Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 193(Pt 1): 108-121, 2022 11 20.
Article in English | MEDLINE | ID: mdl-36241072

ABSTRACT

Doxorubicin (DOX), a clinical chemotherapeutic drug, is often annoyed by its cardiotoxicity which involves ferroptosis in its pathological progress. Human umbilical cord mesenchymal stem cells (HucMSCs)-derived exosomes (HucMSCs-Exo) are proven effective in treating cardiovascular diseases. This study aimed to compare the therapeutic effects between normoxic HucMSCs-Exo (Exo) and hypoxic HucMSCs-Exo (Hypo-Exo) on DOX-induced ferroptosis and explore the underlying mechanisms. An acute cardiotoxicity model was successfully constructed by administrating two doses intraperitoneal injections of DOX (25 mg/kg in total). Exo and Hypo-Exo were extracted by ultracentrifugation and characterized. Compared with Exo, Hypo-Exo and Ferrostatin-1 (Fer-1) exerted superior effects on inhibiting DOX-induced ferroptosis, as evidenced by decreasing malondialdehyde (MDA), iron content and increasing glutathione (GSH) level as well as ferroptosis-related genes expression including prostaglandin-endoperoxide synthase 2 (Ptgs2) mRNA level and glutathione peroxidase 4 (GPX4) protein level. Based on quantitative proteomics analysis, we found that thioredoxin1 (Trx1) was remarkably upregulated in Hypo-Exo and exhibited anti-ferroptosis activity via activating the mechanistic target of rapamycin complex 1 (mTORC1) in neonatal rat cardiomyocytes (NRCMs). Trx1 knockdown and rapamycin (an mTORC1 inhibitor) partially abolished the protective effects of Hypo-Exo. Furthermore, our data indicated that solute carrier family 7 member 11 (SLC7A11) was critical for GPX4 protein synthesis. In conclusion, Hypo-Exo exhibited a better suppression of ferroptosis in DOX-induced cardiotoxicity. Trx1-mediated mTORC1 activation is critical for the Hypo-Exo anti-ferroptosis process, which involves increased GPX4 protein synthesis and decreased iron overload. This study indicated that Hypo-Exo may present a potential strategy against ferroptosis in DOX-induced cardiotoxicity.


Subject(s)
Cardiotoxicity , Mesenchymal Stem Cells , Animals , Humans , Rats , Apoptosis/genetics , Doxorubicin/toxicity , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mesenchymal Stem Cells/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Thioredoxins/metabolism , Umbilical Cord/cytology
2.
Eur J Pharmacol ; 919: 174756, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35032486

ABSTRACT

Thioredoxin-1 (Trx-1) is a small redox-active protein normally found in mammalian cells that responds to the changing redox environment by contributing electrons or regulating related proteins. There is growing evidence that Trx-1 has multiple functions, including cytoprotective, anti-apoptotic, antioxidant and anti-inflammatory effects. To date, researchers have found that Trx-1 deficiency leads to severe damage in various disease models, such as atherosclerosis, cerebral ischemia, diabetes and tumors. Conversely, activation of Trx-1 has a protective effect against these diseases. Accordingly, a variety of Trx-1 inducers have been widely used in the clinic with significant therapeutic value. In this paper, we summarize the pathogenesis of Trx-1 involvement in the above-mentioned diseases and describe the protective effects of Trx-1 inducers on them.


Subject(s)
Antioxidants/metabolism , Coronary Artery Disease/metabolism , Thioredoxins/metabolism , Humans
3.
Antioxidants (Basel) ; 10(7)2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34356388

ABSTRACT

Thioredoxin 1 (Trx1) is a major antioxidant that acts adaptively to protect the heart during the development of diabetic cardiomyopathy. The molecular mechanism(s) responsible for regulating the Trx1 level and/or activity during diabetic cardiomyopathy is unknown. ß-hydroxybutyrate (ßHB), a major ketone body in mammals, acts as an alternative energy source in cardiomyocytes under stress, but it also appears to be involved in additional mechanisms that protect the heart against stress. ßHB upregulated Trx1 in primary cultured cardiomyocytes in a dose- and a time-dependent manner and a ketogenic diet upregulated Trx1 in the heart. ßHB protected cardiomyocytes against H2O2-induced death, an effect that was abolished in the presence of Trx1 knockdown. ßHB also alleviated the H2O2-induced inhibition of mTOR and AMPK, known targets of Trx1, in a Trx1-dependent manner, suggesting that ßHB potentiates Trx1 function. It has been shown that ßHB is a natural inhibitor of HDAC1 and knockdown of HDAC1 upregulated Trx1 in cardiomyocytes, suggesting that ßHB may upregulate Trx1 through HDAC inhibition. ßHB induced Trx1 acetylation and inhibited Trx1 degradation, suggesting that ßHB-induced inhibition of HDAC1 may stabilize Trx1 through protein acetylation. These results suggest that ßHB potentiates the antioxidant defense in cardiomyocytes through the inhibition of HDAC1 and the increased acetylation and consequent stabilization of Trx1. Thus, modest upregulation of ketone bodies in diabetic hearts may protect the heart through the upregulation of Trx1.

4.
Eur J Immunol ; 51(1): 115-124, 2021 01.
Article in English | MEDLINE | ID: mdl-32902872

ABSTRACT

Thioredoxin-1 (Trx1) is a vital component for cellular redox homeostasis. In T cells, Trx1 donates electrons for the de novo synthesis of deoxyribonucleotides to allow rapid cell proliferation. The Trx-interacting protein (Txnip) binds to the reduced Trx1 and inhibits its activity. However, the role of Txnip in adaptive immunity in vivo is unknown. Here, we show that absence of Txnip increased proliferation of effector T cells and GC B-cell responses in response to lymphocytic choriomeningitis virus and Qß virus-like particles, respectively, but did not affect development and homeostasis of T and B cells. While downregulation of Txnip and concomitant upregulation of Trx1 is critical for rapid T-cell expansion upon viral infection, re-expression of Txnip and consequently inhibition of Trx1 is important to restrain late T-cell expansion. Importantly, we demonstrated that T-cell receptor (TCR) engagement but not CD28 costimulation is critically required for Txnip downregulation. Thus, this study further uncovers positive and negative control of lymphocyte proliferation by the Trx1 system.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Carrier Proteins/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thioredoxins/antagonists & inhibitors , Thioredoxins/immunology , Animals , B-Lymphocytes/cytology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Proliferation , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , In Vitro Techniques , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidation-Reduction , T-Lymphocytes/cytology , Thioredoxins/genetics , Thioredoxins/metabolism
5.
Arch Biochem Biophys ; 692: 108515, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32791141

ABSTRACT

Apoptosis-inducing factor (AIF) is a flavoprotein and essential partner of the CHCHD4 redox protein during the mitochondrial intermembrane space import machinery. Mammalian AIF has three cysteine residues, which have received little attention. Previous reports have evidenced a redox interaction between AIF and thioredoxin 1 (Trx1), particularly after oxidant conditions. Therefore, we asked whether the cysteine residues of the human AIF could be oxidized. Our data showed that endogenous AIF could be oxidized to disulfide-linked conjugates (DLC). Overexpressed WT AIF in HEK293T cells, as well as recombinant WT AIF, formed DLC. Expression of C256S, C317S or C441S AIF mutants severely inhibited DLC formation in cells exposed to oxidants. In vitro, DLC formation was completely precluded with C256S and C441S AIF mutants and partially inhibited with the C317S mutant. DLC was shown to enhance cellular susceptibility to apoptosis induced by staurosporine, likely by preventing AIF to maintain mitochondrial oxidative phosphorylation. Cells with decreased expression of Trx1 produced more AIF DLC than those with normal Trx1 levels, and in vitro, Trx1 was able to decrease the amount of AIF DLC. Finally, confocal analysis, as well as immunoblotting of mitochondrial fraction, indicated that a fraction of Trx1 is present in mitochondria. Overall, these data provide evidence that all three cysteine residues of AIF can be oxidized to DLC, which can be disrupted by mitochondrial Trx1.


Subject(s)
Apoptosis Inducing Factor , Apoptosis , Disulfides , Amino Acid Substitution , Apoptosis Inducing Factor/chemistry , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Disulfides/chemistry , Disulfides/metabolism , HEK293 Cells , HeLa Cells , Humans , Mutation, Missense , Oxidation-Reduction , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Staurosporine/pharmacology
6.
Cardiovasc Res ; 116(10): 1742-1755, 2020 08 01.
Article in English | MEDLINE | ID: mdl-31584633

ABSTRACT

AIMS: Thioredoxin 1 (Trx1) is an evolutionarily conserved oxidoreductase that cleaves disulphide bonds in oxidized substrate proteins such as mechanistic target of rapamycin (mTOR) and maintains nuclear-encoded mitochondrial gene expression. The cardioprotective effect of Trx1 has been demonstrated via cardiac-specific overexpression of Trx1 and dominant negative Trx1. However, the pathophysiological role of endogenous Trx1 has not been defined with a loss-of-function model. To address this, we have generated cardiac-specific Trx1 knockout (Trx1cKO) mice. METHODS AND RESULTS: Trx1cKO mice were viable but died with a median survival age of 25.5 days. They developed heart failure, evidenced by contractile dysfunction, hypertrophy, and increased fibrosis and apoptotic cell death. Multiple markers consistently indicated increased oxidative stress and RNA-sequencing revealed downregulation of genes involved in energy production in Trx1cKO mice. Mitochondrial morphological abnormality was evident in these mice. Although heterozygous Trx1cKO mice did not show any significant baseline phenotype, pressure-overload-induced cardiac dysfunction, and downregulation of metabolic genes were exacerbated in these mice. mTOR was more oxidized and phosphorylation of mTOR substrates such as S6K and 4EBP1 was impaired in Trx1cKO mice. In cultured cardiomyocytes, Trx1 knockdown inhibited mitochondrial respiration and metabolic gene promoter activity, suggesting that Trx1 maintains mitochondrial function in a cell autonomous manner. Importantly, mTOR-C1483F, an oxidation-resistant mutation, prevented Trx1 knockdown-induced mTOR oxidation and inhibition and attenuated suppression of metabolic gene promoter activity. CONCLUSION: Endogenous Trx1 is essential for maintaining cardiac function and metabolism, partly through mTOR regulation via Cys1483.


Subject(s)
Energy Metabolism , Heart Failure/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , TOR Serine-Threonine Kinases/metabolism , Thioredoxins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Energy Metabolism/genetics , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Oxidative Stress , Rats, Wistar , Signal Transduction , Thioredoxins/genetics
7.
J Zhejiang Univ Sci B ; 19(10): 750-763, 2018.
Article in English | MEDLINE | ID: mdl-30269443

ABSTRACT

Helicobacter pylori infection is related to the development of gastric diseases. Our previous studies showed that high thioredoxin-1 (Trx1) expression in H. pylori can promote gastric carcinogenesis. To explore the underlying molecular mechanisms, we performed an isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomic analysis of stomach tissues from Mongolian gerbil infected with H. pylori expressing high and low Trx1. Differences in the profiles of the expressed proteins were analyzed by bioinformatics and verified using Western blot analysis. We found three candidate proteins, 14-3-3α/ß, glutathione-S-transferase (GST), and heat shock protein 70 (HSP70), in high Trx1 tissues compared with low Trx1 tissues and concluded that cellular stress and redox activity-related proteins were involved in the pathogenesis of gastric cancer associated with H. pylori Trx1.


Subject(s)
Helicobacter Infections/complications , Helicobacter pylori , Stomach Neoplasms/etiology , Stress, Physiological , Thioredoxins/physiology , 14-3-3 Proteins/physiology , Animals , Computational Biology , Gerbillinae , Glutathione Transferase/physiology , HSP70 Heat-Shock Proteins/physiology , Oxidation-Reduction
8.
Article in English | WPRIM (Western Pacific) | ID: wpr-1010416

ABSTRACT

Helicobacter pylori infection is related to the development of gastric diseases. Our previous studies showed that high thioredoxin-1 (Trx1) expression in H. pylori can promote gastric carcinogenesis. To explore the underlying molecular mechanisms, we performed an isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomic analysis of stomach tissues from Mongolian gerbil infected with H. pylori expressing high and low Trx1. Differences in the profiles of the expressed proteins were analyzed by bioinformatics and verified using Western blot analysis. We found three candidate proteins, 14-3-3α/β, glutathione-S-transferase (GST), and heat shock protein 70 (HSP70), in high Trx1 tissues compared with low Trx1 tissues and concluded that cellular stress and redox activity-related proteins were involved in the pathogenesis of gastric cancer associated with H. pylori Trx1.


Subject(s)
Animals , 14-3-3 Proteins/physiology , Computational Biology , Gerbillinae , Glutathione Transferase/physiology , HSP70 Heat-Shock Proteins/physiology , Helicobacter Infections/complications , Helicobacter pylori , Oxidation-Reduction , Stomach Neoplasms/etiology , Stress, Physiological , Thioredoxins/physiology
9.
Respir Physiol Neurobiol ; 201: 38-46, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24973470

ABSTRACT

Antioxidant therapy attenuates pulmonary hypertension (PH). In the present study, we tested the antioxidant effects of fasudil against PH in rats. Monocrotaline (MCT)-induced and chronic hypoxia-induced PH models of rats were established, and the haemodynamic and pathomorphologic results of three different doses of fasudil (10 mg/kg, 30 mg/kg, and 75 mg/kg per day) were subsequently compared with those of bosentan (30 mg/kg per day). Additionally, the protein expressions of thioredoxin-1 (Trx1) and hypoxia inducible factor-1α (HIF-1α), the content of superoxide dismutase (SOD), and the levels of hydrogen peroxide (H2O2), malonyldialdehyde (MDA), and hydroxy radical (·OH) were investigated. Fasudil effectively reduced the right ventricular systolic pressure (RVSP) and alleviated right ventricle (RV) hypertrophy, as well as the histological changes in the pulmonary arterioles. Moreover, fasudil markedly lessened the expression of Trx1 and HIF-1α, up-regulated the concentration of SOD, and lowered the levels of H2O2, MDA, and ·OH. In conclusion, fasudil is a notably attractive potential therapy for PH.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Gene Expression Regulation/drug effects , Hypertension, Pulmonary/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oxidative Stress/drug effects , Thioredoxins/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Analysis of Variance , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Hemodynamics/drug effects , Hydrogen Peroxide/metabolism , Hypertension, Pulmonary/etiology , Hypoxia/chemically induced , Hypoxia/complications , Male , Malondialdehyde/metabolism , Monocrotaline/toxicity , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use
10.
Mol Hum Reprod ; 20(2): 148-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24130091

ABSTRACT

Uterine leiomyomas are benign tumors that develop from smooth muscle cells (SMCs). The reactive oxygen species (ROS) have been shown to be involved in the signaling pathways that stimulate proliferation of a variety of cell types. Thioredoxin-1 (TRX-1) is a redox-regulating protein, which is overexpressed in various tumors. In the present study, we investigated the expressions of TRX-1 and its related molecules in uterine leiomyomas and matched adjacent myometrium. Our results showed the expression of TRX-1 was increased in leiomyomas compared with the matched adjacent myometrium by quantitative RT-PCR and western blotting. FOXO3A expression was increased in leiomyomas compared with myometrium by western blotting. The mRNA levels of hypoxia-inducible factor-1α, cyclooxygenase-2 and cyclin D1 were increased in leiomyomas compared with the adjacent myometrium. The mRNA level of (thioredoxin-1-binding protein) TBP-2 in leiomyomas was not altered when compared with the matched adjacent myometrium. These results suggest that TRX-1 and some of its related molecules are associated with the pathogenesis of uterine leiomyomas. The identification of TRX-1 signaling pathways leading to cell proliferation points to another potential therapeutic target for treatment and/or prevention of uterine leiomyomas.


Subject(s)
Gene Expression Regulation, Neoplastic , Leiomyoma/genetics , Myometrium/metabolism , RNA, Messenger/genetics , Thioredoxins/genetics , Uterine Neoplasms/genetics , Adult , Blotting, Western , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leiomyoma/metabolism , Leiomyoma/pathology , Leiomyoma/surgery , Middle Aged , Myometrium/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidative Stress , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , TATA Box Binding Protein-Like Proteins/genetics , TATA Box Binding Protein-Like Proteins/metabolism , Thioredoxins/metabolism , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Uterine Neoplasms/surgery
11.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-565991

ABSTRACT

Aim To investigate the effects of antioxidant probucol on vascular smooth muscle cells(VSMCs) apoptosis induced by H2O2.Methods H2O2 (1 mmol?L-1) was used to induce VSMCs apoptosis.The VSMCs were treated with probucol(100,10,1 ?mol?L-1) for 6 hours.For the evaluation of apoptosis,Annexin V-FITC staining,Hoechest33258 staining and the TUNEL assay were used.The expressions of ASK-1 and Trx-1 were detected by Western blot analysis.Results H2O2 could promote the apoptosis of VSMCs,increase the expression of ASK-1 and decrease the expression of Trx-1.Probucol could attenuate the apoptosis induced by H2O2 in a dose-dependent,down-regulate ASK-1 expression and increase Trx-1 expression.Conclusion Probucol can antagonize the apoptosis of VSMCs induced by H2O2.The mechanism may be correlated with a decreased expression of ASK-1 and an increased expression of Trx-1.

SELECTION OF CITATIONS
SEARCH DETAIL
...