Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Environ Toxicol ; 38(12): 2836-2844, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37584494

RESUMEN

BACKGROUND: Formaldehyde (FA) is associated with the occurrence of leukemia, and oxidative stress is considered to be a major reason. As an endogenous biomarker of oxidative stress, few studies focus on the relationship between peroxiredoxin III (PrxIII) and FA toxicity. Our previous research observed high expression of PrxIII occurred in the process of apoptosis of bone marrow cells (BMCs) induced by FA, however the exact mechanism is unclear. Therefore, this paper aimed to explore the possible association between FA toxicity and PrxIII gene. METHODS: We first, used a Cell Counting Kit-8 (CCK-8) to detect the viability of BMCs after they were exposed to different doses of FA (50, 100, 200 µmol/L) for different exposure time (12, 24, 48 h), then chose 24 h as an exposure time to detect the expression of PrxIII for exposing different doses of FA by Quantitative reverse transcription-PCR (qRT-PCR) and Western blot analysis. Based on our preliminary experimental results, we chose 100 µmol/L FA as an exposure dose to expose for 24 h, and used a small interfering RNA (siRNA) to silenced PrxIII to examine the cell viability by CCK-8, reactive oxygen species (ROS) level by DCFH-DA, apoptosis by Annexin V/PI double staining and cell cycle by flow cytometry (FCM) so as to explore the possible regulatory effect of PrxIII silencing on FA-induced bone marrow toxicity. RESULTS: High expression of PrxIII occurred in the process of FA-induced oxidative stress. Silencing of PrxIII prevented FA from inducing oxidative stress, thus increasing cell viability, decreasing ROS level, rescuing G0 -G1 and G2 -M arrest, and reducing cell apoptosis. CONCLUSION: PrxIII silencing might be a potential target for alleviating FA-induced oxidative damage.


Asunto(s)
Estrés Oxidativo , Peroxiredoxina III , Animales , Ratones , Peroxiredoxina III/metabolismo , Peroxiredoxina III/farmacología , Especies Reactivas de Oxígeno/metabolismo , Ratones Endogámicos BALB C , Estrés Oxidativo/genética , Formaldehído/toxicidad , Apoptosis/genética , Células de la Médula Ósea
2.
Brain Res Bull ; 193: 95-105, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36566946

RESUMEN

Subarachnoid hemorrhage (SAH), a type of hemorrhagic stroke, is a neurological emergency associated with a high morbidity and mortality rate. After SAH, early brain injury (EBI) is the leading cause of poor prognosis in SAH patients. Peroxiredoxins (PRDXs) are a family of sulphhydryl-dependent peroxidases. Peroxiredoxin-3 (PRDX3) is mainly located in the mitochondria of neurons, which can remove hydrogen peroxide (H2O2); however, the effect of PRDX3 on EBI after SAH remains unclear. In this study, an endovascular perforation model was used to mimic SAH in Sprague Dawley rats in vivo. The results revealed that after SAH, PRDX3 levels decreased in the neurons. PRDX3 overexpression by neuron-specific adeno-associated viruses upregulated PRDX3 levels. Furthermore, PRDX3 overexpression improved long- and short-term behavioral outcomes and alleviated neuronal impairment in rats. Nissl staining revealed that the upregulation of PRDX3 promoted cortical neuron survival. PRDX3 overexpression decreased the H2O2 content and downregulated caspase-9 expression. In conclusion, PRDX3 participates in neuronal protection by inhibiting the neuronal mitochondria-mediated death pathway; PRDX3 may be an important target for EBI intervention after SAH.


Asunto(s)
Lesiones Encefálicas , Fármacos Neuroprotectores , Hemorragia Subaracnoidea , Animales , Ratas , Apoptosis , Lesiones Encefálicas/metabolismo , Peróxido de Hidrógeno/farmacología , Neuroprotección , Fármacos Neuroprotectores/farmacología , Peroxiredoxina III/farmacología , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/metabolismo
3.
Front Cell Infect Microbiol ; 12: 907043, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35873171

RESUMEN

Trypanosoma cruzi, the causal agent of Chagas disease, has peroxiredoxins (PRXs) expressed in all stages of the parasite and whose function is to detoxify oxidizing agents, such as reactive oxygen species (ROS). These proteins are central for the survival and replication of the parasite and have been proposed as virulence factors. Because of their importance, they have also been considered as possible therapeutic targets, although there is no specific drug against them. One of them, the mitochondrial PRX (TcMPX), is important in the detoxification of ROS in this organelle and has a role in the infectivity of T. cruzi. However, their structural characteristics are unknown, and possible inhibitors have not been proposed. The aim was to describe in detail some structural characteristics of TcMPX and compare it with several PRXs to find possible similarities and repositioning the antibiotic Thiostrepton as a potential inhibitor molecule. It was found that, in addition to the characteristic active site of a 2-cys PRX, this protein has a possible transmembrane motif and motifs involved in resistance to hyper oxidation. The homology model suggests a high structural similarity with human PRX3. This similarity was corroborated by cross-recognition using an anti-human PRX antibody. In addition, molecular docking showed that Thiostrepton, a potent inhibitor of human PRX3, could bind to TcMPX and affect its function. Our results show that Thiostrepton reduces the proliferation of T. cruzi epimastigotes, cell-derived trypomastigotes, and blood trypomastigotes with low cytotoxicity on Vero cells. We also demonstrated a synergic effect of Thriostepton and Beznidazol. The convenience of seeking treatment alternatives against T. cruzi by repositioning compounds as Thiostrepton is discussed.


Asunto(s)
Enfermedad de Chagas , Trypanosoma cruzi , Animales , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/parasitología , Chlorocebus aethiops , Humanos , Simulación del Acoplamiento Molecular , Peroxiredoxina III/metabolismo , Peroxiredoxina III/farmacología , Peroxiredoxina III/uso terapéutico , Peroxirredoxinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tioestreptona/metabolismo , Tioestreptona/farmacología , Tioestreptona/uso terapéutico , Trypanosoma cruzi/metabolismo , Células Vero
4.
Neuroscience ; 487: 88-98, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35026318

RESUMEN

The experimental investigations on the pathogenesis of remifentanil-induced hyperalgesia (RIH) have been primarily conducted, but the effective treatment of RIH remains unclear. Recent reports highlight the necessity of ionotropic glutamate receptors in oxidative damage in spinal nociceptive transduction. Artesunate, the 1st-line anti-malaria drug, has been identified to be valid in removing superoxide in several pathological conditions. This study evaluated whether artesunate inhibits RIH via regulating metabotropic glutamate receptor 5 (mGluR5) and mitochondrial antioxidant enzyme peroxiredoxin-3 in rats. Artesunate was injected intrathecally 10 min before intravenous infusion of remifentanil (1 µg·kg-1·min-1 for 60 min) in rats. The antinociception of artesunate was verified by assessment of paw withdrawal mechanical threshold and paw withdrawal thermal latency. Spinal mGluR5 expression and peroxiredoxin-3 hyperacetylation were examined. Also, both the mGluR5 agonist DHPG and antagonist MPEP were utilized to explore the involvement of mGluR5 in the anti-hyperalgesic property of artesunate. Here, we found that artesunate (10 µg and 100 µg but not 1 µg) prevented RIH in a dose-dependent manner. Artesunate reduced remifentanil-related spinal over-expression of mGluR5 gene and protein, and hyperacetylation of peroxiredoxin-3. Intrathecal application of MPEP (10 nmol and 100 nmol but not 1 nmol) inhibited behavioral RIH and peroxiredoxin-3 acetylation. Moreover, hyperalgesia and peroxiredoxin-3 hyperacetylation were attenuated after the combination of artesunate (1 µg) and MPEP (1 nmol). Additionally, artesunate treatment reversed acute pain and peroxiredoxin-3 hyperacetylation following spinal exposure to DHPG. In conclusion, intrathecal injection of artesunate impairs RIH by down-regulating spinal mGluR5 expression and peroxiredoxin-3 hyperacetylation-mediated oxidative stress in rats.


Asunto(s)
Hiperalgesia , Receptor del Glutamato Metabotropico 5 , Animales , Artesunato/metabolismo , Artesunato/uso terapéutico , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Peroxiredoxina III/metabolismo , Peroxiredoxina III/farmacología , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5/metabolismo , Remifentanilo/efectos adversos , Médula Espinal/metabolismo
5.
J Cancer Res Ther ; 18(7): 1994-2000, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36647961

RESUMEN

Objective: This study aims to investigate peroxiredoxin 3 (PRDX3) expression in gastric cancer tissue and its effects on cisplatin resistance in gastric cancer cells and its possible mechanism. Methods: PRDX3 expression in human gastric cancer tissue microarrays was detected via immunohistochemistry. The PRDX3 small interfering RNA (siPRDX3 group) and the negative control siNC (siNC group) were transfected into AGS and MKN-74 cell lines, respectively, whereas a blank control group was set up. Each group was treated with different cisplatin concentrations (0, 5, 10, 15, 20, 25, and 30 µg/ml), and the half-inhibitory concentration (IC50) of each group of the two cell lines was calculated using the CCK8 assay. The corresponding IC50 concentration of the siPRDX3 group in the two cell lines was used to treat cells of each group. Flow cytometry was used to detect cell apoptosis, and Western blotting was used to detect the expression levels of cleaved caspase-3 and Bax in each group. Results: PRDX3 was overexpressed in gastric adenocarcinoma tissue compared with adjacent noncancer tissue (P = 0.0053). After cisplatin treatment, the IC50 in the siPRDX3 group of AGS cells (5.91 ± 0.18 µg/ml) and the siPRDX3 group of MKN-74 cells (3.48 ± 0.30 µg/ml) was significantly lower than in the corresponding siNC groups (10.01 ± 0.99 and 6.39 ± 0.70 µg/ml; P = 0.0022 and 0.0027, respectively). AGS cells (38.81% ± 1.69%) and MKN-74 cells (25.03% ± 2.80%) in the siPRDX3 group showed significantly higher apoptosis rates than in the corresponding siNC groups (23.17% ± 1.43% and 16.7% ± 1.39%; P = 0.0003 and 0.0099, respectively). The expression levels of cleaved caspase-3 and Bax were significantly higher in the siPRDX3 group of both cell lines than in the siNC group (P < 0.0001). Conclusion: PRDX3 increases the gastric cancer cell resistance to cisplatin by reducing apoptosis and thus may serve as a target to overcome cisplatin resistance.


Asunto(s)
Cisplatino , Neoplasias Gástricas , Humanos , Cisplatino/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Caspasa 3/genética , Peroxiredoxina III/genética , Peroxiredoxina III/metabolismo , Peroxiredoxina III/farmacología , Proteína X Asociada a bcl-2/genética , Apoptosis , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Proliferación Celular
6.
Oxid Med Cell Longev ; 2019: 2828493, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31636803

RESUMEN

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a lipid and protein phosphatase that coordinates various cellular processes. Its activity is regulated by the reversible oxidation of an active-site cysteine residue by H2O2 and thioredoxin. However, the potential role of lipid peroxides in the redox regulation of PTEN remains obscure. To evaluate this, 15-hydroperoxy-eicosatetraenoic acid (15s-HpETE), a lipid peroxide, was employed to investigate its effect on PTEN using molecular and cellular-based assays. Exposure to 15s-HpETE resulted in the oxidation of recombinant PTEN. Reversible oxidation of PTEN was also observed in mouse embryonic fibroblast (MEF) cells treated with a 15s-HpETE and Lipofectamine mixture. The oxidative dimerization of thioredoxin was found simultaneously. In addition, the absence of peroxiredoxin III aggravated 15s-HpETE-induced PTEN oxidation in MEF cells. Our study provides novel insight into the mechanism linking lipid peroxidation to the etiology of tumorigenesis.


Asunto(s)
Leucotrienos/uso terapéutico , Peróxidos Lipídicos/uso terapéutico , Fosfohidrolasa PTEN/efectos de los fármacos , Peroxiredoxina III/uso terapéutico , Animales , Humanos , Leucotrienos/farmacología , Peróxidos Lipídicos/farmacología , Ratones , Oxidación-Reducción , Peroxiredoxina III/farmacología , Transfección
7.
J Invest Dermatol ; 137(6): 1333-1342, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28202400

RESUMEN

UVB light induces generation of reactive oxygen species, ultimately leading to skin cell damage. Mitochondria are a major source of reactive oxygen species in UVB-irradiated skin cells, with increased levels of mitochondrial reactive oxygen species having been implicated in keratinocyte apoptosis. Peroxiredoxin III (PrxIII) is the most abundant and potent H2O2-removing enzyme in the mitochondria of most cell types. Here, the protective role of PrxIII against UVB-induced apoptosis of epidermal keratinocytes was investigated. Mitochondrial H2O2 levels were differentiated from other types of ROS using mitochondria-specific fluorescent H2O2 indicators. Upon UVB irradiation, PrxIII-knockdown HaCaT human keratinocytes and PrxIII-deficient (PrxIII-/-) mouse primary keratinocytes exhibited enhanced accumulation of mitochondrial H2O2 compared with PrxIII-expressing controls. Keratinocytes lacking PrxIII were subsequently sensitized to apoptosis through mitochondrial membrane potential loss, cardiolipin oxidation, cytochrome c release, and caspase activation. Increased UVB-induced epidermal tissue damage in PrxIII-/- mice was attributable to increased caspase-dependent keratinocyte apoptosis. Our findings show that mitochondrial H2O2 is a key mediator in UVB-induced apoptosis of keratinocytes and that PrxIII plays a critical role in protecting epidermal keratinocytes against UVB-induced apoptosis through eliminating mitochondrial H2O2. These findings support the concept that reinforcing mitochondrial PrxIII defenses may help prevent UVB-induced skin damage such as inflammation, sunburn, and photoaging.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Peróxido de Hidrógeno/metabolismo , Queratinocitos/efectos de la radiación , Peroxiredoxina III/farmacología , Animales , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Células Epidérmicas , Epidermis/metabolismo , Epidermis/efectos de la radiación , Humanos , Queratinocitos/citología , Potencial de la Membrana Mitocondrial/efectos de la radiación , Ratones , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/efectos de la radiación , Rayos Ultravioleta/efectos adversos
8.
J Proteome Res ; 15(5): 1506-14, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-26983019

RESUMEN

PRDX3 is a mitochondrial peroxide reductase that regulates cellular redox state. It has been reported that PRDX3 is overexpressed in liver cancer, but how PRDX3 is involved in hepatocellular carcinoma (HCC) tumorigenesis and progression has not been well-characterized. In the present study, we established two stable cell lines by overexpressing or knocking down PRDX3 in HepG2 cells. We found that PRDX3 silencing decreased the growth rate of HepG2 cells and increased mtDNA oxidation. Quantitative proteomics identified 475 differentially expressed proteins between the PRDX3 knockdown and the control cells. These proteins were involved in antioxidant activity, angiogenesis, cell adhesion, cell growth, ATP synthesis, nucleic acid binding, redox, and chaperones. PRDX3 knockdown led to the down-regulation of ATP synthases and the decreased cellular ATP level, contributing to the slow-down of cell growth. Furthermore, silencing PRDX3 enhanced invasive properties of HepG2 cells via TIMP-1 down-regulation and the increased ECM degradation. Taken together, our results indicate that PRDX3 promotes HCC growth and mediates cell migration and invasiveness and is a potential therapeutic target for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Peroxiredoxina III/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Silenciador del Gen , Células Hep G2 , Humanos , Invasividad Neoplásica , Peroxiredoxina III/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...