Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 17934, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087733

ABSTRACT

PRDX4, a member of peroxiredoxin family, is largely concentrated in the endoplasmic reticulum (ER) and plays a pivotal role in the redox relay during oxidative protein folding as well as in peroxidase reactions. A testis-specific PRDX4 variant transcript (PRDX4t) lacks the conventional exon 1, which encodes the signal peptide that is required for entry into the ER lumen, but instead carries alternative exon 1, which is transcribed from the upstream promoter in a testis-specific manner and results in the PRDX4t protein being localized in the cytosol. However, the potential roles of PRDX4t in male genital action remain unknown. Using a CRISPR/Cas9 system, we first disrupted the testis-specific promoter/exon 1 and generated mice that were specifically deficient in PRDX4t. The resulting PRDX4t knockout (KO) mice underwent normal spermatogenesis and showed no overt abnormalities in the testis. Mating PRDX4t KO male mice with wild-type (WT) female mice produced normal numbers of offspring, indicating that a PRDX4t deficiency alone had no effect on fertility in the male mice. We then generated mice lacking both PRDX4 and PRDX4t by disrupting exon 2, which is communal to these variants. The resulting double knockout (DKO) mice were again fertile, and mature sperm isolated from the epididymis of DKO mice exhibited a normal fertilizing ability in vitro. In the meantime, the protein levels of glutathione peroxidase 4 (GPX4), which plays an essential role in the disulfide bond formation during spermatogenesis, were significantly increased in the testis and caput epididymis of the DKO mice compared with the WT mice. Based on these results, we conclude that the disruption of the function of PRDX4t in the spermatogenic process appears to be compensated by other factors including GPX4.


Subject(s)
Fertility/genetics , Genetic Variation/genetics , Peroxiredoxins/genetics , Peroxiredoxins/physiology , Spermatogenesis/genetics , Animals , Exons , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/genetics , Peroxiredoxins/deficiency , Peroxiredoxins/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology , Pregnancy , Testis/metabolism
2.
Exp Mol Med ; 52(9): 1587-1601, 2020 09.
Article in English | MEDLINE | ID: mdl-32929220

ABSTRACT

Abdominal aortic aneurysm (AAA) is an inflammatory vascular disease characterized by structural deterioration of the aorta caused by inflammation and oxidative stress, leading to aortic dilatation and rupture. Peroxiredoxin 2 (PRDX2), an antioxidant enzyme, has been reported as a potential negative regulator of inflammatory vascular diseases, and it has been identified as a protein that is increased in patients with ruptured AAA compared to patients with nonruptured AAA. In this study, we demonstrated that PRDX2 was a pivotal factor involved in the inhibition of AAA progression. PRDX2 levels were increased in AAA compared with those in normal aortas in both humans and mice. Ultrasound imaging revealed that the loss of PRDX2 accelerated the development of AAA in the early stages and increased AAA incidence in mice infused with angiotensin II (Ang II). Prdx2-/- mice infused with Ang II exhibited increased aortic dilatation and maximal aortic diameter without a change in blood pressure. Structural deterioration of the aortas from Prdx2-/- mice infused with Ang II was associated with increases in the degradation of elastin, oxidative stress, and intramural thrombi caused by microhemorrhages, immature neovessels, and the activation of matrix metalloproteinases compared to that observed in controls. Moreover, an increase in inflammatory responses, including the production of cell adhesion molecules and the accumulation of inflammatory cells and proinflammatory cytokines due to PRDX2 deficiency, accelerated Ang II-induced AAA progression. Our data confirm that PRDX2 plays a role as a negative regulator of the pathological process of AAA and suggest that increasing PRDX2 activity may be a novel strategy for the prevention and treatment of AAA.


Subject(s)
Angiotensin II/adverse effects , Aortic Aneurysm, Abdominal/etiology , Aortic Aneurysm, Abdominal/pathology , Disease Susceptibility , Peroxiredoxins/deficiency , Animals , Aortic Aneurysm, Abdominal/diagnostic imaging , Biomarkers , Biopsy , Cell Adhesion Molecules/metabolism , Cytokines/metabolism , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Mice , Mice, Knockout , Models, Biological , Myocytes, Smooth Muscle/metabolism , Peroxiredoxins/genetics , Reactive Oxygen Species , Ultrasonography
3.
Biochem Biophys Res Commun ; 529(3): 635-641, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32736685

ABSTRACT

Keratinocyte hyperproliferation is an essential link in skin cancer pathogenesis. Peroxiredoxin I (Prx I) is known to regulate cancer cell proliferation, differentiation, and apoptosis, but its role in skin cancer remains unclear. This study aimed to elucidate the role and mechanism of Prx I in skin cancer pathogenesis. Dimethylbenz[a]anthracene (DMBA) and 12-O-tetradecanoyl-phorbol-13-acetate (TPA) were used to create a skin tumor model of the initiation/promotion stage of cancer. The role of Prx I in H2O2-induced keratinocyte apoptosis was also investigated. After DMBA/TPA treatment, Prx I deficiency was significantly associated with less skin tumors, lower Bcl-2 expression, and higher p-p38 and cleaved caspase-3 expressions in Prx I knockout tumors than in wild-type controls. H2O2 stimulation caused more cellular apoptosis in Prx I knockdown HaCaT cells than in normal HaCaT cells. The signaling study revealed that Bcl-2, p-p38, and cleaved caspase-3 expressions were consistent with the results in the tumors. In conclusion, the deletion of Prx I triggered the DMBA/TPA-induced skin tumor formation in vivo and in vitro by regulating the reactive oxygen species (ROS)-p38 mitogen-activated protein kinase (MAPK) pathway. These findings provide a theoretical basis for treating skin cancer.


Subject(s)
Apoptosis/genetics , Keratinocytes/metabolism , Peroxiredoxins/genetics , Reactive Oxygen Species/metabolism , Skin Neoplasms/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line , HEK293 Cells , Humans , Hydrogen Peroxide/pharmacology , Keratinocytes/cytology , Keratinocytes/drug effects , Mice, 129 Strain , Mice, Knockout , Oxidants/pharmacology , Peroxiredoxins/deficiency , RNA Interference , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
4.
Reprod Fertil Dev ; 32(8): 783-791, 2020 May.
Article in English | MEDLINE | ID: mdl-32389179

ABSTRACT

Peroxiredoxin 2 (Prdx2), an antioxidant enzyme, is expressed in the ovary during the ovulatory process. The aim of the present study was to examine the physiological role of Prdx2 during ovulation using Prdx2-knockout mice and mouse cumulus-oocyte complex (COC) from WT mice. Two days of treatment of immature mice (21-23 days old) with equine chorionic gonadotrophin and followed by treatment with human chorionic gonadotrophin greatly impaired cumulus expansion and oocyte maturation in Prdx2-knockout but not wild-type mice. Treatment of COCs in culture with conoidin A (50µM), a 2-cys Prdx inhibitor, abolished epiregulin (EPI)-induced cumulus expansion. Conoidin A treatment also inhibited EPI-stimulated signal molecules, including signal transducer and activator of transcription-3, AKT and mitogen-activated protein kinase 1/2. Conoidin A treatment also reduced the gene expression of EPI-stimulated expansion-inducing factors (hyaluronan synthase 2 (Has2), pentraxin 3 (Ptx3), TNF-α induced protein 6 (Tnfaip6) and prostaglandin-endoperoxide synthase 2 (Ptgs2)) and oocyte-derived factors (growth differentiation factor 9 (Gdf9) and bone morphogenetic protein 15 (Bmp15)). Furthermore, conoidin A inhibited EPI-induced oocyte maturation and the activity of connexins 43 and 37. Together, these results demonstrate that Prdx2 plays a role in regulating cumulus expansion and oocyte maturation during the ovulatory process in mice, probably by modulating epidermal growth factor receptor signalling.


Subject(s)
Cumulus Cells/physiology , Oocytes/growth & development , Ovulation/physiology , Peroxiredoxins/physiology , Animals , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Cumulus Cells/drug effects , Female , Gonadotropins, Equine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/drug effects , Peroxiredoxins/antagonists & inhibitors , Peroxiredoxins/deficiency , Quinoxalines/pharmacology
5.
Cell Death Dis ; 11(3): 204, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32205843

ABSTRACT

Iron is an essential element for cellular functions, including those of neuronal cells. However, an imbalance of iron homeostasis, such as iron overload, has been observed in several neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Iron overload causes neuronal toxicity through mitochondrial fission, dysregulation of Ca2+, ER-stress, and ROS production. Nevertheless, the precise mechanisms between iron-induced oxidative stress and iron toxicity related to mitochondria and endoplasmic reticulum (ER) in vivo are not fully understood. Here, we demonstrate the role of peroxiredoxin 5 (Prx5) in iron overload-induced neurotoxicity using Prx5-deficient mice. Iron concentrations and ROS levels in mice fed a high iron diet were significantly higher in Prx5-/- mice than wildtype (WT) mice. Prx5 deficiency also exacerbated ER-stress and ER-mediated mitochondrial fission via Ca2+/calcineurin-mediated dephosphorylation of Drp1 at Serine 637. Moreover, immunoreactive levels of cleaved caspase3 in the CA3 region of the hippocampus were higher in iron-loaded Prx5-/- mice than WT mice. Furthermore, treatment with N-acetyl-cysteine, a reactive oxygen species (ROS) scavenger, attenuated iron overload-induced hippocampal damage by inhibiting ROS production, ER-stress, and mitochondrial fission in iron-loaded Prx5-/- mice. Therefore, we suggest that iron overload-induced oxidative stress and ER-mediated mitochondrial fission may be essential for understanding iron-mediated neuronal cell death in the hippocampus and that Prx5 may be useful as a novel therapeutic target in the treatment of iron overload-mediated diseases and neurodegenerative diseases.


Subject(s)
Hippocampus/metabolism , Iron Overload/metabolism , Mitochondrial Dynamics/physiology , Neurons/metabolism , Peroxiredoxins/deficiency , Animals , Cell Death/physiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress , Female , Hippocampus/pathology , Mice , Mice, Inbred C57BL , Neurons/pathology , Peroxiredoxins/genetics , Pregnancy , Signal Transduction
6.
Microbiologyopen ; 8(10): e889, 2019 10.
Article in English | MEDLINE | ID: mdl-31177643

ABSTRACT

Peroxiredoxins (Prxs) play an essential role in the antioxidant activity and symbiotic capacity of Mesorhizobium huakuii. A mutation in the M. huakuii prxA gene (encoding a Prx5-like peroxiredoxin) was generated by homologous recombination. The mutation of prxA did not affect M. huakuii growth, but the strain displayed decreased antioxidative capacity under organic cumene hydroperoxide (CUOOH) conditions. The higher resistance of the prxA mutant strain compared with the wild-type strain to more than 1 mmol/L H2 O2 was associated with a significantly higher level of glutathione reductase activity and a significantly lower level of intracellular hydrogen peroxide content. Real-time quantitative PCR showed that under 1 mmol/L H2 O2 conditions, expression of the stress-responsive genes katG and katE was significantly upregulated in the prxA mutant. Although the prxA mutant can form nodules, the symbiotic ability was severely impaired, which led to an abnormal nodulation phenotype coupled to a 53.25% reduction in nitrogen fixation capacity. This phenotype was linked to an absence of bacteroid differentiation and deregulation of the transcription of the symbiotic genes nifH, nifD, and fdxN. Expression of the prxA gene was induced during symbiosis. Thus, the PrxA protein is essential for antioxidant capacity and symbiotic nitrogen fixation, playing independent roles in bacterial differentiation and cellular antioxidative systems.


Subject(s)
Antioxidants/metabolism , Mesorhizobium/growth & development , Mesorhizobium/metabolism , Nitrogen Fixation , Peroxiredoxins/metabolism , Symbiosis , Astragalus Plant/microbiology , Gene Expression Profiling , Oxidative Stress , Peroxiredoxins/deficiency , Peroxiredoxins/genetics , Plant Root Nodulation , Real-Time Polymerase Chain Reaction
7.
Parasite ; 25: 57, 2018.
Article in English | MEDLINE | ID: mdl-30474598

ABSTRACT

Cystic echinococcosis (CE) is a cosmopolitan parasitic disease caused by infection with the larval stage of Echinococcus granulosus sensu lato. Thioredoxin peroxidase (TPx) may play an essential role in the antioxidant defence system of E. granulosus s.l. as neither catalase nor glutathione peroxidase activities have been detected in the parasite. However, it is not known whether TPx affects the survival and growth of E. granulosus s.l. during development. In this study, three fragments of siRNA specific for EgTPx (siRNA-1/2/3) were designed and transfected into protoscoleces of E. granulosus sensu stricto by electroporation. Quantitative real-time PCR and Western blotting analysis showed that siRNA-3 significantly reduced the expression of EgTPx. Coincidentally, knockdown of EgTPx expression in protoscoleces with siRNA-3 significantly reduced the viability of the parasite under oxidative stress induced by 0.6 mM H2O2. In vitro culture studies showed that protoscoleces treated with siRNA-3 reduced pre-microcyst formation. In vivo experiments showed that injecting mice intraperitoneally with protoscoleces treated with siRNA-3 resulted in a significant reduction in the number, size and weight of CE cysts compared with those of control animals. Silencing of EgTPx led to the impairment of growth of E. granulosus s.s. both in vitro and in vivo, indicating that EgTPx is an important factor for protoscoleces survival and plays an important role in the antioxidant defence against the host during development.


Subject(s)
Echinococcus granulosus/enzymology , Echinococcus granulosus/physiology , Peroxiredoxins/deficiency , Peroxiredoxins/genetics , Animals , Blotting, Western , Echinococcosis/parasitology , Echinococcus granulosus/genetics , Echinococcus granulosus/growth & development , Electroporation , Female , Gene Silencing , Hydrogen Peroxide/pharmacology , Larva/physiology , Mice , Mice, Inbred BALB C , RNA Interference , RNA, Small Interfering/physiology , Real-Time Polymerase Chain Reaction , Sheep , Transfection
8.
Autophagy ; 14(1): 120-133, 2018.
Article in English | MEDLINE | ID: mdl-28605287

ABSTRACT

Oxidative stress activates macroautophagy/autophagy and contributes to atherogenesis via lipophagic flux, a form of lipid removal by autophagy. However, it is not known exactly how endogenous antioxidant enzymes are involved in lipophagic flux. Here, we demonstrate that the antioxidant PRDX1 (peroxiredoxin 1) has a crucial role in the maintenance of lipophagic flux in macrophages. PRDX1 is more highly expressed than other antioxidant enzymes in monocytes and macrophages. We determined that Prdx1 deficiency induced excessive oxidative stress and impaired maintenance of autophagic flux in macrophages. Prdx1-deficient macrophages had higher intracellular cholesterol mass and lower cholesterol efflux compared with wild type. This perturbation in cholesterol homeostasis was due to impaired lipophagic cholesterol hydrolysis caused by excessive oxidative stress, resulting in the inhibition of free cholesterol formation and the reduction of NR1H3 (nuclear receptor subfamily 1, group H, member 3) activity. Notably, impairment of both lipophagic flux and cholesterol efflux was restored by the 2-Cys PRDX-mimics ebselen and gliotoxin. Consistent with this observation, apoe -/- mice transplanted with bone marrow from prdx1-/-apoe-/- mice had increased plaque formation compared with apoe-/- BM-transplanted recipients. This study reveals that PRDX1 is crucial to regulating lipophagic flux and maintaining macrophage cholesterol homeostasis against oxidative stress. We suggest that PRDX1-dependent control of oxidative stress may provide a strategy for treating atherosclerosis and autophagy-related human diseases.


Subject(s)
Autophagy , Cholesterol/metabolism , Macrophages/metabolism , Oxidative Stress , Peroxiredoxins/deficiency , Animals , Atherosclerosis/enzymology , Cells, Cultured , Humans , Liver X Receptors/metabolism , Mice , Mice, Knockout , Peroxiredoxins/chemistry , Peroxiredoxins/genetics , Peroxiredoxins/therapeutic use
9.
Sci Rep ; 6: 35995, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27786251

ABSTRACT

Peroxiredoxin (PRX), a family of peroxidases, is associated with various biological processes such as the detoxification of oxidants and cell apoptosis. Besides, the anti-apoptosis effect of estrogen results partially from its anti-oxidant function. The purpose of this study was to investigate the expression of PRXs in ovariectomy (OVX) mice and the related anti-oxidative mechanism of estrogen. Eight-week-old mice were subjected to ovariectomy. MC3T3-E1 cells were pretreatment with 17b-estradiol and N-acetyl cysteine followed by oxidative injury induced with H2O2. Western blot and real time-PCR were applied to clarify the expressions of PRX1 and caspase-3, with both wild-type and PRX1 knockout MC3T3-E1 cells generated by CRISPR/Cas9 technology. The results showed PRX1 and PRX5 were upregulated in osteoblasts in the proximal tibial metaphysis of ovariectomy mice. Interestingly, PRX1 and PRX5 showed different distribution patterns, with PRX1 mainly accumulated in cell nuclei and PRX5 in the cytoplasm. Gene expression analysis showed significantly reduced expressions of PRX1 and caspase-3 in the pretreatment groups when compared with cells treated with H2O2 alone. Also, a decrease of caspase-3 expressions was observed in PRX1 knockout MC3T3-E1 cells with or without H2O2 in comparison to wild-type cells. These findings suggested that PRX may play important roles in estrogen-deficient osteoporosis. (200 words).


Subject(s)
Osteoblasts/metabolism , Peroxiredoxins/metabolism , 3T3 Cells , Animals , Apoptosis/drug effects , CRISPR-Cas Systems , Caspase 3/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Estrogens/deficiency , Female , Gene Knockout Techniques , Hydrogen Peroxide/pharmacology , Mice , Osteoblasts/drug effects , Osteoblasts/pathology , Osteoporosis/etiology , Osteoporosis/metabolism , Osteoporosis/pathology , Ovariectomy/adverse effects , Peroxiredoxins/deficiency , Peroxiredoxins/genetics , Up-Regulation
10.
J Immunol ; 197(8): 3233-3244, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27605010

ABSTRACT

Peroxiredoxin (PRDX)1 is an antioxidant that detoxifies hydrogen peroxide and peroxinitrite. Compared with wild-type (WT) mice, Prdx1-deficient (Prdx1-/-) mice showed increased susceptibility to Mycobacterium tuberculosis and lower levels of IFN-γ and IFN-γ-producing CD4+ T cells in the lungs after M. tuberculosis infection. IL-12 production, c-Rel induction, and p38 MAPK activation levels were lower in Prdx1-/- than in WT bone marrow-derived macrophages (BMDMs). IFN-γ-activated Prdx1-/- BMDMs did not kill M. tubercuosis effectively. NO production levels were lower, and arginase activity and arginase 1 (Arg1) expression levels were higher, in IFN-γ-activated Prdx1-/- than in WT BMDMs after M. tuberculosis infection. An arginase inhibitor, Nω-hydroxy-nor-arginine, restored antimicrobial activity and NO production in IFN-γ-activated Prdx1-/- BMDMs after M. tuberculosis infection. These results suggest that PRDX1 contributes to host defenses against M. tuberculosis PRDX1 positively regulates IL-12 production by inducing c-Rel and activating p38 MAPK, and it positively regulates NO production by suppressing Arg1 expression in macrophages infected with M. tuberculosis.


Subject(s)
Mycobacterium tuberculosis/immunology , Peroxiredoxins/immunology , Animals , Interleukin-12/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Peroxiredoxins/deficiency
11.
Neuroscience ; 311: 539-51, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26546471

ABSTRACT

Parkinson's disease (PD) is a well-characterized neurological disorder with regard to its neuropathological and symptomatic appearance. At the genetic level, mutations of particular genes, e.g. Parkin and DJ-1, were found in human hereditary PD with early onset. Neurotransmitter receptors constitute decisive elements in neural signal transduction. Furthermore, since they are often altered in neurological and psychiatric diseases, receptors have been successful targets for pharmacological agents. However, the consequences of PD-associated gene mutations on the expression of transmitter receptors are largely unknown. Therefore, we studied the expression of 16 different receptor binding sites of the neurotransmitters glutamate, GABA, acetylcholine, adrenaline, serotonin, dopamine and adenosine by means of quantitative receptor autoradiography in Parkin and DJ-1 knockout mice. These knockout mice exhibit electrophysiological and behavioral deficits, but do not show the typical dopaminergic cell loss. We demonstrated differential changes of binding site densities in eleven brain regions. Most prominently, we found an up-regulation of GABA(B) and kainate receptor densities in numerous cortical areas of Parkin and DJ-1 knockout mice, as well as increased NMDA but decreased AMPA receptor densities in different brain regions of the Parkin knockout mice. The alterations of three different glutamate receptor types may indicate the potential relevance of the glutamatergic system in the pathogenesis of PD. Furthermore, the cholinergic M1, M2 and nicotinic receptors as well as the adrenergic α2 and the adenosine A(2A) receptors showed differentially increased densities in Parkin and DJ-1 knockout mice. Taken together, knockout of the PD-associated genes Parkin or DJ-1 results in differential changes of neurotransmitter receptor densities, highlighting a possible role of altered non-dopaminergic, and in particular of glutamatergic neurotransmission in PD pathogenesis.


Subject(s)
Brain/metabolism , Oncogene Proteins/genetics , Peroxiredoxins/genetics , Receptors, Neurotransmitter/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Autoradiography , Brain/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins/deficiency , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Peroxiredoxins/deficiency , Protein Deglycase DJ-1 , Ubiquitin-Protein Ligases/deficiency
12.
Life Sci ; 143: 50-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26408915

ABSTRACT

AIMS: The purpose of this study is to explore whether antioxidant DJ-1 protein affects the atrophy of skeletal muscle cell induced by undernutrition. MAIN METHODS: To determine cell atrophic responses, L6 cell line and skeletal primary cells from mouse hind limbs were cultivated under condition of FBS-free and low glucose. Changes of protein expression were analyzed using Western blot. Overexpression and knockdown of DJ-1 was performed in cells to assess its influence on cell atrophic responses. KEY FINDINGS: Undernutrition decreased cell size and increased the abundance of oxidized form and total form of DJ-1 protein in L6 myoblasts. The undernourished cells revealed an elevation in the expression of muscle-specific RING finger-1 (MuRF-1) and atrogin-1, and in the phosphorylations of p38 mitogen-activated protein kinase (MAPK) and stress-activated protein kinase/c-Jun N-terminal kinase compared with control groups. Moreover, DJ-1-knockout mice showed a decrease in cell size and an enhancement in the expression of MuRF-1 and atrogin-1, as well as in the phosphorylation of MAPKs in gastrocnemius muscles; these changes were also observed in L6 cells transfected with siRNA of DJ-1. On the other hand, L6 cells overexpressing full-length DJ-1 did not exhibit the alterations in cell size and ubiquitin ligases seen after undernourished states of control cells. Myotubes differentiated from L6 cells also showed elevated expression of MuRF-1 and atrogin-1 in response to undernutrition. SIGNIFICANCE: These results suggest that DJ-1 protein may contribute to undernutrition-induced atrophy via MAPKs/ubiquitin ligase pathway in skeletal muscle cells.


Subject(s)
MAP Kinase Signaling System/physiology , Malnutrition/metabolism , Myoblasts/enzymology , Oncogene Proteins/deficiency , Peroxiredoxins/deficiency , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Animals , Atrophy/enzymology , Atrophy/prevention & control , Cell Line , Female , Male , Malnutrition/prevention & control , Mice , Mice, Knockout , Myoblasts/pathology , Organ Culture Techniques , Protein Deglycase DJ-1 , Rats
13.
Cell Res ; 25(6): 691-706, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26021615

ABSTRACT

Inappropriate inflammation responses contribute to mortality during sepsis. Through Toll-like receptors (TLRs), reactive oxygen species (ROS) produced by NADPH oxidase could modulate the inflammation responses. Parkinson disease (autosomal recessive, early onset) 7 (Park7) has a cytoprotective role by eliminating ROS. However, whether Park7 could modulate inflammation responses and mortality in sepsis is unclear. Here, we show that, compared with wild-type mice, Park7(-/-) mice had significantly increased mortality and bacterial burdens in sepsis model along with markedly decreased systemic and local inflammation, and drastically impaired macrophage phagocytosis and bacterial killing abilities. Surprisingly, LPS and phorbol-12-myristate-13-acetate stimulation failed to induce ROS and proinflammatory cytokine production in Park7(-/-) macrophages and Park7-deficient RAW264.7 cells. Through its C-terminus, Park7 binds to p47(phox), a subunit of the NADPH oxidase, to promote NADPH oxidase-dependent production of ROS. Restoration of Park7 expression rescues ROS production and improves survival in LPS-induced sepsis. Together, our study shows that Park7 has a protective role against sepsis by controlling macrophage activation, NADPH oxidase activation and inflammation responses.


Subject(s)
NADPH Oxidases/metabolism , Oncogene Proteins/metabolism , Peroxiredoxins/metabolism , Reactive Oxygen Species/metabolism , Sepsis/prevention & control , Animals , Lipopolysaccharides/administration & dosage , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins/biosynthesis , Oncogene Proteins/deficiency , Peroxiredoxins/biosynthesis , Peroxiredoxins/deficiency , Protein Deglycase DJ-1 , Sepsis/chemically induced , Sepsis/metabolism
14.
Aging Cell ; 14(4): 558-68, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25808059

ABSTRACT

Peroxiredoxins (Prx) are abundant thiol peroxidases with a conserved anti-ageing role. In contrast to most animals, the nematode worm, Caenorhabditis elegans, encodes a single cytosolic 2-Cys Prx, PRDX-2, rendering it an excellent model for examining how peroxiredoxins affect animal physiology and ageing. Our previous work revealed that, although PRDX-2 protects against the toxicity of peroxides, enigmatically, prdx-2-mutant animals are hyper-resistant to other forms of oxidative stress. Here, we have investigated the basis for this increased resistance. Mammalian FOXO and Nrf2 transcription factors directly promote the expression of a range of detoxification enzymes. We show that the FOXO orthologue, DAF-16, and the Nrf2 orthologue, SKN-1, are required for the increased stress resistance of prdx-2-mutant worms. Our data suggest that PRDX-2 is required for normal levels of insulin secretion and hence the inhibition of DAF-16 and SKN-1 by insulin/IGF-1-like signalling (IIS) under nutrient-rich conditions. Intriguingly, loss of PRDX-2 increases DAF-16 and SKN-1 activities sufficiently to increase arsenite resistance without initiating other IIS-inhibited processes. Together, these data suggest that loss of peroxiredoxin function may increase stress resistance by reducing insulin secretion, but that further changes in insulin signalling are required for the reprogramming of development and fat metabolism. In addition, we reveal that the temperature-dependent prolongevity function of PRDX-2 is required for the extended lifespan associated with several pathways, including further reductions in IIS.


Subject(s)
Aging/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Insulin/metabolism , Longevity/genetics , Peroxiredoxins/genetics , Aging/metabolism , Animals , Arsenites/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Insulin Secretion , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Larva/drug effects , Larva/genetics , Larva/metabolism , Lipid Metabolism/drug effects , Oxidative Stress , Peroxiredoxins/deficiency , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Biol Chem ; 290(18): 11432-42, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25802339

ABSTRACT

Collagen-induced platelet signaling is mediated by binding to the primary receptor glycoprotein VI (GPVI). Reactive oxygen species produced in response to collagen have been found to be responsible for the propagation of GPVI signaling pathways in platelets. Therefore, it has been suggested that antioxidant enzymes could down-regulate GPVI-stimulated platelet activation. Although the antioxidant enzyme peroxiredoxin II (PrxII) has emerged as having a role in negatively regulating signaling through various receptors by eliminating H2O2 generated upon receptor stimulation, the function of PrxII in collagen-stimulated platelets is not known. We tested the hypothesis that PrxII negatively regulates collagen-stimulated platelet activation. We analyzed PrxII-deficient murine platelets. PrxII deficiency enhanced GPVI-mediated platelet activation through the defective elimination of H2O2 and the impaired protection of SH2 domain-containing tyrosine phosphatase 2 (SHP-2) against oxidative inactivation, which resulted in increased tyrosine phosphorylation of key components for the GPVI signaling cascade, including Syk, Btk, and phospholipase Cγ2. Interestingly, PrxII-mediated antioxidative protection of SHP-2 appeared to occur in the lipid rafts. PrxII-deficient platelets exhibited increased adhesion and aggregation upon collagen stimulation. Furthermore, in vivo experiments demonstrated that PrxII deficiency facilitated platelet-dependent thrombus formation in injured carotid arteries. This study reveals that PrxII functions as a protective antioxidant enzyme against collagen-stimulated platelet activation and platelet-dependent thrombosis.


Subject(s)
Antioxidants/metabolism , Blood Platelets/drug effects , Blood Platelets/physiology , Collagen/pharmacology , Peroxiredoxins/metabolism , Platelet Membrane Glycoproteins/metabolism , Animals , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/physiopathology , Hydrogen Peroxide/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Peroxiredoxins/deficiency , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Thrombosis/metabolism , Thrombosis/physiopathology , Tyrosine/metabolism
16.
Cancer Lett ; 361(2): 262-70, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25779674

ABSTRACT

Bone is a common site of metastasis from breast and prostate carcinoma, where activation of bone resorbing osteoclasts is important for cancer progression. A large body of evidence indicates that soluble factors produced by the cancer cells act to promote osteoclast formation. Using mass spectrometry, we identified peroxiredoxin (PRDX) as a secreted mediator of cancer-induced osteoclastogenesis. Both breast (MCF7 and MDA-MB-231) and prostate (PC3 and LNCaP) carcinoma cells secreted PRDX4. PRDX4 knockdown using shRNA (shPRDX4) diminished PRDX4 secretion from MDA-MB-231 and PC3 cells and significantly decreased the ability of cancer-derived factors to induce osteoclast formation from late precursors in vitro. Tibial injection of shPRDX4 PC3 cells led to the development of significantly smaller osteolytic lesions characterized by significantly reduced osteoclast numbers compared to control PC3 cells. A meta-analysis demonstrated an increase in PRDX4 mRNA expression in carcinoma and metastatic breast and prostate tissues. Moreover, high expression of PRDX4 in the primary breast tumor was consistently associated with metastasis at 5 years. These data identify a novel function of secreted PRDX4 in mediating osteoclast activation by cancer cells.


Subject(s)
Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Osteoclasts/metabolism , Osteogenesis , Peroxiredoxins/metabolism , Animals , Bone Neoplasms/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , MCF-7 Cells , Male , Mice , Mice, Nude , Neoplasm Metastasis , Osteoclasts/pathology , Peroxiredoxins/deficiency , Peroxiredoxins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
17.
J Nutr Biochem ; 26(1): 75-81, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25448609

ABSTRACT

DJ-1 constitutes a ubiquitously expressed, oxidative stress-responsive protein with multiple functions. DJ-1 emerged as a candidate from our previous proteome analysis investigating alterations in the hypothalamus in three mouse strains differing in their susceptibility to diet-induced obesity (DIO). Validation studies demonstrated a high-fat diet (HFD)-induced shift in the DJ-1 isoform pattern in the hypothalamus and several other tissues of mice. Others found HFD-induced alterations in DJ-1 protein abundance in adipose tissue and pancreatic islets in wild-type rodents. Here, we investigated the gene-diet interaction by challenging Dj-1(-/-) mice with a HFD. We demonstrate that the development of diet-induced obesity (DIO) Dj-1(-/-) mice is according to wild-type mice with the exception of transient higher gains in fat mass at the expense of lean mass after 14 weeks of feeding.


Subject(s)
Diet, High-Fat/adverse effects , Obesity/physiopathology , Oncogene Proteins/deficiency , Peroxiredoxins/deficiency , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Calorimetry, Indirect , Energy Intake , Female , Glucose Tolerance Test , Hypothalamus/metabolism , Insulin/blood , Islets of Langerhans/metabolism , Leptin/administration & dosage , Leptin/blood , Male , Mice , Mice, Knockout , Oncogene Proteins/genetics , Oxidative Stress , Peroxiredoxins/genetics , Protein Deglycase DJ-1 , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Sequence Analysis, RNA
18.
Biosci Biotechnol Biochem ; 78(4): 593-601, 2014.
Article in English | MEDLINE | ID: mdl-25036955

ABSTRACT

Euglena gracilis lacks catalase and contains ascorbate peroxidase (APX) which is localized exclusively in the cytosol. Other enzymes that scavenge reactive oxygen species (ROS) in Euglena have not yet been identified; therefore, ROS metabolism, especially in organelles, remains unclear in Euglena. The full-length cDNAs of four Euglena peroxiredoxins (EgPrxs) were isolated in this study. EgPrx1 and -4 were predicted to be localized in the cytosol, and EgPrx2 and -3 in plastids and mitochondria, respectively. The catalytic efficiencies of recombinant EgPrxs were similar to those of plant thiol-peroxidases, but were markedly lower than those of APX from Euglena. However, transcript levels of EgPrx1, -2, and -3 were markedly higher than those of APX. The growth rate of Euglena cells, in which the expression of EgPrx1 and -4 was suppressed by gene silencing, was markedly reduced under normal conditions, indicating physiological significance of Prx proteins.


Subject(s)
Euglena gracilis/enzymology , Peroxiredoxins/metabolism , Amino Acid Sequence , Cell Proliferation , Euglena gracilis/cytology , Euglena gracilis/genetics , Gene Knockdown Techniques , Hydrogen Peroxide/metabolism , Isoenzymes/chemistry , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Sequence Data , Peroxiredoxins/chemistry , Peroxiredoxins/deficiency , Peroxiredoxins/genetics
19.
PLoS One ; 9(6): e98764, 2014.
Article in English | MEDLINE | ID: mdl-24904997

ABSTRACT

In order to understand the molecular mechanisms of Bifidobacterium infantis thymidine kinase/nucleoside analogue ganciclovir (BI-TK/GCV) treatment system which was proven to exhibit sustainable anti-tumor growth activity and induce apoptosis in bladder cancer, a proteomic approach of isobaric tags for relative and absolute quantification (iTRAQ), followed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used. 192 down-regulated and 210 up-regulated proteins were identified after treatment with BI-TK/GCV system in Sprague-Dawley (SD) rats. Western blot analysis and immunohistochemistry analysis confirmed that Peroxiredoxin-I (Prx-I) was significantly down-regulated in bladder cancer after treatment. Prx-I silencing by transfection of Prx-I shRNA significantly suppressed growth, promoted apoptosis and regulated the cell cycle in T24 cells and reduced the phospho-NF-κB p50 and p65 protein expression which revealed the links between Prx-I and NF-κB pathway implied by Ingenuity pathway analysis (IPA). These findings yield new insights into the therapy of bladder cancer, revealing Prx-I as a new therapeutic target and indicating BI-TK/GCV system as a prospective therapy by down-regulation of Prx-I through NF-κB signaling pathway.


Subject(s)
Bifidobacterium/enzymology , Ganciclovir/pharmacology , Peroxiredoxins/metabolism , Proteomics , Thymidine Kinase/genetics , Urinary Bladder Neoplasms/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Bifidobacterium/genetics , Bifidobacterium/physiology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Ganciclovir/therapeutic use , Gene Knockdown Techniques , Genetic Therapy , NF-kappa B/metabolism , Peroxiredoxins/deficiency , Peroxiredoxins/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects , Transcriptome/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
20.
PLoS One ; 9(4): e94826, 2014.
Article in English | MEDLINE | ID: mdl-24733019

ABSTRACT

Parkinson's disease (PD) is one of the most prevalent neurodegenerative brain diseases; it is accompanied by extensive loss of dopamine (DA) neurons of the substantia nigra that project to the putamen, leading to impaired motor functions. Several genes have been associated with hereditary forms of the disease and transgenic mice have been developed by a number of groups to produce animal models of PD and to explore the basic functions of these genes. Surprisingly, most of the various mouse lines generated such as Parkin KO, Pink1 KO, DJ-1 KO and LRRK2 transgenic have been reported to lack degeneration of nigral DA neuron, one of the hallmarks of PD. However, modest impairments of motor behavior have been reported, suggesting the possibility that the models recapitulate at least some of the early stages of PD, including early dysfunction of DA axon terminals. To further evaluate this possibility, here we provide for the first time a systematic comparison of DA release in four different mouse lines, examined at a young age range, prior to potential age-dependent compensations. Using fast scan cyclic voltammetry in striatal sections prepared from young, 6-8 weeks old mice, we examined sub-second DA overflow evoked by single pulses and action potential trains. Unexpectedly, none of the models displayed any dysfunction of DA overflow or reuptake. These results, compatible with the lack of DA neuron loss in these models, suggest that molecular dysfunctions caused by the absence or mutation of these individual genes are not sufficient to perturb the function and survival of mouse DA neurons.


Subject(s)
Dopamine/metabolism , Mutation/genetics , Oncogene Proteins/deficiency , Peroxiredoxins/deficiency , Protein Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/deficiency , Animals , Electrochemical Techniques , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Mice, Knockout , Neostriatum , Oncogene Proteins/metabolism , Peroxiredoxins/metabolism , Protein Deglycase DJ-1 , Protein Kinases/metabolism , Synaptic Transmission , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...