Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Int J Biol Sci ; 20(5): 1707-1728, 2024.
Article in English | MEDLINE | ID: mdl-38481805

ABSTRACT

Acute pancreatitis (AP) is a common abdominal disease that typically resolves on its own, but the mortality rate dramatically increases when it progresses to severe acute pancreatitis (SAP). In this study, we investigated the molecular mechanism underlying the development of SAP from AP. We utilized two SAP models induced by pancreatic duct ligation and caerulein administration. Transcriptomic and proteomic analyses were subsequently performed to determine the mRNA and protein expression profiles of pancreatic samples from SAP and AP model and normal mice. To explore the role of Hspb1 in SAP, we used Hspb1 knockout (KO) mice, a genetically engineered chronic pancreatitis strain (T7D23A), Anxa2 KO mice, and acinar cell-specific Prdx1 knockout mice. Additionally, various in vivo and in vitro assays were performed to elucidate the molecular events and direct targets of Hspb1 in acinar cells. We found that Hspb1 expression was upregulated in AP samples but significantly reduced in acinar cells from SAP samples. KO or inhibition of Hspb1 worsened AP, while AAV8-Hspb1 administration mitigated the severity of SAP and reduced remote organ damage in mice. Furthermore, AAV8-Hspb1 treatment prevented the development of chronic pancreatitis. We found that KO or inhibition of Hspb1 promoted acinar cell death through apoptosis and ferroptosis but not necroptosis or autophagy by increasing reactive oxygen species (ROS) and lipid ROS levels. Mechanistically, Hspb1 directly interacted with Anxa2 to decrease its aggregation and phosphorylation, interact with the crucial antioxidant enzyme Prdx1, and maintain its antioxidative activity by decreasing Thr-90 phosphorylation. Notably, the overexpression of Hspb1 did not have a protective effect on acinar-specific Prdx1 knockout mice. In summary, our findings shed light on the role of Hspb1 in acinar cells. We showed that targeting Hspb1/Anxa2/Prdx1 could serve as a potential therapeutic strategy for SAP.


Subject(s)
Ferroptosis , Pancreatitis, Chronic , Animals , Mice , Acute Disease , Antioxidants/pharmacology , Apoptosis/genetics , Mice, Knockout , Peroxiredoxins/genetics , Peroxiredoxins/pharmacology , Proteomics , Reactive Oxygen Species
2.
CNS Neurosci Ther ; 30(3): e14681, 2024 03.
Article in English | MEDLINE | ID: mdl-38516845

ABSTRACT

BACKGROUND: Peroxiredoxin 2 (Prx2), an intracellular protein that regulates redox reactions, released from red blood cells is involved in inflammatory brain injury after intracerebral hemorrhage (ICH). Toll-like receptor 4 (TLR4) may be crucial in this process. This study investigated the role of the Prx2-TLR4 inflammatory axis in brain injury following experimental ICH in mice. METHODS: First, C57BL/6 mice received an intracaudate injection of autologous arterial blood or saline and their brains were harvested on day 1 to measure Prx2 levels. Second, mice received an intracaudate injection of either recombinant mouse Prx2 or saline. Third, the mice were co-injected with autologous arterial blood and conoidin A, a Prx2 inhibitor, or vehicle. Fourth, the mice received a Prx2 injection and were treated with TAK-242, a TLR4 antagonist, or saline (intraperitoneally). Behavioral tests, magnetic resonance imaging, western blot, immunohistochemistry/immunofluorescence staining, and RNA sequencing (RNA-seq) were performed. RESULTS: Brain Prx2 levels were elevated after autologous arterial blood injection. Intracaudate injection of Prx2 caused brain swelling, microglial activation, neutrophil infiltration, neuronal death, and neurological deficits. Co-injection of conoidin A attenuated autologous arterial blood-induced brain injury. TLR4 was expressed on the surface of microglia/macrophages and neutrophils and participated in Prx2-induced inflammation. TAK-242 treatment attenuated Prx2-induced inflammation and neurological deficits. CONCLUSIONS: Prx2 can cause brain injury following ICH through the TLR4 pathway, revealing the Prx2-TLR4 inflammatory axis as a potential therapeutic target.


Subject(s)
Brain Injuries , Sulfonamides , Toll-Like Receptor 4 , Animals , Mice , Brain Injuries/etiology , Cerebral Hemorrhage/metabolism , Inflammation/etiology , Inflammation/pathology , Mice, Inbred C57BL , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Peroxiredoxins/therapeutic use , Toll-Like Receptor 4/metabolism
3.
Plant Physiol Biochem ; 201: 107876, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37413942

ABSTRACT

Plant 2-cysteine peroxiredoxin (2-Cys Prx) is a mercaptan peroxidase localized in chloroplasts and has unique catalytic properties. To explore the salt stress tolerance mechanisms of 2-Cys Prx in plants, we analyzed the effects of overexpressing the 2-CysPrx gene on the physiological and biochemical metabolic processes of tobacco under NaHCO3 stress through joint physiological and transcriptomic analysis. These parameters included growth phenotype, chlorophyll, photosynthesis, and antioxidant system. After NaHCO3 stress treatment, a total of 5360 differentially expressed genes (DEGs) were identified in 2-Cysprx overexpressed (OE) plants, and the number of DEGs was significantly lower than 14558 in wild-type (WT) plants. KEGG enrichment analysis showed that DEGs were mainly enriched in photosynthetic pathways, photosynthetic antenna proteins, and porphyrin and chlorophyll metabolism. Overexpressing 2-CysPrx significantly reduced the growth inhibition of tobacco induced by NaHCO3 stress, alleviating the down-regulation of the DEGs related to chlorophyll synthesis, photosynthetic electron transport and the Calvin cycle and the up-regulation of those related to chlorophyll degradation. In addition, it also interacted with other redox systems such as thioredoxins (Trxs) and the NADPH-dependent Trx reductase C (NTRC), and mediated the positive regulation of the activities of antioxidant enzymes such as peroxidase (POD) and catalase (CAT) and the expression of related genes, thereby reducing the accumulation of superoxide anion (O2·-), hydrogen peroxide (H2O2) and malondialdehyde (MDA). In conclusion, 2-CysPrx overexpression could alleviate the NaHCO3 stress-induced photoinhibition and oxidative damage by regulating chlorophyll metabolism, promoting photosynthesis and participating in the regulation of antioxidant enzymes, and thus improve the ability of plants to resist salt stress damage.


Subject(s)
Antioxidants , Peroxiredoxins , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Nicotiana/genetics , Nicotiana/metabolism , Hydrogen Peroxide/metabolism , Cysteine/metabolism , Photosynthesis , Oxidoreductases/metabolism , Peroxidase/metabolism , Chlorophyll
4.
Cells Dev ; 175: 203860, 2023 09.
Article in English | MEDLINE | ID: mdl-37270067

ABSTRACT

Peroxiredoxins (Prdxs) are thiol-dependent enzymes that scavenge peroxides. Previously, we found that Prdxs were hyperoxidized in a Parkinson's disease model induced by paraquat (PQ), which led to their inactivation, perpetuating reactive oxygen species (ROS) formation. Herein, we evaluated the redox state of the typical 2-Cys-Prx subgroup. We found that PQ induces ROS compartmentalization in different organelles, reflected by the 2-Cys-Prdx hyperoxidation pattern detected by redox eastern blotting. 2-Cys Prdxs are most vulnerable to hyperoxidation, while atypical 2-Cys Peroxiredoxin 5 (Prdx5) is resistant and is expressed in multiple organelles, such as mitochondria, peroxisomes, and cytoplasm. Therefore, we overexpressed human Prdx5 in the dopaminergic SHSY-5Y cell line using the adenoviral vector Ad-hPrdx5. Prdx5 overexpression was confirmed by western blotting and immunofluorescence (IF) and effectively decreased PQ-mediated mitochondrial and cytoplasmic ROS assessed with a mitochondrial superoxide indicator and DHE through IF or flow cytometry. Decreased ROS mediated by Prdx5 in the main subcellular compartments led to overall cell protection against PQ-induced cell death, which was demonstrated by flow cytometry using Annexin V labeling and 7-AAD. Therefore, Prdx5 is an attractive therapeutic target for PD, as its overexpression protects dopaminergic cells from ROS and death, which warrants further experimental animal studies for its subsequent application in clinical trials.


Subject(s)
Oxidative Stress , Paraquat , Animals , Humans , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Paraquat/pharmacology , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Cell Death/genetics
5.
Brain Res ; 1813: 148428, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37263551

ABSTRACT

Protracted opioid withdrawal is considered to be a traumatic event with many adverse effects. However, little attention is paid to its consequences on the protein expression in the rat brain. A better understanding of the changes at the molecular level is essential for designing future innovative drug therapies. Our previous proteomic data indicated that long-term morphine withdrawal is associated with altered proteins functionally involved in energy metabolism, cytoskeletal changes, oxidative stress, apoptosis, or signal transduction. In this study, we selected peroxiredoxin II (PRX II) as a marker of oxidative stress, 14-3-3 proteins as adaptors, and creatine kinase-B (CK-B) as a marker of energy metabolism to detect their amounts in the brain cortex and hippocampus isolated from rats after 3-month (3 MW) and 6-month morphine withdrawal (6 MW). Methodically, our work was based on immunoblotting accompanied by 2D resolution of PRX II and 14-3-3 proteins. Our results demonstrate significant upregulation of PRX II in the rat brain cortex (3-fold) and hippocampus (1.3-fold) after 3-month morphine abstinence, which returned to the baseline six months since the drug was withdrawn. Interestingly, the level of 14-3-3 proteins was downregulated in both brain areas in 3 MW samples and remained decreased only in the brain cortex of 6 MW. Our findings suggest that the rat brain cortex and hippocampus exhibit the oxidative stress-induced vulnerability represented by compensatory upregulation of PRX II after three months of morphine withdrawal.


Subject(s)
Morphine Dependence , Substance Withdrawal Syndrome , Rats , Animals , Morphine/metabolism , 14-3-3 Proteins/metabolism , Up-Regulation , Proteomics , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Hippocampus/metabolism , Brain/metabolism , Substance Withdrawal Syndrome/metabolism
6.
J Assist Reprod Genet ; 40(7): 1573-1587, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37227568

ABSTRACT

PURPOSE: PE is a pregnancy-specific syndrome and one of the main causes of maternal, fetal, and neonatal mortality. PRDX1 is an antioxidant that regulates cell proliferation, differentiation, and apoptosis. The aim of this study is to investigate the effect of PRDX1 on the regulation of trophoblast function by affecting autophagy and oxidative stress in preeclampsia. METHODS: Western blotting, RT-qPCR, and immunofluorescence were used to examine the expression of PRDX1 in placentas. PRDX1-siRNA was transfected to knockdown PRDX1 in HTR-8/SVneo cells. The biological function of HTR-8/SVneo cells was detected by wound healing, invasion, tube formation, CCK-8, EdU, flow cytometry, and TUNEL assays. Western blotting was used to detect the protein expression of cleaved-Caspase3, Bax, LC3II, Beclin1, PTEN, and p-AKT. DCFH-DA staining was used to detect ROS levels by flow cytometry. RESULTS: PRDX1 was significantly decreased in placental trophoblasts in PE patients. Following the exposure of HTR-8/SVneo cells to H2O2, PRDX1 expression was significantly decreased, LC3II and Beclin1 expression was notably increased, and ROS level was also markedly increased. PRDX1 knockdown impaired migration, invasion, and tube-formation abilities and promoted apoptosis, which was accompanied by an increased expression of cleaved-Caspase3 and Bax. PRDX1 knockdown induced a significant decrease in LC3II and Beclin1 expression, along with an elevated p-AKT expression and a decreased PTEN expression. PRDX1 knockdown increased intracellular ROS levels, and NAC attenuated PRDX1 knockdown-induced apoptosis. CONCLUSION: PRDX1 regulated trophoblast function through the PTEN/AKT signaling pathway to affect cell autophagy and ROS level, which provided a potential target for the treatment of PE.


Subject(s)
Pre-Eclampsia , Trophoblasts , Infant, Newborn , Humans , Pregnancy , Female , Trophoblasts/metabolism , Placenta/metabolism , Cell Line , Proto-Oncogene Proteins c-akt/genetics , bcl-2-Associated X Protein , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Beclin-1/metabolism , Beclin-1/pharmacology , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Reactive Oxygen Species/metabolism , Cell Proliferation , Oxidative Stress/genetics , Autophagy/genetics , Apoptosis
7.
Biosci Biotechnol Biochem ; 87(7): 696-706, 2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37024271

ABSTRACT

Obesity is caused by the accumulation of excess lipids due to an energy imbalance. Differentiation of pre-adipocytes induces abnormal lipid accumulation, and reactive oxygen species (ROS) generated in this process promote the differentiation of pre-adipocytes through mitogen-activated protein kinase (MAPK) signaling. Peroxiredoxin (Prx) is a potent antioxidant enzyme, and peroxiredoxin 5 (Prx5), which is mainly expressed in cytosol and mitochondria, inhibits adipogenesis by regulating ROS levels. Based on previous findings, the present study was performed to investigate whether cytosolic Prx5 (CytPrx5) or mitochondrial Prx5 (MtPrx5) has a greater effect on the inhibition of adipogenesis. In this study, MtPrx5 decreased insulin-mediated ROS levels to reduce adipogenic gene expression and lipid accumulation more effectively than CytPrx5. In addition, we found that p38 MAPK mainly participates in adipogenesis. Furthermore, we verified that MtPrx5 overexpression suppressed the phosphorylation of p38 during adipogenesis. Thus, we suggest that MtPrx5 inhibits insulin-induced adipogenesis more effectively than CytPrx5.


Subject(s)
Adipogenesis , Insulin , p38 Mitogen-Activated Protein Kinases , Animals , Mice , 3T3-L1 Cells , Cell Differentiation , Insulin/metabolism , Lipids/pharmacology , Mitochondria/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Phosphorylation , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Neuroscience ; 512: 59-69, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36642396

ABSTRACT

Peroxiredoxin-6 (PRDX6), a member of the peroxiredoxin family, has progressively emerged as a possible therapeutic target for a variety of brain diseases, particularly Alzheimer's disease and ischemic stroke. However, the role of PRDX6 in neurons under ischemic conditions has remained elusive. Here, we found that astrocytes could release PRDX6 extracellularly after OGD/R, and that PRDX6 release actually worsened neuroapoptosis under OGD/R. We discovered a unique PRDX6/RAGE/JNK signaling pathway that contributes to the effect of neuroapoptosis. We applied a specific inhibitor of the RAGE signaling pathway in a mouse MCAO model and observed significant alterations in animal behavior. Considered together, our findings show the crucial role of the astrocyte-released PRDX6 in the process of neuroapoptosis caused by OGD/R, and could provide novel insights for investigating the molecular mechanism of protecting brain function from ischemia-reperfusion injury.


Subject(s)
Astrocytes , Brain Ischemia , Peroxiredoxins , Animals , Mice , Apoptosis/physiology , Astrocytes/metabolism , Brain Ischemia/metabolism , Ischemia/metabolism , Peroxiredoxin VI/metabolism , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Reperfusion Injury/metabolism
9.
Plant Physiol Biochem ; 194: 524-532, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36521289

ABSTRACT

The effects of overexpression of the thioredoxin-like protein CDSP32 (Trx CDSP32) on reactive oxygen species (ROS) metabolism in tobacco leaves exposed to cadmium (Cd) were studied by combining physiological measures and proteomics technology. Thus, the number of differentially expressed proteins (DEPs) in plants overexpressing the Trx CDSP32 gene in tobacco (OE) was observed to be evidently lower than that in wild-type (WT) tobacco under Cd exposure, especially the number of down-regulated DEPs. Cd exposure induced disordered ROS metabolism in tobacco leaves. Although Cd exposure inhibited the activities of superoxide dismutase (SOD), catalase (CAT), and l-ascorbate peroxidase (APX) and the expression of proteins related to the thioredoxin-peroxiredoxin (Trx-Prx) pathway, the increase in the activities of peroxidase (POD), monodehydroascorbate reductase (MDHAR), dehydroascorbate reductase (DHAR), glutathione reductase (GR), glutathione peroxidase (GPX), and glutathione S-transferase (GST) and their protein expression levels played an important role in the physiological response to Cd exposure. Notably, Trx CDSP32 was observed to alleviate the decrease in the expression and activities of SOD and CAT caused by Cd exposure and enhance the function of POD. Trx CDSP32 was observed to increase the H2O2 scavenging capacity of the ascorbic acid-glutathione (AsA-GSH) cycle and Trx-Prx pathway under Cd exposure, and it can especially regulate 2-Cys peroxiredoxin (2-Cys Prx) protein expression and thioredoxin peroxidase (TPX) activity. Thus, overexpression of the Trx CDSP32 gene can alleviate the oxidative damage that occurs in tobacco leaves under Cd exposure by modulating antioxidant defense systems.


Subject(s)
Antioxidants , Cadmium , Antioxidants/metabolism , Cadmium/toxicity , Nicotiana/genetics , Nicotiana/metabolism , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , Oxidative Stress , Glutathione/metabolism , Superoxide Dismutase/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Thioredoxins/genetics , Thioredoxins/metabolism , Thioredoxins/pharmacology
10.
Microbiol Spectr ; 10(5): e0166722, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36066254

ABSTRACT

Antibiotic tolerance not only enables bacteria to survive acute antibiotic exposures but also provides bacteria with a window of time in which to develop antibiotic resistance. The increasing prevalence of Campylobacter jejuni isolates resistant to clinically important antibiotics, particularly fluoroquinolones (FQs), is a global public health concern. Currently, little is known about antibiotic tolerance and its effects on resistance development in C. jejuni. Here, we show that exposure to ciprofloxacin or tetracycline at concentrations 10 and 100 times higher than the MIC induces antibiotic tolerance in C. jejuni, whereas gentamicin or erythromycin treatment causes cell death. Interestingly, FQ resistance rapidly develops in C. jejuni after tolerance induction by ciprofloxacin and tetracycline. Furthermore, after tolerance is induced, alkyl hydroperoxide reductase (AhpC) plays a critical role in reducing FQ resistance development by alleviating oxidative stress. Together, these results demonstrate that exposure of C. jejuni to antibiotics can induce antibiotic tolerance and that FQ-resistant (FQR) C. jejuni clones rapidly emerge after tolerance induction. This study elucidates the mechanisms underlying the high prevalence of FQR C. jejuni and provides insights into the effects of antibiotic tolerance on resistance development. IMPORTANCE Antibiotic tolerance compromises the efficacy of antibiotic treatment by extending bacterial survival and facilitating the development of mutations associated with antibiotic resistance. Despite growing public health concerns about antibiotic resistance in C. jejuni, antibiotic tolerance has not yet been investigated in this important zoonotic pathogen. Here, our results show that exposure of C. jejuni to ciprofloxacin or tetracycline leads to antibiotic tolerance development, which subsequently facilitates the emergence of FQR C. jejuni. Importantly, these antibiotics are commonly used in animal agriculture. Moreover, our study suggests that the use of non-FQ drugs in animal agriculture promotes FQ resistance development, which is crucial because antibiotic-resistant C. jejuni is primarily transmitted from animals to humans. Overall, these findings increase our understanding of the mechanisms of resistance development through the induction of antibiotic tolerance.


Subject(s)
Campylobacter jejuni , Drug Resistance, Bacterial , Fluoroquinolones , Anti-Bacterial Agents/pharmacology , Bacteria , Campylobacter jejuni/drug effects , Campylobacter jejuni/genetics , Ciprofloxacin/pharmacology , Drug Resistance, Bacterial/genetics , Erythromycin/pharmacology , Fluoroquinolones/pharmacology , Gentamicins/pharmacology , Microbial Sensitivity Tests , Peroxiredoxins/pharmacology , Tetracycline/pharmacology
11.
Int J Biol Sci ; 18(9): 3747-3761, 2022.
Article in English | MEDLINE | ID: mdl-35813474

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignancies worldwide, yet successful treatment still remains a challenge. In this study, we found that oxiconazole (OXI), a broad-spectrum antifungal agent, exhibits certain anti-tumor effect against CRC. Autophagy arrest and subsequent apoptosis are characterized as pivotal events involving OXI-induced growth suppression of CRC cells. Mechanistically, OXI downregulates the protein levels of peroxiredoxin-2 (PRDX2), an antioxidant enzyme, for reactive oxygen species (ROS) detoxication, to initiate autophagy by inactivating the Akt/mTOR pathway and inhibiting RAB7A-mediated fusion of autophagosome and lysosome, which lead to extreme accumulation of autophagosomes and subsequent growth suppression of CRC cells. Consistently, interfering with autophagy or overexpressing PRDX2 significantly impedes OXI-induced growth suppression of CRC cells. Moreover, OXI plus oxaliplatin, a mainstay drug for CRC treatment, achieves an improved anti-tumor effect. Taken together, our findings bring novel mechanistic insights into OXI-induced autophagy arrest and the growth inhibitory effect on CRC cells, and suggest a promisingly therapeutic role of OXI for CRC treatment.


Subject(s)
Colorectal Neoplasms , Peroxiredoxins , Apoptosis/genetics , Autophagy , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Repositioning , Humans , Imidazoles , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology
12.
Front Endocrinol (Lausanne) ; 12: 718235, 2021.
Article in English | MEDLINE | ID: mdl-34557160

ABSTRACT

Oxidative stress is hypothesized to play a role in pancreatic ß-cell damage, potentially contributing to ß-cell dysfunction and death in both type 1 and type 2 diabetes. Oxidative stress arises when naturally occurring reactive oxygen species (ROS) are produced at levels that overwhelm the antioxidant capacity of the cell. ROS, including superoxide and hydrogen peroxide, are primarily produced by electron leak during mitochondrial oxidative metabolism. Additionally, peroxynitrite, an oxidant generated by the reaction of superoxide and nitric oxide, may also cause ß-cell damage during autoimmune destruction of these cells. ß-cells are thought to be susceptible to oxidative damage based on reports that they express low levels of antioxidant enzymes compared to other tissues. Furthermore, markers of oxidative damage are observed in islets from diabetic rodent models and human patients. However, recent studies have demonstrated high expression of various isoforms of peroxiredoxins, thioredoxin, and thioredoxin reductase in ß-cells and have provided experimental evidence supporting a role for these enzymes in promoting ß-cell function and survival in response to a variety of oxidative stressors. This mini-review will focus on the mechanism by which thioredoxins and peroxiredoxins detoxify ROS and on the protective roles of these enzymes in ß-cells. Additionally, we speculate about the role of this antioxidant system in promoting insulin secretion.


Subject(s)
Antioxidants/pharmacology , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 2/prevention & control , Insulin-Secreting Cells/drug effects , Oxidative Stress , Peroxiredoxins/pharmacology , Thioredoxins/pharmacology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Insulin-Secreting Cells/pathology
13.
Arch Biochem Biophys ; 697: 108671, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33181129

ABSTRACT

PURPOSE: Peroxiredoxin 1 (Prx1) is known to be a multifunctional antioxidant enzyme playing an essential role in protecting the organism against oxidative stress. We hypothesized that administration of exogenous recombinant Prx1 may provide additional protection of the mammalian organism during the development of acute oxidative stress induced by ionizing radiation. Hence, the aim of the present work was to study the radioprotective properties of exogenous Prx1. MATERIALS AND METHODS: Recombinant Prx1 was obtained by genetic engineering. The properties of Prx1 were studied using physicochemical methods. An immunoblotting and ELISA were used for the determination of the level of endogenous and exogenous Prx1 in animal blood. The survival rate of irradiated animals was assessed for 30 days with various modes of administration (intraperitoneal, intramuscular, intravenously) Prx1. Using a hematological analyzer and microscopic analysis, the changes in the level of leukocytes and platelets were assessed in animals that received and did not receive an intravenous injection of Prx1 before irradiation. Genoprotective properties of Prx1 were confirmed by micronucleus test. Real-time PCR was used to investigate the effect of Prx1 on the expression of genes involved in response to oxidative stress. RESULTS: Recombinant Prx1 was shown to significantly reduce oxidative damage to biological macromolecules. Prx1 is an effective radioprotector which decreases the severity of radiation-induced leuko- and thrombocytopenia, plus protects bone marrow cells from damage. The half-life of Prx1 in the bloodstream is more than 1 h, while within 1 h there is a loss of the antioxidant activity of Prx1 by almost 50%, which limits its use long (2 h) before irradiation. The introduction of Prx1 after irradiation has no significant radiomitigating effect. The most effective way of using Prx1 is intravenous administration shortly (15-30 min) before exposure to ionizing radiation, with a dose reduction factor of 1.3. Under the action of ionizing radiation a dose-dependent appearance of endogenous Prx1 in the bloodstream was also observed. The appearance of Prx1 in the bloodstream alters the expression of stress response genes (especial antioxidant response and DNA repair) in the cells of red bone marrow, promoting the activation of repair processes. CONCLUSION: The recombinant Prx1 can be considered as an effective radioprotector for minimizing the risks of injury of animal's body by ionizing radiation.


Subject(s)
Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Peroxiredoxins/pharmacology , Radiation-Protective Agents/pharmacology , Whole-Body Irradiation/adverse effects , Animals , Dose-Response Relationship, Radiation , Hematology , Male , Mice , Survival Analysis
14.
Cell Biol Int ; 44(11): 2231-2242, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32716104

ABSTRACT

This study demonstrates both the antioxidant and anticancer potential of the novel short molecule YT12 derived from peroxiredoxin (Prx) of spirulina, Arthrospira platensis (Ap). ApPrx showed significant reduction in reactive oxygen species (ROS) against hydrogen peroxide (H2 O2 ) stress. The complementary DNA sequence of ApPrx contained 706 nucleotides and its coding region possessed 546 nucleotides between position 115 and 660. Real-time quantitative reverse transcription polymerase chain reaction analysis confirmed the messenger RNA expression of ApPrx due to H2 O2 exposure in spirulina cells at regular intervals, in which the highest expression was noticed on Day 20. Cytotoxicity assay was performed using human peripheral blood mononuclear cells, and revealed that at 10 µM, the YT12 did not exhibit any notable toxicity. Furthermore, ROS scavenging activity of YT12 was performed using DCF-DA assay, in which YT12 scavenged a significant amount of ROS at 25 µM in H2 O2 -treated blood leukocytes. The intracellular ROS in human colon adenocarcinoma cells (HT-29) was regulated by oxidative stress, where the YT12 scavenges ROS in HT-29 cells at 12.5 µM. Findings show that YT12 peptide has anticancer activity, when treated against HT-29 cells. Through the MTT assay, YT12 showed vital cytotoxicity against HT-29 cells. These finding suggested that YT12 is a potent antioxidant molecule which defends ROS against oxidative stress and plays a role in redox balance.


Subject(s)
Peroxiredoxins/metabolism , Spirulina/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antioxidants/metabolism , Antioxidants/pharmacology , Cell Line, Tumor , Humans , Hydrogen Peroxide/metabolism , Leukocytes, Mononuclear/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/physiology , Peptides/metabolism , Peptides/pharmacology , Peroxiredoxins/pharmacology , Reactive Oxygen Species/metabolism , Spirulina/genetics
15.
Biotech Histochem ; 95(8): 626-633, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32362142

ABSTRACT

Local action of nicotine on oral mucosa contributes to the pathogenesis of precancerous and cancerous lesions. Nicotine participation in the mechanism of apoptosis in normal mucosa has not been established. Peroxiredoxin 1 (Prx1) is a cellular antioxidant that participates in regulating apoptosis. We investigated expression of Prx1 and proteins in apoptosis-related downstream signaling by mitogen-activated protein kinases (MAPKs) in nicotine-treated tongue tissues of wild-type and Prx1 knockout (Prx1±) mice; we also investigated these processes in mouse embryonic fibroblast (MEF) cells in vitro. Nicotine increased the expression of Prx1 mRNA in tongue tissues in vivo. The rate of apoptosis was similar among the nicotine-treated mice, nicotine-treated + Prx1± mice and untreated controls. The expression of p-JNK was greater in Prx1± mice compared to control mice. In MEF cells, nicotine increased the expression of Prx1 and inhibited apoptosis and expression of p-p38 and p-JNK. Prx1 knockdown animals exhibited increased apoptotic rate and expression of p-p38 and p-JNK in MEFs. Nicotine-regulated apoptosis might occur via a Prx1-dependent pathway.


Subject(s)
Apoptosis/drug effects , Nicotine/toxicity , Peroxiredoxins/pharmacology , Animals , Gene Expression Regulation/drug effects , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Stroke ; 51(5): 1578-1586, 2020 05.
Article in English | MEDLINE | ID: mdl-32279622

ABSTRACT

Background and Purpose- Our recent study demonstrated that release of Prx2 (peroxiredoxin 2) from red blood cells (RBCs) is involved in the inflammatory response and brain injury after intracerebral hemorrhage. The current study investigated the role of extracellular Prx2 in hydrocephalus development after experimental intraventricular hemorrhage. Methods- There were 4 parts in this study. First, Sprague-Dawley rats received an intraventricular injection of lysed RBC or saline and were euthanized at 1 hour for Prx2 measurements. Second, rats received an intraventricular injection of Prx2, deactivated Prx2, or saline. Third, lysed RBC was coinjected with conoidin A, a Prx2 inhibitor, or vehicle. Fourth, rats received Prx2 injection and were treated with minocycline or saline (i.p.). The effects of Prx2 and the inhibitors were examined using magnetic resonance imaging assessing ventriculomegaly, histology assessing ventricular wall damage, and immunohistochemistry to assess inflammation, particularly at the choroid plexus. Results- Intraventricular injection of lysed RBC resulted in increased brain Prx2 and hydrocephalus. Intraventricular injection of Prx2 alone caused hydrocephalus, ventricular wall damage, activation of choroid plexus epiplexus cells (macrophages), and an accumulation of neutrophils. Conoidin A attenuated lysed RBC-induced injury. Systemic minocycline treatment reduced the epiplexus cell activation and hydrocephalus induced by Prx2. Conclusions- Prx2 contributed to the intraventricular hemorrhage-induced hydrocephalus, probably by inducing inflammatory responses in choroid plexus and ventricular wall damage.


Subject(s)
Cerebral Intraventricular Hemorrhage/metabolism , Choroid Plexus/metabolism , Hydrocephalus/metabolism , Inflammation/metabolism , Macrophages/metabolism , Peroxiredoxins/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cerebral Intraventricular Hemorrhage/complications , Choroid Plexus/drug effects , Choroid Plexus/pathology , Disease Models, Animal , Ependyma/drug effects , Ependyma/pathology , Female , Hydrocephalus/etiology , Hylobatidae , Inflammation/pathology , Injections, Intraventricular , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/pathology , Male , Minocycline/pharmacology , Neutrophils/drug effects , Neutrophils/pathology , Peroxiredoxins/antagonists & inhibitors , Peroxiredoxins/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley
17.
Parasit Vectors ; 12(1): 542, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31727141

ABSTRACT

BACKGROUND: Larvae of Echinococcus granulosus (sensu lato) dwell in host organs for a long time but elicit only a mild inflammatory response, which indicates that the resolution of host inflammation is necessary for parasite survival. The recruitment of alternatively activated macrophages (AAMs) has been observed in a variety of helminth infections, and emerging evidence indicates that AAMs are critical for the resolution of inflammation. However, whether AAMs can be induced by E. granulosus (s.l.) infection or thioredoxin peroxidase (TPx), one of the important molecules secreted by the parasite, remains unclear. METHODS: The activation status of peritoneal macrophages (PMs) derived from mice infected with E. granulosus (sensu stricto) was analyzed by evaluating the expression of phenotypic markers. PMs were then treated in vivo and in vitro with recombinant EgTPx (rEgTPx) and its variant (rvEgTPx) in combination with parasite excretory-secretory (ES) products, and the resulting activation of the PMs was evaluated by flow cytometry and real-time PCR. The phosphorylation levels of various molecules in the PI3K/AKT/mTOR pathway after parasite infection and antigen stimulation were also detected. RESULTS: The expression of AAM-related genes in PMs was preferentially induced after E. granulosus (s.s.) infection, and phenotypic differences in cell morphology were detected between PMs isolated from E. granulosus (s.s.)-infected mice and control mice. The administration of parasite ES products or rEgTPx induced the recruitment of AAMs to the peritoneum and a notable skewing of the ratio of PM subsets, and these effects are consistent with those obtained after E. granulosus (s.s.) infection. ES products or rEgTPx also induced PMs toward an AAM phenotype in vitro. Interestingly, this immunomodulatory property of rEgTPx was dependent on its antioxidant activity. In addition, the PI3K/AKT/mTOR pathway was activated after parasite infection and antigen stimulation, and the activation of this pathway was suppressed by pre-treatment with an AKT/mTOR inhibitor. CONCLUSIONS: This study demonstrates that E. granulosus (s.s.) infection and ES products, including EgTPx, can induce PM recruitment and alternative activation, at least in part, via the PI3K/AKT/mTOR pathway. These results suggest that EgTPx-induced AAMs might play a key role in the resolution of inflammation and thereby favour the establishment of hydatid cysts in the host.


Subject(s)
Echinococcus granulosus/immunology , Macrophages, Peritoneal/immunology , Oncogene Protein v-akt/metabolism , Peroxiredoxins/immunology , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Echinococcosis/parasitology , Echinococcus granulosus/enzymology , Female , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/parasitology , Mice , Mice, Inbred BALB C , Peroxiredoxins/pharmacology , Phenotype , Phosphorylation , Signal Transduction , Specific Pathogen-Free Organisms
18.
Neurotoxicology ; 74: 242-251, 2019 09.
Article in English | MEDLINE | ID: mdl-31408635

ABSTRACT

Diethylhexyl phthalate (DEHP) is used in many plastic products, such as perfumes, lunch boxes, bags, and building materials. As DEHP is not covalently bound to the plastic, humans can be easily exposed to it. DEHP induces neurobehavioral changes and neuronal cell death; however, the exact mechanism behind this is still unclear. We hypothesized that the neurotoxic mechanism is related to DEHP-induced oxidative stress leading to apoptosis through mitochondrial fission. We demonstrated that DEHP-induced oxidative stress triggers neuronal cell death via mitochondrial fission in mouse hippocampal HT-22 cells. Furthermore, we identified that peroxiredoxin 5 (Prx5), an antioxidant enzyme induced by DEHP, prevents DEHP-induced mitochondrial fission by inhibiting the production of reactive oxygen species. We conclude that Prx5 may be a promising therapeutic target for mitigating DEHP-induced neuronal cell death.


Subject(s)
Cell Death/genetics , Diethylhexyl Phthalate/toxicity , Hippocampus/pathology , Mitochondria/genetics , Neurons/drug effects , Peroxiredoxins/pharmacology , Animals , Apoptosis/genetics , Cell Line , Gene Knockdown Techniques , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mitochondria/ultrastructure , Neurons/pathology , Oxidative Stress/drug effects , Peroxiredoxins/genetics , Reactive Oxygen Species
19.
J Neuroinflammation ; 15(1): 87, 2018 Mar 19.
Article in English | MEDLINE | ID: mdl-29554978

ABSTRACT

BACKGROUND: Peroxiredoxin (Prx) protein family have been reported as important damage-associated molecular patterns (DAMPs) in ischemic stroke. Since peroxiredoxin 2 (Prx2) is the third most abundant protein in erythrocytes and the second most protein in the cerebrospinal fluid in traumatic brain injury and subarachnoid hemorrhage (SAH) patients, we assessed the role of extracellular Prx2 in the context of SAH. METHODS: We introduced a co-culture system of primary neurons and microglia. Prx2 was added to culture medium with oxyhemoglobin (OxyHb) to mimic SAH in vitro. Neuronal cell viability was assessed by lactate dehydrogenase (LDH) assay, and neuronal apoptosis was determined by TUNEL staining. Inflammatory factors in culture medium were measured by ELISA, and their mRNA levels in microglia were determined by qPCR. Toll-like receptor 4 knockout (TLR4-KO) mice were used to provide TLR4-KO microglia; ST-2825 was used to inhibit MyD88, and pyrrolidine dithiocarbamate (PDTC) was used to inhibit NF-κB. Related cellular signals were analyzed by Western blot. Furthermore, we detected the level of Prx2 in aneurysmal SAH patients' cerebrospinal fluids (CSF) and compared its relationship with Hunt-Hess grades. RESULTS: Prx2 interacted with TLR4 on microglia after SAH and then activated microglia through TLR4/MyD88/NF-κB signaling pathway. Pro-inflammatory factors were expressed and released, eventually caused neuronal apoptosis. The levels of Prx2 in SAH patients positively correlated with Hunt-Hess grades. CONCLUSIONS: Extracellular Prx2 in CSF after SAH is a DAMP which resulted in microglial activation via TLR4/MyD88/NF-κB pathway and then neuronal apoptosis. Prx2 in patients' CSF may be a potential indicator of brain injury and prognosis.


Subject(s)
Microglia/drug effects , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Subarachnoid Hemorrhage/cerebrospinal fluid , Toll-Like Receptor 4/metabolism , Animals , Animals, Newborn , Antioxidants/pharmacology , Cerebral Cortex/cytology , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , In Situ Nick-End Labeling , L-Lactate Dehydrogenase/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Oxyhemoglobins/pharmacology , Pyrrolidines/pharmacology , RNA, Messenger/metabolism , Spiro Compounds/pharmacology , Thiocarbamates/pharmacology , Toll-Like Receptor 4/genetics
20.
J Nat Prod ; 81(2): 298-306, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29394050

ABSTRACT

Acute myeloid leukemia (AML) is a hematologic malignancy that is characterized by clonal proliferation of myeloid blasts. Despite the progress that has been made in the treatment of various malignant hematopoietic diseases, the effective treatment of AML remains very challenging. Differentiation therapy has emerged as a promising approach for leukemia treatment, and new and effective chemical agents to trigger the differentiation of AML cells, especially drug-resistant cells, are urgently required. Herein, the natural product jungermannenone C, a tetracyclic diterpenoid isolated from liverworts, is reported to induce cell differentiation in AML cells. Interestingly, the unnatural enantiomer of jungermannenone C (1) was found to be more potent than jungermannenone C in inducing cell differentiation. Furthermore, compound 1 targets peroxiredoxins I and II by selectively binding to the conserved cysteine residues and leads to cellular reactive oxygen species accumulation. Accordingly, ent-jungermannenone C (1) shows potential for further investigation as an effective differentiation therapy against AML.


Subject(s)
Cell Differentiation/drug effects , Diterpenes/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Hepatophyta/chemistry , Humans , Leukemia, Myeloid, Acute/metabolism , Peroxiredoxins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...