Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
Sleep Breath ; 27(2): 421-429, 2023 05.
Article in English | MEDLINE | ID: mdl-35624400

ABSTRACT

PURPOSE: Obstructive sleep apnea-hypopnea syndrome (OSAHS) is characterized by recurrent upper airway disturbances during sleep leading to episodes of hypopnea or apnea, followed by hypoxemia and subsequent reoxygenation. It is believed that this reoxygenation/reperfusion stage leads to oxidative stress, which then leads to inflammation and cardiovascular diseases. The treatments of patient with OSAHS include surgical and non-surgical therapies with various side effects and common complaints. Therefore, it is important to develop a new, safe, and effective therapeutic treatment. As a small-molecule multifunctional protein, thioredoxin (TRX) has antioxidant and redox regulatory functions at the active site Cys-Gly-Pro. TRX prevents inflammation by suppressing the production of pro-inflammatory cytokines rather than suppressing the immune response. METHODS: We review the papers on the pathophysiological process of OSAHS and the antioxidative and anti-inflammatory effects of TRX. RESULTS: TRX may play a role in OSAHS by scavenging ROS, blocking the production of inflammatory cytokines, inhibiting the migration and activation of neutrophils, and controlling the activation of ROS-dependent inflammatory signals by regulating the redox state of intracellular target particles. Furthermore, TRX regulates the synthesis, stability, and activity of hypoxia-inducible factor 1 (HIF-1). TRX also has an inhibitory effect on endoplasmic reticulum- and mitochondria-induced apoptosis by regulating the expression of BAX, BCL2, p53, and ASK1. CONCLUSION: Understanding the function of TRX may be useful for the treatment of OSAHS.


Subject(s)
Sleep Apnea, Obstructive , Humans , Reactive Oxygen Species/metabolism , Sleep Apnea, Obstructive/therapy , Sleep Apnea, Obstructive/metabolism , Antioxidants , Cytokines , Inflammation , Thioredoxins
2.
Nat Immunol ; 11(10): 897-904, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20835230

ABSTRACT

Interleukin 1ß (IL-1ß) is an important inflammatory mediator of type 2 diabetes. Here we show that oligomers of islet amyloid polypeptide (IAPP), a protein that forms amyloid deposits in the pancreas during type 2 diabetes, triggered the NLRP3 inflammasome and generated mature IL-1ß. One therapy for type 2 diabetes, glyburide, suppressed IAPP-mediated IL-1ß production in vitro. Processing of IL-1ß initiated by IAPP first required priming, a process that involved glucose metabolism and was facilitated by minimally oxidized low-density lipoprotein. Finally, mice transgenic for human IAPP had more IL-1ß in pancreatic islets, which localized together with amyloid and macrophages. Our findings identify previously unknown mechanisms in the pathogenesis of type 2 diabetes and treatment of pathology caused by IAPP.


Subject(s)
Amyloid/metabolism , Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/immunology , Interleukin-1beta/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diabetes Mellitus, Type 2/metabolism , Glyburide/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Islet Amyloid Polypeptide , Islets of Langerhans/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Rats , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics
3.
J Biol Chem ; 292(46): 18988-19000, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28939765

ABSTRACT

Thioredoxin 1 (Trx1) is a 12-kDa oxidoreductase that catalyzes thiol-disulfide exchange reactions to reduce proteins with disulfide bonds. As such, Trx1 helps protect the heart against stresses, such as ischemia and pressure overload. Mechanistic target of rapamycin (mTOR) is a serine/threonine kinase that regulates cell growth, metabolism, and survival. We have shown previously that mTOR activity is increased in response to myocardial ischemia-reperfusion injury. However, whether Trx1 interacts with mTOR to preserve heart function remains unknown. Using a substrate-trapping mutant of Trx1 (Trx1C35S), we show here that mTOR is a direct interacting partner of Trx1 in the heart. In response to H2O2 treatment in cardiomyocytes, mTOR exhibited a high molecular weight shift in non-reducing SDS-PAGE in a 2-mercaptoethanol-sensitive manner, suggesting that mTOR is oxidized and forms disulfide bonds with itself or other proteins. The mTOR oxidation was accompanied by reduced phosphorylation of endogenous substrates, such as S6 kinase (S6K) and 4E-binding protein 1 (4E-BP1) in cardiomyocytes. Immune complex kinase assays disclosed that H2O2 treatment diminished mTOR kinase activity, indicating that mTOR is inhibited by oxidation. Of note, Trx1 overexpression attenuated both H2O2-mediated mTOR oxidation and inhibition, whereas Trx1 knockdown increased mTOR oxidation and inhibition. Moreover, Trx1 normalized H2O2-induced down-regulation of metabolic genes and stimulation of cell death, and an mTOR inhibitor abolished Trx1-mediated rescue of gene expression. H2O2-induced oxidation and inhibition of mTOR were attenuated when Cys-1483 of mTOR was mutated to phenylalanine. These results suggest that Trx1 protects cardiomyocytes against stress by reducing mTOR at Cys-1483, thereby preserving the activity of mTOR and inhibiting cell death.


Subject(s)
Myocytes, Cardiac/metabolism , Oxidative Stress , TOR Serine-Threonine Kinases/metabolism , Thioredoxins/metabolism , Animals , Cell Death , Cells, Cultured , Hydrogen Peroxide/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/cytology , Phosphorylation , Rats, Wistar , Ribosomal Protein S6 Kinases/metabolism
4.
J Obstet Gynaecol Res ; 43(1): 50-56, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27762463

ABSTRACT

AIM: Thioredoxin binding protein-2 (TBP-2), which is identical to thioredoxin interacting protein (Txnip), controls cellular proliferation and differentiation. The aim of the present study was to compare TBP-2 protein and mRNA expression in human placenta during the three trimesters of pregnancy and to investigate the role of hypoxia in the change of these expressions in placental tissue. A secondary objective was to determine the gene expression of peroxisome proliferator-activated receptors (PPARs) in TBP-2 deficient placenta using TBP-2 gene disrupted mice (TBP-2-/- ). METHODS: Protein and mRNA expression of TBP-2 in human placenta from each trimester were analyzed by immunohistochemistry, Western blots, and by quantitative reverse-transcriptase-polymerase chain reaction. The effect of hypoxia on TBP-2 expression was tested using an explant culture of human placenta. In TBP-2-/- mouse placenta, we detected PPAR mRNA expression. RESULTS: TBP-2 was located in syncytiotrophoblasts and cytotrophoblasts, and also in the endothelium in human placenta. Its expression in the placenta was low in the first trimester, and increased in the second and third trimesters. Hypoxia decreased TBP-2 mRNA and protein expression in human placental explant culture. In TBP-2-/- mice, placental mRNA levels of PPARα and γ were significantly suppressed compared with those in wild-type mice. CONCLUSION: Hypoxia suppresses TBP-2 gene expression, which may ultimately alter placental development.


Subject(s)
Carrier Proteins/metabolism , Hypoxia/metabolism , Placenta/metabolism , Animals , Carrier Proteins/genetics , Female , Humans , Mice , Mice, Knockout , Peroxisome Proliferator-Activated Receptors/genetics , Pregnancy , Pregnancy Trimesters , RNA, Messenger/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism , Trophoblasts/metabolism
5.
Arch Biochem Biophys ; 595: 88-93, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27095222

ABSTRACT

A 12-kDa protein with redox-active dithiol in the active site -Cys-Gly-Pro-Cys-, human thioredoxin 1 (TRX) has demonstrated an excellent anti-inflammatory effect in various animal models. TRX is induced by various oxidative stress factors, including ultraviolet rays, radiation, oxidation, viral infections, ischemia reperfusion and anticancer agents, and are involved in the pathogenesis and progression of various diseases. We have demonstrated that systemic administration and transgenic overexpression of TRX is effective in a wide variety of in vivo inflammatory disease models, such as viral pneumonia, acute lung injury, chronic obstructive pulmonary disease, indomethacin-induced gastric injury, and dermatitis. Our recent studies indicate that topically applied TRX prevents skin inflammation via the inhibition of local formation of inflammatory cytokines and chemokines. These indicate that the activation of inflammasome in skin and mucosa may be regulated by TRX. These suggest that application of TRX may be useful for the treatment of various skin and mucosal inflammatory disorders. Based on these results, we are conducting clinical studies to develop human recombinant thioredoxin 1 (rhTRX) pharmaceuticals. We have also developed substances that increase the expression of TRX in the body (TRX-inducing substances) in vegetables and other plant ingredients, and we are also developing skin-care products and functional foods that take advantage of the anti-inflammation and anti-allergic action of TRX.


Subject(s)
Inflammation/prevention & control , Oxidative Stress , Thioredoxins/metabolism , Animals , Humans , Oxidation-Reduction
6.
Exp Eye Res ; 152: 71-76, 2016 11.
Article in English | MEDLINE | ID: mdl-27664905

ABSTRACT

Interleukin (IL)-1ß, a proinflammatory cytokine, is a key mediator in several acute and chronic neurological diseases. Thioredoxin-1 (TRX1) acts as an antioxidant and plays a protective role in certain neurons. We examined whether exogenous TRX1 exerts axonal protection and affects IL-1ß levels in tumor necrosis factor (TNF)-induced optic nerve degeneration in rats. Immunoblot analysis showed that IL-1ß was upregulated in the optic nerve after intravitreal injection of TNF. Treatment with recombinant human (rh) TRX1 exerted substantial protective effects against TNF-induced axonal loss. The increase in the IL-1ß level in the optic nerve was abolished by rhTRX1. Treatment with rhTRX1 also significantly inhibited increased glial fibrillary acidic protein (GFAP) levels induced by TNF. Immunohistochemical analysis showed substantial colocalization of IL-1ß and GFAP in the optic nerve after TNF injection. These results suggest that IL-1ß is upregulated in astrocytes in the optic nerve after TNF injection and that exogenous rhTRX1 exerts axonal protection with an inhibitory effect on IL-1ß.


Subject(s)
Interleukin-1beta/antagonists & inhibitors , Nerve Degeneration/prevention & control , Optic Nerve Diseases/prevention & control , Optic Nerve/pathology , Recombinant Proteins/administration & dosage , Thioredoxins/administration & dosage , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Blotting, Western , Humans , Immunoblotting , Immunohistochemistry , Interleukin-1beta/metabolism , Intravitreal Injections , Male , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Optic Nerve/drug effects , Optic Nerve Diseases/metabolism , Optic Nerve Diseases/pathology , Rats , Tumor Necrosis Factor-alpha/toxicity
7.
Pediatr Res ; 80(3): 433-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27100048

ABSTRACT

BACKGROUND: Maternal intrauterine infection/inflammation represents the major etiology of preterm delivery and the leading cause of neonatal mortality and morbidity. The aim of this study was to investigate the anti-inflammatory properties of thioredoxin-1 in vivo and its potential ability to attenuate the rate of inflammation-induced preterm delivery. METHODS: Two intraperitoneal injections of lipopolysaccharide from Escherichia coli were administered in pregnant mice on gestational day 15, with a 3-h interval between the injections. From either 1 h before or 1 h after the first lipopolysaccharide injection, mice received three intravenous injections of either recombinant human thioredoxin-1, ovalbumin, or vehicle, with a 3-h interval between injections. RESULTS: Intraperitoneal injection of lipopolysaccharide induced a rise of tumor necrosis factor-α, interferon-γ, monocyte chemotactic protein 1, and interleukin-6 in maternal serum levels and provoked preterm delivery. Recombinant human thoredoxin-1 prevented the rise in these proinflammatory cytokine levels. After the inflammatory challenge, placentas exhibited severe maternal vascular dilatation and congestion and a marked decidual neutrophil activation. These placental pathological findings were ameliorated by recombinant human thioredoxin-1, and the rate of inflammation-induced preterm delivery was attenuated. CONCLUSION: Thioredoxin-1 may thus represent a novel effective treatment to delay inflammation-induced preterm delivery.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Obstetric Labor, Premature/drug therapy , Thioredoxins/pharmacology , Animals , Animals, Newborn , Chemokine CCL2/blood , Cytokines/blood , Female , Humans , Inflammation , Interferon-gamma/blood , Interleukin-6/blood , Lipopolysaccharides , Macrophages/metabolism , Male , Mice , Mice, Inbred C3H , Obstetric Labor, Premature/chemically induced , Placenta/metabolism , Pregnancy , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology , Thioredoxins/physiology , Tumor Necrosis Factor-alpha/blood
8.
Am J Physiol Renal Physiol ; 307(12): F1342-51, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25350977

ABSTRACT

Oxidative stress is a major determinant of acute kidney injury (AKI); however, the effects of an AKI on renal redox system are unclear, and few existing AKI markers are suitable for evaluating oxidative stress. We measured urinary levels of the redox-regulatory protein thioredoxin 1 (TRX1) in patients with various kinds of kidney disease and in mice with renal ischemia-reperfusion injury. Urinary TRX1 levels were markedly higher in patients with AKI than in those with chronic kidney disease or in healthy subjects. In a receiver operating characteristic curve analysis to differentiate between AKI and other renal diseases, the area under the curve for urinary TRX1 was 0.94 (95% confidence interval, 0.90-0.98), and the sensitivity and specificity were 0.88 and 0.88, respectively, at the optimal cutoff value of 43.0 µg/g creatinine. Immunostaining revealed TRX1 to be diffusely distributed in the tubules of normal kidneys, but to be shifted to the brush borders or urinary lumen in injured tubules in both mice and humans with AKI. Urinary TRX1 in AKI was predominantly in the oxidized form. In cultured human proximal tubular epithelial cells, hydrogen peroxide specifically and dose dependently increased TRX1 levels in the culture supernatant, while reducing intracellular levels. These findings suggest that urinary TRX1 is an oxidative stress-specific biomarker useful for distinguishing AKI from chronic kidney disease and healthy kidneys.


Subject(s)
Acute Kidney Injury/urine , Kidney/metabolism , Oxidative Stress , Reperfusion Injury/urine , Thioredoxins/urine , Acute Kidney Injury/diagnosis , Adult , Aged , Aged, 80 and over , Animals , Area Under Curve , Biomarkers/urine , Case-Control Studies , Diagnosis, Differential , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mice , Middle Aged , Oxidation-Reduction , Predictive Value of Tests , ROC Curve , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/urine , Reperfusion Injury/diagnosis , Time Factors , Up-Regulation
9.
Biol Pharm Bull ; 37(8): 1352-8, 2014.
Article in English | MEDLINE | ID: mdl-25087957

ABSTRACT

Although supplementation with the selenocompound, sodium selenite has been shown to stimulate the concanavalin A-induced T-cell mitogenic response, the mechanisms responsible remain unclear. This study was conducted to evaluate the relationships between the induction of apoptosis, formation of tumor necrosis factor (TNF)-alpha and reactive oxygen species (ROS), activation of apoptosis signal-regulating kinase (ASK) 1 and the thioredoxin (Trx) system when mitogenesis was stimulated by selenite. TNF-alpha was dose-dependently released by mouse splenocytes treated with selenite, and apoptosis was induced when TNF-alpha was added at the indicated concentrations. However, supplementation with selenite at low concentrations inhibited the accumulation of ROS with the increased expression of Trx reductase 1 and induction of apoptosis in wild-type splenocytes, and also at high concentrations in Trx-1-transgenic mouse splenocytes. The suppression of apoptosis was accompanied by a decrease in the expression of phospho-ASK1. These results suggest that the stimulation of T-cell mitogenesis by selenite may be partly attributed to the inhibited accumulation of ROS due to a reduced Trx-1/TR1 system, the inactivation of ASK1, and the suppression of apoptosis.


Subject(s)
MAP Kinase Kinase Kinase 5/metabolism , Reactive Oxygen Species/metabolism , Selenious Acid/pharmacology , T-Lymphocytes/drug effects , Thioredoxins/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Concanavalin A/pharmacology , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitogens/pharmacology , Spleen/cytology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thioredoxins/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Biosci Biotechnol Biochem ; 78(7): 1221-30, 2014.
Article in English | MEDLINE | ID: mdl-25229862

ABSTRACT

Thioredoxin (TRX) is a redox regulating protein which has protective effects against oxidative stress-induced damage to cells and tissues. In this study, we investigated the effects of orally administered TRX derived from edible yeast, Saccharomyces cerevisiae, on gastric mucosa. First, we examined the digestibility of orally administered yeast TRX in mice, and detected yeast TRX in the stomach for 4 h after administration. Next, we investigated the mitigation of gastric mucosal injury after the oral administration of yeast TRX in water-immersion restraint stress and HCl/ethanol-induced gastric ulcer models. Furthermore, we conducted DNA microarray analysis, using the HCl/ethanol-induced model, which revealed that several groups of genes related to tissue repair were upregulated in ulcer regions in the stomachs of rats administered with yeast TRX. These results demonstrated the viability of the use of oral administrations of yeast TRX to protect the gastric mucosa.


Subject(s)
Cytoprotection/drug effects , Gastric Mucosa/drug effects , Gastric Mucosa/injuries , Saccharomyces cerevisiae/chemistry , Thioredoxins/administration & dosage , Thioredoxins/pharmacology , Administration, Oral , Animals , Cell Line , Digestion , Ethanol/adverse effects , Fermentation , Hydrochloric Acid/adverse effects , Male , Mice , Rats , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control , Stomach Ulcer/psychology , Stress, Psychological/prevention & control , Thioredoxins/metabolism
11.
Adv Exp Med Biol ; 801: 829-36, 2014.
Article in English | MEDLINE | ID: mdl-24664777

ABSTRACT

Oxidative stress is a node common to the causes and effects of various ocular diseases. We have shown that thioredoxin has neuroprotective effects on tubby photoreceptors. We also demonstrated that nanoceria (cerium oxide nanoparticles), which are direct antioxidants, have long-term effects on prevention of retinal degeneration in tubby mice. Here, using commercially available PCR array plates, we surveyed the regulation in expression of 89 oxidative stress-associated genes in the eyes of P12 tubby mice which are either intravitreally injected with nanoceria or in which the Trx gene is overexpressed. Our data demonstrate that nanoceria and Trx regulate the same group of genes associated with antioxidative stress and antioxidant defense.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antioxidants/pharmacology , Cerium/pharmacology , Retinal Degeneration , Thioredoxins/pharmacology , Transcriptome/drug effects , Animals , Antioxidants/metabolism , Cerium/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nanoparticles/therapeutic use , Oxidative Stress/drug effects , Oxidative Stress/genetics , Retinal Degeneration/drug therapy , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Thioredoxins/metabolism
12.
Antioxid Redox Signal ; 40(10-12): 595-597, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38386512

ABSTRACT

Recent studies have provided evidence for the direct binding of thioredoxin-1 (TRX1) to a component of inflammasome complex NLR family pyrin domain containing 1 (NLRP-1). This interaction suggests a potential role for TRX1 in the regulation of the NLRP-1 inflammasome. Furthermore, the NLRP-3 inflammasome is known to bind TRX1 and its inhibitor, TRX-binding protein-2/TRX-interacting protein/vitamin D3 upregulated protein-1 (TBP2/TXNIP/VDUP-1). This binding forms a redox-sensitive complex, termed the "Redoxisome," as described previously. However, the specific functions of NLRP-1 within the redoxisome complex remain undefined. Antioxid. Redox Signal. 40, 595-597.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidation-Reduction , Thioredoxins/metabolism
13.
Crit Care Med ; 41(1): 171-81, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23222257

ABSTRACT

OBJECTIVES: Influenza virus infections can cause severe acute lung injury leading to significant morbidity and mortality. Thioredoxin-1 is a redox-active defensive protein induced in response to stress conditions. Animal experiments have revealed that thioredoxin-1 has protective effects against various severe disorders. This study was undertaken to evaluate the protective effects of recombinant human thioredoxin-1 administration on influenza A virus (H1N1)-induced acute lung injury in mice. DESIGN: Prospective animal trial. SETTING: Research laboratory. SUBJECTS: Nine-week-old male C57BL/6 mice inoculated with H1N1. INTERVENTION: The mice were divided into a vehicle-treated group and recombinant human thioredoxin-1-treated group. For survival rate analysis, the vehicle or recombinant human thioredoxin-1 was administered intraperitoneally every second day from day -1 to day 13. For lung lavage and pathological analyses, vehicle or recombinant human thioredoxin-1 was administered intraperitoneally on days -1, 1, and 3. MEASUREMENTS AND MAIN RESULTS: Lung lavage and pathological analyses were performed at 24, 72, and 120 hrs after inoculation. The recombinant human thioredoxin-1 treatment significantly improved the survival rate of H1N1-inoculated mice, although the treatment did not affect virus propagation in the lung. The treatment significantly attenuated the histological changes and neutrophil infiltration in the lung of H1N1-inoculated mice. The treatment significantly attenuated the production of tumor necrosis factor-α and chemokine (C-X-C motif) ligand 1 in the lung and oxidative stress enhancement, which were observed in H1N1-inoculated mice. H1N1 induced expressions of tumor necrosis factor-α and chemokine (C-X-C motif) ligand 1 in murine lung epithelial cells MLE-12, which were inhibited by the addition of recombinant human thioredoxin-1. The recombinant human thioredoxin-1 treatment started 30 mins after H1N1 inoculation also significantly improved the survival of the mice. CONCLUSIONS: Exogenous administration of recombinant human thioredoxin-1 significantly improved the survival rate and attenuated lung histological changes in the murine model of influenza pneumonia. The protective mechanism of thioredoxin-1 might be explained by its potent antioxidative and anti-inflammatory actions. Consequently, recombinant human thioredoxin-1 might be a possible pharmacological strategy for severe influenza virus infection in humans.


Subject(s)
Acute Lung Injury/drug therapy , Antioxidants/therapeutic use , Influenza A Virus, H1N1 Subtype , Influenza, Human/drug therapy , Pneumonia, Viral/drug therapy , Recombinant Proteins/therapeutic use , Thioredoxins/therapeutic use , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Acute Lung Injury/virology , Animals , Antioxidants/pharmacology , Chemokine CXCL1/drug effects , Chemokine CXCL1/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Prospective Studies , Recombinant Proteins/pharmacology , Survival Analysis , Thioredoxins/pharmacology , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism , Viral Load/drug effects
14.
Antioxidants (Basel) ; 12(9)2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37760004

ABSTRACT

Geranylgeranylacetone (GGA) exerts cytoprotective activity against various toxic stressors via the thioredoxin (TRX) redox system; however, its effect on skin inflammation and molecular mechanism on inducing the TRX of GGA is still unknown. We investigated the effects of GGA in a murine irritant contact dermatitis (ICD) model induced by croton oil. Both a topical application and oral administration of GGA induced TRX production and Nrf2 activation. GGA ameliorated ear swelling, neutrophil infiltration, and inhibited the expression of TNF-α, IL-1ß, GM-CSF, and 8-OHdG. GGA's cytoprotective effect was stronger orally than topically in mice. In vitro studies also showed that GGA suppressed the expression of NLRP3, TNF-α, IL-1ß, and GM-CSF and scavenged ROS in PAM212 cells after phorbol myristate acetate stimulation. Moreover, GGA induced endogenous TRX production and Nrf2 nuclear translocation in PAM212 cells (dependent on the presence of ROS) and activated the PI3K-Akt signaling pathway. GGA significantly downregulated thioredoxin-interacting protein (TXNIP) levels in PAM212 cells treated with or without Nrf2 siRNA. After knocking down Nrf2 in PAM212 cells, the effect of GGA on TRX induction was significantly inhibited. This suggests that GGA suppress ICD by inducing endogenous TRX, which may be regulated by PI3K/Akt/Nrf2 mediation of the TRX redox system.

15.
Biochem Biophys Res Commun ; 425(3): 656-61, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22846575

ABSTRACT

There is increasing evidence demonstrating that glutaredoxin 1 (GRX1), a cytosolic enzyme responsible for the catalysis of protein deglutathionylation, plays distinct roles in inflammation and apoptosis by inducing changes in the cellular redox system. In this study, we investigated whether and how the overexpression of GRX1 protects cardiomyocytes against nitric oxide (NO)-induced apoptosis. Cardiomyocytes (H9c2 cells) were transfected with the expression vector for mouse GRX1 cDNA, and mock-transfected cells were used as a control. Compared with the mock-transfected cells, the GRX1-transfected cells were more resistant to NO-induced apoptosis. Stimulation with NO significantly increased the nuclear translocation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a pro-apoptotic protein, in the mock-transfected cells, but did not change GAPDH localization in the GRX1-transfected cells. Furthermore, we found that NO stimulation clearly induced the oxidative modification of GAPDH in the mock-transfected cells, whereas less modification of GAPDH was observed in the GRX1-transfected cells. These data suggest that the overexpression of GRX1 could protect cardiomyocytes against NO-induced apoptosis, likely through the inhibition of the oxidative modification and the nuclear translocation of GAPDH.


Subject(s)
Apoptosis , Cell Nucleus/enzymology , Glutaredoxins/biosynthesis , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Myocytes, Cardiac/physiology , Nitric Oxide/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Glutaredoxins/genetics , Mice , Myocytes, Cardiac/cytology , Oxidation-Reduction , Rats
16.
HPB (Oxford) ; 14(9): 573-82, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22882193

ABSTRACT

BACKGROUND: Multifunctional redox protein human thioredoxin (TRX-1) is reduced by thioredoxin reductase (TRX-R). The aim of the present study was to examine the distribution of TRX-1 and TRX-R expressions in gallbladder carcinoma (GBC) to clarify their usefulness as prognostic factors after surgical resection. METHODS: Immunohistochemical staining for TRX-1 and TRX-R was performed in GBC tissue from 38 patients who underwent surgical resection, and TRX-1/TRX-R localization in relation to outcome was examined. RESULTS: TRX-1 protein levels were significantly higher in GBC samples than in cholecystolithiasis samples (P = 0.0174). TRX-1 expression was observed in 100% (38/38) of tumour samples and in the nucleus in 76% (29/38), with nuclear expression in the invasion front observed in 45% (13/29). TRX-R expression was only detected in the cytoplasm of cancer cells and in the invasion front in 28 samples. In all of the samples, the depth of tumour invasion, lymph node metastasis, surgical margin, curability and nuclear expression of TRX-1 in the invasion front were significant prognostic factors by univariate analysis. In 27 selected patients who underwent curative resection, both TRX-1 nuclear expression and TRX-R cytoplasmic expression in the invasion front was a significantly prognostic factor. CONCLUSION: TRX-1 nuclear expression in the GBC invasion front is a significant prognostic marker. Patients with both TRX-1 nuclear expression and TRX-R cytoplasmic expression in the tumour invasion front should be observed carefully even if after curative resection.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma/chemistry , Cell Nucleus/chemistry , Gallbladder Neoplasms/chemistry , Thioredoxins/analysis , Adult , Aged , Aged, 80 and over , Blotting, Western , Carcinoma/mortality , Carcinoma/secondary , Carcinoma/surgery , Cytoplasm/chemistry , Female , Gallbladder Neoplasms/mortality , Gallbladder Neoplasms/pathology , Gallbladder Neoplasms/surgery , Humans , Immunohistochemistry , Japan , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Thioredoxin-Disulfide Reductase/analysis , Treatment Outcome , Up-Regulation
17.
Antioxid Redox Signal ; 36(13-15): 1051-1065, 2022 05.
Article in English | MEDLINE | ID: mdl-34541903

ABSTRACT

Significance: The development and progression of renal diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD), are the result of heterogeneous pathophysiology that reflects a range of environmental factors and, in a lesser extent, genetic mutations. The pathophysiology specific to most kidney diseases is not currently identified; therefore, these diseases are diagnosed based on non-pathological factors. For that reason, pathophysiology-based companion diagnostics for selection of pathophysiology-targeted treatments have not been available, which impedes personalized medicine in kidney disease. Recent Advances: Pathophysiology-targeted therapeutic agents are now being developed for the treatment of redox dysregulation. Redox modulation therapeutics, including bardoxolone methyl, suppresses the onset and progression of AKI and CKD. On the other hand, pathophysiology-targeted diagnostics for renal redox dysregulation are also being developed. Urinary thioredoxin (TXN) is a biomarker that can be used to diagnose tubular redox dysregulation. AKI causes oxidation and urinary excretion of TXN, which depletes TXN from the tubules, resulting in tubular redox dysregulation. Urinary TXN is selectively elevated at the onset of AKI and correlates with the progression of CKD in diabetic nephropathy. Critical Issues: Diagnostic methods should provide information about molecular mechanisms that aid in the selection of appropriate therapies to improve the prognosis of kidney disease. Future Directions: A specific diagnostic method enabling detection of redox dysregulation based on pathological molecular mechanisms is much needed and could provide the first step toward personalized medicine in kidney disease. Urinary TXN is a candidate for a companion diagnostic method to identify responders to redox-modulating therapeutics. Antioxid. Redox Signal. 36, 1051-1065.


Subject(s)
Acute Kidney Injury , Renal Insufficiency, Chronic , Acute Kidney Injury/diagnosis , Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Biomarkers/urine , Female , Humans , Male , Oleanolic Acid/analogs & derivatives , Oxidation-Reduction , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/etiology , Thioredoxins
18.
Front Immunol ; 13: 883116, 2022.
Article in English | MEDLINE | ID: mdl-35572600

ABSTRACT

Thioredoxin-1 (Trx1) is an important regulator of cellular redox homeostasis that comprises a redox-active dithiol. Trx1 is induced in response to various stress conditions, such as oxidative damage, infection or inflammation, metabolic dysfunction, irradiation, and chemical exposure. It has shown excellent anti-inflammatory and immunomodulatory effects in the treatment of various human inflammatory disorders in animal models. This review focused on the protective roles and mechanisms of Trx1 in allergic diseases, such as allergic asthma, contact dermatitis, food allergies, allergic rhinitis, and drug allergies. Trx1 plays an important role in allergic diseases through processes, such as antioxidation, inhibiting macrophage migration inhibitory factor (MIF), regulating Th1/Th2 immune balance, modulating allergic inflammatory cells, and suppressing complement activation. The regulatory mechanism of Trx1 differs from that of glucocorticoids that regulates the inflammatory reactions associated with immune response suppression. Furthermore, Trx1 exerts a beneficial effect on glucocorticoid resistance of allergic inflammation by inhibiting the production and internalization of MIF. Our results suggest that Trx1 has the potential for future success in translational research.


Subject(s)
Asthma , Rhinitis, Allergic , Animals , Asthma/drug therapy , Inflammation/drug therapy , Oxidation-Reduction , Thioredoxins/metabolism
19.
Carcinogenesis ; 32(10): 1459-66, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21771725

ABSTRACT

Thioredoxin-interacting protein (TXNIP), which has a tumor-suppressive function, is underexpressed in some human cancers. The function of TXNIP in vivo in carcinogenesis is not fully understood. Here, we show TXNIP to be downregulated in human bladder cancer according to grade and stage and also that loss of TXNIP expression facilitates bladder carcinogenesis using a mouse bladder cancer model. N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced bladder cancer was found in 100% of Txnip knockout (KO) mice at week 8 of 0.025% BBN administration but in only 22% of wild-type (WT) mice at the same point. Among growth stimulators, phospho-extracellular signal-regulated kinase (pERK) expression was stronger during bladder carcinogenesis in Txnip-KO mice than in WT mice. We then evaluated TXNIP's effects on ERK activation through various growth stimulators and their receptors. Overexpression of TXNIP in human bladder cancer cells attenuated pERK expression upon stimulation with stromal cell-derived factor-1 (SDF-1) but not with epidermal growth factor or insulin-like growth factor-1. In Txnip-KO mice, immunohistochemical analysis showed enhanced expression of C-X-C chemokine receptor type 4 (CXCR4), the receptor of SDF-1, and of pERK in urothelial cells during BBN-induced bladder carcinogenesis. Finally, subcutaneous injection of CXCR4 antagonist, TF14016, attenuated pERK in urothelial cells and suppressed bladder carcinogenesis. These data indicate that TXNIP negatively regulates bladder carcinogenesis by attenuating SDF-1-CXCR4-induced ERK activation. This signal transduction pathway can be a potent target in preventing or treating bladder cancer.


Subject(s)
Carrier Proteins/metabolism , Carrier Proteins/physiology , Signal Transduction , Thioredoxins/physiology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Aged , Animals , Blotting, Western , Butylhydroxybutylnitrosamine/toxicity , Carrier Proteins/genetics , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Thioredoxins/metabolism , Urinary Bladder Neoplasms/chemically induced
20.
Plant Mol Biol ; 76(3-5): 335-44, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21290168

ABSTRACT

The production of human therapeutic proteins in plants provides opportunities for low-cost production, and minimizes the risk of contamination from potential human pathogens. Chloroplast genetic engineering is a particularly promising strategy, because plant chloroplasts can produce large amounts of foreign target proteins. Oxidative stress is a key factor in various human diseases. Human thioredoxin 1 (hTrx1) is a stress-induced protein that functions as an antioxidant against oxidative stress, and overexpression of hTrx1 has been shown to suppress various diseases in mice. Therefore, hTrx1 is a prospective candidate as a new human therapeutic protein. We created transplastomic lettuce expressing hTrx1 under the control of the psbA promoter. Transplastomic plants grew normally and were fertile. The hTrx1 protein accumulated to approximately 1% of total soluble protein in mature leaves. The hTrx1 protein purified from lettuce leaves was functionally active, and reduced insulin disulfides. The purified protein protected mouse insulinoma line 6 cells from damage by hydrogen peroxide, as reported previously for a recombinant hTrx1 expressed in Escherichia coli. This is the first report of expression of the biologically active hTrx1 protein in plant chloroplasts. This research opens up possibilities for plant-based production of hTrx1. Considering that this expression host is an edible crop plant, this transplastomic lettuce may be suitable for oral delivery of hTrx1.


Subject(s)
Chloroplasts/metabolism , Lactuca/metabolism , Thioredoxins/biosynthesis , Base Sequence , DNA Primers , Humans , Plastids , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL