreatment with SC79, an Akt activator, could significantly attenuated As3+-induced Akt inactivation as well as autophagic and apoptotic events. Expectedly, inhibition of Akt signaling with LY294002 obviously enhanced As3+-triggered autophagy and apoptosis. Exposure to As3+ also dramatically increased the phosphorylation level of AMPKα. Pretreatment of AMPK inhibitor (Compound C) could markedly abrogate the As3+-induced phosphorylated AMPKα expression, and autophagy and apoptosis activation. Taken together, these results indicated that As3+ exerted its cytotoxicity in neuronal cells via the Akt inactivation/AMPK activation downstream-regulated autophagy-dependent apoptosis pathways, which ultimately lead to cell death. Our findings suggest that the regulation of Akt/AMPK signals may be a promising intervention to against As3+-induced neurotoxicity and NDs.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Arsenic/toxicity , Autophagy/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/physiology , Autophagy/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Mice , Neurons/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
6.
Toxicol In Vitro ; 63: 104739, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31756540

ABSTRACT

Human exposure to silica nanoparticles (SiNPs) has been widely applied as vehicles for drug delivery and cellular manipulations in nanoneuromedicine. SiNPs may cause adverse effects in the brain, but potential mechanisms underlying SiNPs-induced neurotoxicity are remained unclear. Here, we examined cytotoxic effects and the cellular mechanisms of SiNPs-induced neuronal cell death. In this study, the results showed that SiNPs significantly decreased cell viability and induced apoptosis in Neuro-2a cells as evidenced by the increase caspase-3 activity and the activation of caspase cascades and poly (ADP-ribose) polymerase (PARP). In addition, endoplasmic reticulum (ER) stress was triggered as indicated by several key molecules including glucose-regulated protein (GRP)78 and 94, C/EBP homologous protein (CHOP), activation transcription factor (ATF)-4, and caspase-12. Pretreatment of Neuro-2a cells with specific pharmacological inhibitor of ER stress (4-phenylbutyric acid (4-PBA)) effectively alleviated the SiNPs-induced ER stress and apoptotic related signals. Furthermore, 2',7'-Dichlorofluorescein fluorescence as an indicator of reactive oxygen species (ROS) formation after exposure of Neuro-2a cells to SiNPs significantly increased ROS levels. Antioxidant N-acetylcyseine (NAC) effectively reversed SiNPs-induced cellular responses. Taken together, these results suggest that SiNPs exposure exerts its neurotoxicity in cultured neuronal cells by inducing apoptosis via a ROS generation-activated downstream ER stress signaling pathway.


Subject(s)
Nanoparticles/toxicity , Neurons/drug effects , Silicon Dioxide/toxicity , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Mice , Neurons/metabolism , Reactive Oxygen Species/metabolism
7.
Front Microbiol ; 6: 1508, 2015.
Article in English | MEDLINE | ID: mdl-26779163

ABSTRACT

Bamboo mosaic virus (BaMV) has a 6.4-kb (+) sense RNA genome with a 5' cap and a 3' poly(A) tail. ORF1 of this potexvirus encodes a 155-kDa replication protein responsible for the viral RNA replication/transcription and 5' cap formation. To learn more about the replication complex of BaMV, a protein preparation enriched in the 155-kDa replication protein was obtained from Nicotiana benthamiana by a protocol involving agroinfiltration and immunoprecipitation. Subsequent analysis by SDS-PAGE and mass spectrometry identified a handful of host proteins that may participate in the viral replication. Among them, the cytoplasmic exoribonuclease NbXRN4 particularly caught our attention. NbXRN4 has been shown to have an antiviral activity against Tomato bushy stunt virus and Tomato mosaic virus. In Arabidopsis, the enzyme could reduce RNAi- and miRNA-mediated RNA decay. This study found that downregulation of NbXRN4 greatly decreased BaMV accumulation, while overexpression of NbXRN4 resulted in an opposite effect. Mutations at the catalytically essential residues abolished the function of NbXRN4 in the increase of BaMV accumulation. Nonetheless, NbXRN4 was still able to promote BaMV accumulation in the presence of the RNA silencing suppressor P19. In summary, the replication efficiency of BaMV may be improved by the exoribonuclease activity of NbXRN4.

SELECTION OF CITATIONS
SEARCH DETAIL
...