Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.407
Filter
1.
Biomolecules ; 14(7)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39062479

ABSTRACT

Unlike plants and animals, the phytoflagellate Euglena gracilis lacks catalase and contains a non-selenocysteine glutathione peroxidase-like protein (EgGPXL), two peroxiredoxins (EgPrx1 and EgPrx4), and one ascorbate peroxidase in the cytosol to maintain reactive oxygen species (ROS) homeostasis. In the present study, the full-length cDNA of three cytosolic EgGPXLs was obtained and further characterized biochemically and functionally. These EgGPXLs used thioredoxin instead of glutathione as an electron donor to reduce the levels of H2O2 and t-BOOH. The specific peroxidase activities of these enzymes for H2O2 and t-BOOH were 1.3 to 4.9 and 0.79 to 3.5 µmol/min/mg protein, respectively. Cytosolic EgGPXLs and EgPrx1/EgPrx4 were silenced simultaneously to investigate the synergistic effects of these genes on the physiological function of E. gracilis. The suppression of cytosolic EgGPXL genes was unable to induce any critical phenomena in Euglena under normal (100 µmol photons m-2 s-1) and high-light conditions (350 µmol photons m-2 s-1) at both autotrophic and heterotrophic states. Unexpectedly, the suppression of EgGPXL genes was able to rescue the EgPrx1/EgPrx4-silenced cell line from a critical situation. This study explored the potential resilience of Euglena to ROS, even with restriction of the cytosolic antioxidant system, indicating the involvement of some compensatory mechanisms.


Subject(s)
Cytosol , Euglena gracilis , Glutathione Peroxidase , Thioredoxins , Euglena gracilis/genetics , Euglena gracilis/metabolism , Euglena gracilis/enzymology , Thioredoxins/metabolism , Thioredoxins/genetics , Glutathione Peroxidase/metabolism , Glutathione Peroxidase/genetics , Cytosol/metabolism , Hydrogen Peroxide/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Reactive Oxygen Species/metabolism , Peroxiredoxins/metabolism , Peroxiredoxins/genetics
2.
Curr Opin Chem Biol ; 81: 102496, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38959751

ABSTRACT

H2O2 signals trigger adaptive responses affecting cell division, differentiation, migration, and survival. These signals are transduced by selective oxidation of cysteines on specific target proteins, with redox-sensitive cysteines now identified in many proteins, including both kinases and phosphatases. Assessing the contribution of these oxidation events to cell signalling presents several challenges including understanding how and when the selective oxidation of specific proteins takes place in vivo. In recent years, a combination of biochemical, structural, genetic, and computational approaches in fungi, plants, and animals have revealed different ways in which thiol peroxidases (peroxiredoxins) are bypassed or utilised in relaying these signals. Together, these mechanisms provide a conceptual framework for selectively oxidising proteins that will further advance understanding of how redox modifications contribute to health and disease.


Subject(s)
Hydrogen Peroxide , Oxidation-Reduction , Signal Transduction , Animals , Hydrogen Peroxide/metabolism , Humans , Peroxiredoxins/metabolism
3.
Sci Rep ; 14(1): 15642, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977865

ABSTRACT

Oxidative stress plays an essential role in the progression of Alzheimer's disease (AD), the most common age-related neurodegenerative disorder. Streptozotocin (STZ)-induced abnormal brain insulin signaling and oxidative stress play crucial roles in the progression of Alzheimer's disease (AD)-like pathology. Peroxiredoxins (Prxs) are associated with protection from neuronal death induced by oxidative stress. However, the molecular mechanisms underlying Prxs on STZ-induced progression of AD in the hippocampal neurons are not yet fully understood. Here, we evaluated whether Peroxiredoxin 1 (Prx1) affects STZ-induced AD-like pathology and cellular toxicity. Prx1 expression was increased by STZ treatment in the hippocampus cell line, HT-22 cells. We evaluated whether Prx1 affects STZ-induced HT-22 cells using overexpression. Prx1 successfully protected the forms of STZ-induced AD-like pathology, such as neuronal apoptosis, synaptic loss, and tau phosphorylation. Moreover, Prx1 suppressed the STZ-induced increase of mitochondrial dysfunction and fragmentation by down-regulating Drp1 phosphorylation and mitochondrial location. Prx1 plays a role in an upstream signal pathway of Drp1 phosphorylation, cyclin-dependent kinase 5 (Cdk5) by inhibiting the STZ-induced conversion of p35 to p25. We found that STZ-induced of intracellular Ca2+ accumulation was an important modulator of AD-like pathology progression by regulating Ca2+-mediated Calpain activation, and Prx1 down-regulated STZ-induced intracellular Ca2+ accumulation and Ca2+-mediated Calpain activation. Finally, we identified that Prx1 antioxidant capacity affected Ca2+/Calpain/Cdk5-mediated AD-like pathology progress. Therefore, these findings demonstrated that Prx1 is a key factor in STZ-induced hippocampal neuronal death through inhibition of Ca2+/Calpain/Cdk5-mediated mitochondrial dysfunction by protecting against oxidative stress.


Subject(s)
Alzheimer Disease , Calcium , Calpain , Cyclin-Dependent Kinase 5 , Hippocampus , Mitochondria , Neurons , Peroxiredoxins , Streptozocin , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/etiology , Cyclin-Dependent Kinase 5/metabolism , Cyclin-Dependent Kinase 5/genetics , Streptozocin/toxicity , Hippocampus/metabolism , Hippocampus/pathology , Neurons/metabolism , Neurons/pathology , Calpain/metabolism , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Mitochondria/metabolism , Mice , Calcium/metabolism , Cell Line , Oxidative Stress , Apoptosis , Dynamins/metabolism , Dynamins/genetics , Phosphorylation , tau Proteins/metabolism , Signal Transduction
4.
Biomed Pharmacother ; 177: 116953, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38955087

ABSTRACT

The second most common mutation in melanoma occurs in NRAS oncogene, being a more aggressive disease that has no effective approved treatment. Besides, cellular plasticity limits better outcomes of the advanced and therapy-resistant patients. Peroxiredoxins (PRDXs) control cellular processes through direct hydrogen peroxide oxidation or by redox-relaying processes. Here, we demonstrated that PRDX2 could act as a modulator of multiple EMT markers in NRAS-mutated melanomas. PRDX2 knockdown lead to phenotypic changes towards invasion in human reconstructed skin and the treatment with a PRDX mimetic (gliotoxin), decreased migration in PRDX2-deficient cells. We also confirmed the favorable clinical outcome of patients expressing PRDX2 in a large primary melanoma cohort. This study contributes to our knowledge about genes involved in phenotype switching and opens a new perspective for PRDX2 as a biomarker and target in NRAS-mutated melanomas.


Subject(s)
Epithelial-Mesenchymal Transition , GTP Phosphohydrolases , Melanoma , Membrane Proteins , Mutation , Neoplasm Invasiveness , Peroxiredoxins , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/drug therapy , Melanoma/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Cell Line, Tumor , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/drug effects , Cell Movement/drug effects , Cell Movement/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/drug therapy , Female , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic
5.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891864

ABSTRACT

According to the World Health Organization (WHO), breast cancer (BC) is the deadliest and the most common type of cancer worldwide in women. Several factors associated with BC exert their effects by modulating the state of stress. They can induce genetic mutations or alterations in cell growth, encouraging neoplastic development and the production of reactive oxygen species (ROS). ROS are able to activate many signal transduction pathways, producing an inflammatory environment that leads to the suppression of programmed cell death and the promotion of tumor proliferation, angiogenesis, and metastasis; these effects promote the development and progression of malignant neoplasms. However, cells have both non-enzymatic and enzymatic antioxidant systems that protect them by neutralizing the harmful effects of ROS. In this sense, antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), thioredoxin reductase (TrxR), and peroxiredoxin (Prx) protect the body from diseases caused by oxidative damage. In this review, we will discuss mechanisms through which some enzymatic antioxidants inhibit or promote carcinogenesis, as well as the new therapeutic proposals developed to complement traditional treatments.


Subject(s)
Antioxidants , Breast Neoplasms , Reactive Oxygen Species , Humans , Antioxidants/metabolism , Antioxidants/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Reactive Oxygen Species/metabolism , Oxidative Stress , Peroxiredoxins/metabolism , Animals , Glutathione Peroxidase/metabolism , Catalase/metabolism , Superoxide Dismutase/metabolism
6.
Exp Gerontol ; 194: 112485, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38876448

ABSTRACT

The natural polyphenol resveratrol (RSV) might counteract the skeletal muscle age-related loss of muscle mass and strength/function partly acting on mitochondria. This work analysed the effects of a six-week administration of RSV (50 mg/kg/day) in the oxidative Soleus (Sol) skeletal muscle of old rats (27 months old). RSV effects on key mitochondrial biogenesis proteins led to un unchanged amount of SIRT1 protein and a marked decrease (60 %) in PGC-1α protein. In addition, Peroxyredoxin 3 (PRXIII) protein decreased by 50 %, which on overall suggested the absence of induction of mitochondrial biogenesis by RSV in old Sol. A novel direct correlation between PGC-1α and PRXIII proteins was demonstrated by correlation analysis in RSV and ad-libitum (AL) rats, supporting the reciprocally coordinated expression of the proteins. RSV supplementation led to an unexpected 50 % increase in the frequency of the oxidized base OH8dG in mtDNA. Furthermore, RSV supplementation induced a 50 % increase in the DRP1 protein of mitochondrial dynamics. In both rat groups an inverse correlation between PGC-1α and the frequency of OH8dG as well as an inverse correlation between PRXIII and the frequency of OH8dG were also found, suggestive of a relationship between oxidative damage to mtDNA and mitochondrial biogenesis activity. Such results may indicate that the antioxidant activity of RSV in aged Sol impinged on the oxidative fiber-specific, ROS-mediated, retrograde communication, thereby affecting the expression of SIRT1, PGC-1α and PRXIII, reducing the compensatory responses to the age-related mitochondrial oxidative stress and decline.


Subject(s)
Aging , Mitochondria, Muscle , Muscle, Skeletal , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats, Wistar , Resveratrol , Sirtuin 1 , Animals , Resveratrol/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Male , Aging/drug effects , Aging/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/metabolism , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Rats , Stilbenes/pharmacology , Antioxidants/pharmacology , Peroxiredoxins/metabolism , DNA, Mitochondrial/metabolism , Oxidative Stress/drug effects , Dynamins/metabolism , Mitochondrial Dynamics/drug effects
7.
Phytomedicine ; 131: 155752, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38833947

ABSTRACT

BACKGROUND: Cutaneous squamous cell carcinoma (cSCC) is one of the most common skin cancers for which effective drugs are urgently needed. Echinatin, a natural compound extracted from Glycyrrhiza plants, has shown promising antitumour effects. However, the efficacy and the direct target of echinatin in cSCC remain unclear. PURPOSE: This study conducted a systematic investigation of the antitumour effects of echinatin on cSCC and the underlying mechanisms involved. STUDY DESIGN AND METHODS: Three cSCC cell lines, a xenograft model, and a UV-induced cSCC mouse model were used to investigate the potential protective effects of echinatin. The interactions between echinatin and glutathione S-transferase mu3 (GSTM3) and between echinatin and peroxiredoxin-2 (PRDX2) were evaluated by a proteome microarray assay, pull-down LC‒MS/MS analysis, surface plasmon resonance, and molecular docking. The potential mechanisms of GSTM3-mediated echinatin activity were analysed by using western blotting, lentivirus infection and small interfering RNA (siRNA) transfection. RESULTS: In this study, we found that echinatin inhibited the proliferation and migration of cSCC cells but had no cytotoxic effect on primary human keratinocytes. Furthermore, echinatin significantly inhibited tumour growth in vivo. Mechanistically, our data showed that echinatin could directly bind to GSTM3 and PRDX2. Notably, echinatin inhibited GSTM3 and PRDX2 levels by promoting their proteasomal degradation, which led to the disruption of ROS production. We then revealed that echinatin increased mitochondrial ROS production by inhibiting GSTM3. Moreover, echinatin triggered ferroptosis by inhibiting GSTM3-mediated ferroptosis negative regulation (FNR) proteins. In addition, echinatin regulated GSTM3-mediated ROS/MAPK signalling. CONCLUSION: Echinatin has good antitumour effects both in vitro and in vivo. Moreover, our findings indicate that GSTM3 and PRDX2 could function as viable targets of echinatin in cSCC. Consequently, echinatin represents a novel treatment for cSCC through the targeting of GSTM3-mediated ferroptosis.


Subject(s)
Carcinoma, Squamous Cell , Ferroptosis , Glutathione Transferase , Skin Neoplasms , Ferroptosis/drug effects , Animals , Skin Neoplasms/drug therapy , Humans , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Mice , Glutathione Transferase/metabolism , Peroxiredoxins/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Mice, Inbred BALB C , Cell Proliferation/drug effects , Molecular Docking Simulation , Mice, Nude , Cell Movement/drug effects , Xenograft Model Antitumor Assays , Keratinocytes/drug effects , Chalcones
8.
BMC Ophthalmol ; 24(1): 237, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844903

ABSTRACT

BACKGROUND: The purpose of this study was to investigate the photoprotection effect of peroxiredoxin 1 (PRDX1) protein in ultraviolet B (UVB) irradiation-induced damage of retinal pigment epithelium (RPE) and its possible molecular mechanism. METHODS: ARPE-19 cell viability and apoptosis were assessed by MTT assay and flow cytometry, respectively. Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to detect the PRDX1 expression. The corresponding kits were employed to measure the levels or activities of lactate dehydrogenase (LDH), 8-hydroxy-2-deoxyguanosine (8-OHdG), reactive oxygen species (ROS), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD). Western blotting was applied to examine PRDX1 expression and mitogen-activated protein kinase (MAPK) signaling pathway-related proteins. RESULTS: After exposure to 20 mJ/cm2 intensity of UVB irradiation for 24 h, ARPE-19 cells viability was decreased, the leakage degree of LDH and 8-OHdG were increased, and cell apoptosis was elevated. The expression of PRDX1 was significantly down-regulated in UVB-induced ARPE-19 cells. The low expression of PRDX1 was involved in high irradiation intensity. Overexpression of PRDX1 increased cell activity, decreased cell apoptosis, and LDH as well as 8-OHdG leakage in UVB-induced ARPE-19 cells. In addition to alleviating UVB-induced cell damage, PRDX1 overexpression also inhibited UVB-induced oxidative stress (down-regulation of ROS and MDA levels, up-regulation of GSH-Px and SOD activities) and the activation of MAPK signaling pathway in ARPE-19 cells. CONCLUSION: PRDX1 exerts a photoprotection effect on RPE by attenuating UVB-induced cell damage and inhibiting oxidative stress, which can be attributed to the inhibition of MAPK signaling pathway activation.


Subject(s)
Apoptosis , Cell Survival , Oxidative Stress , Peroxiredoxins , Reactive Oxygen Species , Retinal Pigment Epithelium , Ultraviolet Rays , Humans , Retinal Pigment Epithelium/radiation effects , Retinal Pigment Epithelium/metabolism , Peroxiredoxins/metabolism , Ultraviolet Rays/adverse effects , Reactive Oxygen Species/metabolism , MAP Kinase Signaling System/physiology , Cell Line , Blotting, Western , Cells, Cultured , 8-Hydroxy-2'-Deoxyguanosine/metabolism , Signal Transduction
9.
Pathol Res Pract ; 260: 155411, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936092

ABSTRACT

BACKGROUND: Oral leukoplakia (OLK) is the most common oral potentially malignant disorder (OPMD), which can be malignantly transformed into oral squamous cell carcinoma (OSCC). Peroxiredoxin1(Prx1) has been predicted to bind to Prohibitin2 (PHB2), which confers to affect OLK progression; however, the mechanism of Prx1/PHB2 mediated mitophagy involved in OLK remains unclear. METHODS: This study aimed to explore the mechanism of the Prx1/PHB2 axis on senescence in OLK through mediating mitophagy. The positive rate of Ki67 and the expression of p21, p16, PHB2, and LC3 in human normal, OLK, and OSCC tissues were detected by immunohistochemical staining. The mitophagy and mitochondrial function changes were then analyzed in Prx1 knockdown and Prx1C52S mutations in dysplastic oral keratinocyte (DOK) cells treated with H2O2. In situ Proximity Ligation Assay combined with co-immunoprecipitation was used to detect the interaction between Prx1 and PHB2. RESULTS: Clinically, the positive rate of Ki67 progressively increased from normal to OLK, OLK with dysplasia, and OSCC. Higher p21, p16, PHB2, and LC3 expression levels were observed in OLK with dysplasia than in normal and OSCC tissues. In vitro, PHB2 and LC3II expression gradually increased with the degree of DOK cell senescence. Prx1/PHB2 regulated mitophagy and affected senescence in H2O2-induced DOK cells. Furthermore, Prx1C52S mutation specifically reduced interaction between Prx1 and PHB2. Prx1Cys52 is associated with mitochondrial reactive oxygen species (ROS) accumulated and cell cycle arrest. CONCLUSION: Prx1Cys52 functions as a redox sensor that binds to PHB2 and regulates mitophagy in the senescence of OLK, suggesting its potential as a clinical target.


Subject(s)
Cellular Senescence , Leukoplakia, Oral , Mitophagy , Prohibitins , Repressor Proteins , Humans , Mitophagy/physiology , Cellular Senescence/physiology , Leukoplakia, Oral/pathology , Leukoplakia, Oral/metabolism , Leukoplakia, Oral/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/genetics
10.
Aging (Albany NY) ; 16(9): 8198-8216, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38738994

ABSTRACT

Disulfidptosis, a newly recognized cell death triggered by disulfide stress, has garnered attention for its potential role in osteoporosis (OP) pathogenesis. Although sulfide-related proteins are reported to regulate the balance of bone metabolism in OP, the precise involvement of disulfidptosis regulators remains elusive. Herein, leveraging the GSE56815 dataset, we conducted an analysis to delineate disulfidptosis-associated diagnostic clusters and immune landscapes in OP. Subsequently, vertebral bone tissues obtained from OP patients and controls were subjected to RNA sequencing (RNA-seq) for the validation of key disulfidptosis gene expression. Our analysis unveiled seven significant disulfidptosis regulators, including FLNA, ACTB, PRDX1, SLC7A11, NUBPL, OXSM, and RAC1, distinguishing OP samples from controls. Furthermore, employing a random forest model, we identified four diagnostic disulfidptosis regulators including FLNA, SLC7A11, NUBPL, and RAC1 potentially predictive of OP risk. A nomogram model integrating these four regulators was constructed and validated using the GSE35956 dataset, demonstrating promising utility in clinical decision-making, as affirmed by decision curve analysis. Subsequent consensus clustering analysis stratified OP samples into two different disulfidptosis subgroups (clusters A and B) using significant disulfidptosis regulators, with cluster B exhibiting higher disulfidptosis scores and implicating monocyte immunity, closely linked to osteoclastogenesis. Notably, RNA-seq analysis corroborated the expression patterns of two disulfidptosis modulators, PRDX1 and OXSM, consistent with bioinformatics predictions. Collectively, our study sheds light on disulfidptosis patterns, offering potential markers and immunotherapeutic avenues for future OP management.


Subject(s)
Osteoporosis , Sequence Analysis, RNA , rac1 GTP-Binding Protein , Humans , Osteoporosis/genetics , Osteoporosis/immunology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , Filamins/genetics , Female , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Nomograms , Male , Peroxiredoxins
11.
J Oral Pathol Med ; 53(6): 404-413, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38797866

ABSTRACT

BACKGROUND: Some studies confirmed that erythroblast transformation-specific-related gene (ERG) may be a pathogenic factor of oral squamous cell carcinoma (OSCC). However, the undergoing molecular mechanism has not been elucidated yet. OBJECTIVE: In this study, the investigation will focus on how the transcription factor ERG modulates the biological behaviors of OSCC. METHODS: In this study, cancer tissue specimens and corresponding paracancer tissues were collected from 54 patients. Real-time polymerase chain reaction analysis and Western blots were employed to detect the expression of multiple genes. Cell proliferation assays, Transwell, and flow cytometry assay were utilized to detect the proliferation, invasion, and apoptosis of OSCC cell, respectively. Dual luciferase reporter gene and chromatin immunoprecipitation assays were conducted to verify the regulation of ERG on PRDX1. RESULTS: ERG exhibits high expression levels in OSCC. Inhibition of ERG has been shown to effectively suppress the malignant growth of OSCC cells. Moreover, ERG has been found to transcriptionally upregulate the expression of PRDX1. The knockdown of PRDX1 has demonstrated its ability to inhibit the malignant growth of OSCC cells. Interestingly, when PRDX1 is overexpressed, it attenuates the inhibitory effect of si-ERG on the malignant growth of OSCC cells. This suggests that PRDX1 may play a crucial role in mediating the impact of ERG on malignancy in OSCC cells. CONCLUSION: The transcription factor ERG promotes the expression of PRDX1, which could enhance the proliferation and invasion while inhibiting the apoptosis of OSCC cells.


Subject(s)
Carcinoma, Squamous Cell , Cell Proliferation , Mouth Neoplasms , Peroxiredoxins , Transcriptional Regulator ERG , Up-Regulation , Humans , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis , Neoplasm Invasiveness , Transcriptional Activation , Female , Male
12.
EMBO J ; 43(13): 2789-2812, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38811853

ABSTRACT

It has remained unknown how cells reduce cystine taken up from the extracellular space, which is a required step for further utilization of cysteine in key processes such as protein or glutathione synthesis. Here, we show that the thioredoxin-related protein of 14 kDa (TRP14, encoded by TXNDC17) is the rate-limiting enzyme for intracellular cystine reduction. When TRP14 is genetically knocked out, cysteine synthesis through the transsulfuration pathway becomes the major source of cysteine in human cells, and knockout of both pathways becomes lethal in C. elegans subjected to proteotoxic stress. TRP14 can also reduce cysteinyl moieties on proteins, rescuing their activities as here shown with cysteinylated peroxiredoxin 2. Txndc17 knockout mice were, surprisingly, protected in an acute pancreatitis model, concomitant with activation of Nrf2-driven antioxidant pathways and upregulation of transsulfuration. We conclude that TRP14 is the evolutionarily conserved enzyme principally responsible for intracellular cystine reduction in C. elegans, mice, and humans.


Subject(s)
Caenorhabditis elegans , Cysteine , Cystine , Mice, Knockout , Oxidation-Reduction , Proteome , Thioredoxins , Animals , Humans , Mice , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Cysteine/metabolism , Cystine/metabolism , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Proteome/metabolism , Thioredoxins/metabolism , Thioredoxins/genetics
13.
J Pharm Biomed Anal ; 247: 116247, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38815521

ABSTRACT

Amino acid epimerization, a process of converting L-amino acids to D-amino acids, will lead to modification in the protein structure and, subsequently, its biological function. This modification causes no change in protein m/z and may be overlooked during protein analysis. Aspartic Acid Epimerization (AAE) is faster than other amino acids and could be accelerated by free radicals and peroxides. In this work, a novel and site-specific HPLC method using a chiral stationary phase for determining the AAE in the active site model peptide (AP) of Peroxiredoxin 2 has been developed and validated. The developed method showed good linearity (1 - 200 µg/mL) and recoveries of the limit of quantification (LOQ), low, medium, and high concentrations were between 85% and 115%. The Kinetics of AAE in AP were studied using the developed method, and the results showed that when ascorbic acid and Cu2+ coexisted, the AP epimerized rapidly. The AAE extent increased with time and was positively correlated with hydrogen peroxide generation.


Subject(s)
Aspartic Acid , Catalytic Domain , Peroxiredoxins , Chromatography, High Pressure Liquid/methods , Kinetics , Peroxiredoxins/chemistry , Peroxiredoxins/analysis , Aspartic Acid/chemistry , Aspartic Acid/analysis , Peptides/chemistry , Peptides/analysis , Stereoisomerism , Hydrogen Peroxide/chemistry , Ascorbic Acid/chemistry , Ascorbic Acid/analysis , Limit of Detection , Copper/chemistry
14.
Redox Biol ; 73: 103201, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795545

ABSTRACT

Reactive oxygen species (ROS), play important roles in cellular signaling, nonetheless are toxic at higher concentrations. Cells have many interconnected, overlapped or backup systems to neutralize ROS, but their regulatory mechanisms remain poorly understood. Here, we reveal an essential role for mitochondrial AMPylase Fmp40 from budding yeast in regulating the redox states of the mitochondrial 1-Cys peroxiredoxin Prx1, which is the only protein shown to neutralize H2O2 with the oxidation of the mitochondrial glutathione and the thioredoxin Trx3, directly involved in the reduction of Prx1. Deletion of FMP40 impacts a cellular response to H2O2 treatment that leads to programmed cell death (PCD) induction and an adaptive response involving up or down regulation of genes encoding, among others the catalase Cta1, PCD inducing factor Aif1, and mitochondrial redoxins Trx3 and Grx2. This ultimately perturbs the reduced glutathione and NADPH cellular pools. We further demonstrated that Fmp40 AMPylates Prx1, Trx3, and Grx2 in vitro and interacts with Trx3 in vivo. AMPylation of the threonine residue 66 in Trx3 is essential for this protein's proper endogenous level and its precursor forms' maturation under oxidative stress conditions. Additionally, we showed the Grx2 involvement in the reduction of Trx3 in vivo. Taken together, Fmp40, through control of the reduction of mitochondrial redoxins, regulates the hydrogen peroxide, GSH and NADPH signaling influencing the yeast cell survival.


Subject(s)
Hydrogen Peroxide , Oxidation-Reduction , Oxidative Stress , Peroxiredoxins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Thioredoxins , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Thioredoxins/metabolism , Thioredoxins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Glutathione/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Cell Survival , Apoptosis , Peroxidases , Glutaredoxins
15.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1509-1522, 2024 May 25.
Article in Chinese | MEDLINE | ID: mdl-38783812

ABSTRACT

In order to investigate the role of Prdx1 in macrophage polarization, mouse leukemia cells of monocyte macrophage (RAW264.7) were treated with lipopolysaccharides (LPS)+ interferon gamma (IFNγ) or IL-4 to induce type 1 macrophage (M1) and type 1 macrophage (M2) macrophages, respectively. The Prdx1 gene knockout cells (Prdx1-/-) were used for the study. Flow cytometry was conducted to detect M1/M2 macrophage markers, and ELISA kits were used to measure M1/M2 cytokine levels. Inducible nitric-oxide synthase (iNOS) activity, arginase-1 (Arg-1) activity, and oxidative damage were also assessed. The Seahorse XFe24 Extracellular Flux Analyzer was employed to measure extracellular acidification rate and oxygen consumption rate. The mitochondrial membrane potential was analyzed using the mitochondrial membrane potential dye (JC-1) fluorescent probe, and mitochondrial superoxide was detected through fluorescence staining. Additionally, the impact of adding a mitochondrial reactive oxygen species (ROS) scavenger on RAW264.7 macrophage polarization was examined. The results demonstrated an increase in ROS, hydrogen peroxide, and 8-hydroxy-2 deoxyguanosine (8-OHDG). Cytotoxicity and mitochondrial toxic effects, including mitochondrial superoxide accumulation, decreased adenosine-triphosphate (ATP) production, reduced mitochondrial membrane potential, and decreased mitochondrial DNA copy number, were observed. Furthermore, down-regulation of translocase of inner mitochondrial membrane 23 (TIM23) mitochondrial protein and mitochondrial stress protein heat shock protein 60 (HSP60) was noted. The extra cellular acidification rate (ECAR) in M1 macrophage polarization in RAW264.7 cells was increased, while oxygen consumption rate (OCR) in M2 macrophages was reduced. These findings indicate that Prdx1 knockout in RAW264.7 cells can inhibit M2 macrophage polarization but promote M1 macrophage polarization by impairing mitochondrial function and reducing oxidative phosphorylation.


Subject(s)
Homeostasis , Macrophages , Mitochondria , Peroxiredoxins , Animals , Mice , Macrophages/metabolism , Macrophages/drug effects , Mitochondria/metabolism , RAW 264.7 Cells , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Reactive Oxygen Species/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation , Membrane Potential, Mitochondrial , Gene Knockout Techniques
16.
Mol Biol Rep ; 51(1): 607, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38704801

ABSTRACT

BACKGROUND: Intracerebral hemorrhage (ICH) is a critical neurological condition with few treatment options, where secondary immune responses and specific cell death forms, like pyroptosis, worsen brain damage. Pyroptosis involves gasdermin-mediated membrane pores, increasing inflammation and neural harm, with the NLRP3/Caspase-1/GSDMD pathway being central to this process. Peroxiredoxin II (Prx II), recognized for its mitochondrial protection and reactive oxygen species (ROS) scavenging abilities, appears as a promising neuronal pyroptosis modulator. However, its exact role and action mechanisms need clearer definition. This research aims to explore Prx II impact on neuronal pyroptosis and elucidate its mechanisms, especially regarding endoplasmic reticulum (ER) stress and oxidative stress-induced neuronal damage modulation. METHODS AND RESULTS: Utilizing MTT assays, Microscopy, Hoechst/PI staining, Western blotting, and immunofluorescence, we found Prx II effectively reduces LPS/ATP-induced pyroptosis and neuroinflammation in HT22 hippocampal neuronal cells. Our results indicate Prx II's neuroprotective actions are mediated through PI3K/AKT activation and ER stress pathway inhibition, diminishing mitochondrial dysfunction and decreasing neuronal pyroptosis through the ROS/MAPK/NF-κB pathway. These findings highlight Prx II potential therapeutic value in improving intracerebral hemorrhage outcomes by lessening secondary brain injury via critical signaling pathway modulation involved in neuronal pyroptosis. CONCLUSIONS: Our study not only underlines Prx II importance in neuroprotection but also opens new therapeutic intervention avenues in intracerebral hemorrhage, stressing the complex interplay between redox regulation, ER stress, and mitochondrial dynamics in neuroinflammation and cell death management.


Subject(s)
Endoplasmic Reticulum Stress , Oxidative Stress , Peroxiredoxins , Pyroptosis , Animals , Mice , Cell Line , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/complications , Endoplasmic Reticulum Stress/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Mitochondria/metabolism , Mitochondria/drug effects , Neurons/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Peroxiredoxins/metabolism , Pyroptosis/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
17.
J Mol Histol ; 55(4): 403-413, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38758520

ABSTRACT

Hypoxia is a key trigger in the transformation of oral leukoplakia into oral cancer. However, it is still too early to determine the role of hypoxia in the development of oral leukoplakia. Prx1, an antioxidant protein, upregulated by hypoxia, regulates cellular autophagy in leukoplakia. This study aimed to understand the mechanisms by which hypoxia induces Prx1 expression during autophagy in oral leukoplakia. We used an experimental model of tongue epithelial hyperplasia induced by 4-nitroquinoline-1-oxide (4NQO) and dysplastic oral keratinocytes. Prx1 knockdown DOK cells, Leuk-1 cells and control cells were harvested, and cell proliferation was assayed using the Cell Counting Kit-8. Several hypoxia and autophagy-related proteins were examined using quantitative real-time polymerase chain reaction, immunohistochemistry, immunofluorescence, and western blotting in cells and mouse tongue tissues. In addition, the ultrastructure of the cells was observed by transmission electron microscopy. Hypoxia induces cell proliferation, autophagic vesicles and the expression of Prx1, BNIP3, LC3II/I and Beclin-1 in DOK and Leuk-1 cells. However, these effects were all attenuated by Prx1 knockdown. Histologically, 4NQO induced epithelial hyperplasia in the tongue mucosa. The expression of proliferation marker PCNA, autophagy-related proteins LC3B and Beclin-1, as well as HIF-1α/BNIP3 was significantly lower in the tongue tissues of Prx1flox/flox:Cre+ mice compared with Prx1flox/flox mice. In Prx1flox/flox:Cre+ mice, an increased expression of HIF-1α/BNIP3, LC3B and Beclin-1 was detected in epithelial hyperplasia tongue tissues compared to normal tissues. The current study suggests that Prx1 may promotes cell proliferation and autophagy in oral leukoplakia cells via the HIF-1α/BNIP3 pathway.


Subject(s)
Autophagy , Cell Proliferation , Hypoxia-Inducible Factor 1, alpha Subunit , Leukoplakia, Oral , Peroxiredoxins , Signal Transduction , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leukoplakia, Oral/pathology , Leukoplakia, Oral/metabolism , Leukoplakia, Oral/genetics , Mice , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , Tongue/pathology , Tongue/metabolism , Cell Hypoxia , Cell Line , Mitochondrial Proteins
18.
Int J Cancer ; 155(7): 1290-1302, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38738971

ABSTRACT

Peroxiredoxin 1 (PRDX1) is an important member of the peroxiredoxin family (PRDX) and is upregulated in a variety of tumors. Previous studies have found that high PRDX1 expression is closely related to the metastasis of oral squamous cell carcinoma (OSCC), but the specific molecular mechanism is elusive. To elucidate the role of PRDX1 in the metastasis process of OSCC, we evaluated the expression of PRDX1 in OSCC clinical specimens and its impact on the prognosis of OSCC patients. Then, the effect of PRDX1 on OSCC metastasis and cytoskeletal reconstruction was explored in vitro and in nude mouse tongue cancer models, and the molecular mechanisms were also investigated. PRDX1 can directly interact with the actin-binding protein Cofilin, inhibiting the phosphorylation of its Ser3 site, accelerating the depolymerization and turnover of actin, promoting OSCC cell movement, and aggravating the invasion and metastasis of OSCC. In clinical samples and mouse tongue cancer models, PRDX1 also increased lymph node metastasis of OSCC and was negatively correlated with the phosphorylation of Cofilin; PRDX1 also reduced the overall survival rate of OSCC patients. In summary, our study identified that PRDX1 may be a potential therapeutic target to inhibit OSCC metastasis.


Subject(s)
Carcinoma, Squamous Cell , Mice, Nude , Mouth Neoplasms , Peroxiredoxins , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Humans , Animals , Mice , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/secondary , Cell Line, Tumor , Male , Female , Prognosis , Phosphorylation , Cell Movement , Lymphatic Metastasis , Middle Aged , Actin Depolymerizing Factors/metabolism , Tongue Neoplasms/pathology , Tongue Neoplasms/metabolism , Tongue Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Cofilin 1/metabolism , Mice, Inbred BALB C
19.
Methods Mol Biol ; 2798: 235-263, 2024.
Article in English | MEDLINE | ID: mdl-38587748

ABSTRACT

A set of peroxidases detoxifies H2O2 and mediates H2O2-dependent signal propagation. The peroxidases include peroxiredoxins, glutathione peroxidases, ascorbate peroxidases, and catalases. This at least partial redundancy impedes addressing individual proteins in living plant cells so that the protein functions are often studied by biochemical assays in vitro. In vivo analysis frequently relies on transgenic insertion lines resulting in the knockdown or knockout of the protein of interest. However, many proteins have multiple isoforms in close genomic arrangement so that even crossing of transgenic lines does not allow for a knockdown of all isoforms. The genes encoding for the three cytosolic peroxiredoxins PRXIIB, C, and D in Arabidopsis thaliana are located in close vicinity on chromosome 1 so that crossing over between the genes most rarely occurs and successful crossing of the plants appears impossible. Genome editing instead allows targeting of multiple isoforms and knocks out several genes at once. This chapter describes how to inactivate the three cytosolic peroxiredoxins by CRISPR/Cas9 in A. thaliana.


Subject(s)
Arabidopsis , Peroxiredoxins , Peroxiredoxins/genetics , Gene Editing , Hydrogen Peroxide , Arabidopsis/genetics , Protein Isoforms
20.
Cell Commun Signal ; 22(1): 231, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637880

ABSTRACT

BACKGROUND: Neurodegenerative diseases are increasingly recognized for their association with oxidative stress, which leads to progressive dysfunction and loss of neurons, manifesting in cognitive and motor impairments. This study aimed to elucidate the neuroprotective role of peroxiredoxin II (Prx II) in counteracting oxidative stress-induced mitochondrial damage, a key pathological feature of neurodegeneration. METHODS: We investigated the impact of Prx II deficiency on endoplasmic reticulum stress and mitochondrial dysfunction using HT22 cell models with knocked down and overexpressed Prx II. We observed alcohol-treated HT22 cells using transmission electron microscopy and monitored changes in the length of mitochondria-associated endoplasmic reticulum membranes and their contact with endoplasmic reticulum mitochondria contact sites (EMCSs). Additionally, RNA sequencing and bioinformatic analysis were conducted to identify the role of Prx II in regulating mitochondrial transport and the formation of EMCSs. RESULTS: Our results indicated that Prx II preserves mitochondrial integrity by facilitating the formation of EMCSs, which are essential for maintaining mitochondrial Ca2+ homeostasis and preventing mitochondria-dependent apoptosis. Further, we identified a novel regulatory axis involving Prx II, the transcription factor ATF3, and miR-181b-5p, which collectively modulate the expression of Armcx3, a protein implicated in mitochondrial transport. Our findings underscore the significance of Prx II in protecting neuronal cells from alcohol-induced oxidative damage and suggest that modulating the Prx II-ATF3-miR-181b-5p pathway may offer a promising therapeutic strategy against neurodegenerative diseases. CONCLUSIONS: This study not only expands our understanding of the cytoprotective mechanisms of Prx II but also offers necessary data for developing targeted interventions to bolster mitochondrial resilience in neurodegenerative conditions.


Subject(s)
MicroRNAs , Mitochondrial Diseases , Neurodegenerative Diseases , Humans , Peroxiredoxins/genetics , Reactive Oxygen Species/metabolism , Oxidative Stress , Apoptosis , Endoplasmic Reticulum Stress , MicroRNAs/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL