Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
CNS Neurosci Ther ; 30(3): e14681, 2024 03.
Article in English | MEDLINE | ID: mdl-38516845

ABSTRACT

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


Subject(s)
Brain Injuries , Sulfonamides , Toll-Like Receptor 4 , Animals , Mice , Brain Injuries/etiology , Cerebral Hemorrhage/metabolism , Inflammation/etiology , Inflammation/pathology , Mice, Inbred C57BL , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Peroxiredoxins/therapeutic use , Toll-Like Receptor 4/metabolism
2.
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
3.
Bull Exp Biol Med ; 160(5): 639-42, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27021100

ABSTRACT

Antioxidant properties of recombinant peroxiredoxin-6 and chimeric protein PSH combining peroxidase and superoxide dismutase activities were studied on the model of retrograde perfusion of isolated rat heart under conditions of H2O2-induced oxidative stress. The exogenous antioxidant proteins exhibited cardioprotective properties manifested in heart rate normalization, maintenance of contractile activity of the myocardium, and prevention of H2O2-induced LPO in oxidative stress. Localization of peroxiredoxin-6 and PSH in the cardiac tissue was determined and myocardial structures most effectively protected by the antioxidant enzymes from ischemia/reperfusion-induced damages were identified. The results suggest that modified peroxiredoxins are promising components of perfusion media for preservation of isolated organs.


Subject(s)
Antioxidants/therapeutic use , Cardiotonic Agents/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Oxidative Stress/drug effects , Peroxiredoxins/therapeutic use , Animals , Heart/physiopathology , Heart Rate/drug effects , Hydrogen Peroxide/adverse effects , Male , Myocardial Contraction/drug effects , Perfusion , Peroxidase/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
4.
Bull Exp Biol Med ; 160(3): 322-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26742734

ABSTRACT

Nephroprotective effect of exogenous chimeric antioxidant enzyme with combined superoxide dismutase and peroxide activities (PSH protein) was studied on the model of ischemia/reperfusion damage of the renal tissue. It was shown that post-ischemic (25- and 45-min ischemia) intravenous administration of PSH protein significantly normalized the levels of creatinine and urea. Histological studies showed that as distinct from ischemic kidney, the structure of renal corpuscles and tubules remained unchanged, the number of atrophied glomeruli and glomeruli with exudates and protein inclusions decreased in the capsular teeth after postischemic intravenous administration of PSH protein. Immunohistochemical investigations showed that post-ischemic intravenous injection of PSH protein significantly reduced the intensity of apoptosis in ischemic renal tissues.


Subject(s)
Antioxidants/therapeutic use , Kidney/drug effects , Animals , Kidney/metabolism , Male , Peroxiredoxins/therapeutic use , Rats , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Superoxide Dismutase/therapeutic use
5.
J Urol ; 195(3): 706-12, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26585682

ABSTRACT

PURPOSE: We compared levels of peroxiredoxin 2 in semen samples from normozoospermic and asthenozoospermic men. The potential effects of TAT-peroxiredoxin 2 fusion protein on sperm motility and DNA integrity were also evaluated. MATERIALS AND METHODS: Semen samples were obtained from 50 normozoospermic and 50 asthenozoospermic men. Lipid peroxidation of semen was determined using a commercial malondialdehyde kit. Sperm DNA fragmentation was evaluated by TUNEL assay. Western blot and immunofluorescence were performed to detect the amount of peroxiredoxin 2 protein in seminal plasma and spermatozoa. Sperm motility, DNA damage and levels of reactive oxygen species were evaluated after TAT-peroxiredoxin 2 fusion protein supplementation to the sperm suspension for 2 and 12 hours of incubation. RESULTS: In asthenozoospermic semen samples a significantly higher level of malondialdehyde and DNA damage was discovered. However, the expression of peroxiredoxin 2 was significantly lower in seminal plasma and spermatozoa compared with that of normozoospermic men. TAT-peroxiredoxin 2 fusion protein was successfully prepared and delivered to the spermatozoa. Interestingly adding TAT-peroxiredoxin 2 in asthenozoospermic sperm suspension effectively defended against the decrease in progressive motility and the increase in DNA damage. CONCLUSIONS: This study shows that supplementation of TAT-peroxiredoxin 2 fusion protein in the sperm suspension from asthenozoospermic men effectively improved sperm motility and DNA integrity by reducing levels of reactive oxygen species. Therefore, we speculate that peroxiredoxin 2 may have an important role as an antioxidant defense in semen and would provide new prevention and therapy alternatives for asthenozoospermia.


Subject(s)
Asthenozoospermia/drug therapy , Asthenozoospermia/genetics , DNA Damage/drug effects , Gene Products, tat/therapeutic use , Peroxiredoxins/analysis , Peroxiredoxins/therapeutic use , Semen/chemistry , Sperm Motility/drug effects , Spermatozoa/chemistry , Spermatozoa/drug effects , Adult , Asthenozoospermia/metabolism , Asthenozoospermia/physiopathology , Gene Products, tat/pharmacology , Humans , Male , Peroxiredoxins/pharmacology , Reactive Oxygen Species , Spermatozoa/physiology
6.
Vet Parasitol ; 195(3-4): 272-85, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23623183

ABSTRACT

The liver fluke, Fasciola hepatica, causes fascioliasis in domestic animals (sheep, cattle), a global disease that is also an important infection of humans. As soon as the parasite invades the gut wall its interaction with various host immune cells (e.g. dendritic cells, macrophages and mast cells) is complex. The parasite secretes a myriad of molecules that direct the immune response towards a favourable non-protective Th2-mediate/regulatory environment. These immunomodulatory molecules, such as cathepsin L peptidase (FhCL1), are under development as the first generation of fluke vaccines. However, this peptidase and other molecules, such as peroxiredoxin (FhPrx) and helminth defence molecule (FhHDM-1), exhibit various immunomodulatory properties that could be harnessed to help treat immune-related conditions in humans and animals.


Subject(s)
Antibodies, Helminth/immunology , Cattle Diseases/prevention & control , Fasciola hepatica/immunology , Fascioliasis/veterinary , Helminth Proteins/immunology , Sheep Diseases/prevention & control , Animals , Autoimmune Diseases/drug therapy , Cathepsin L/immunology , Cathepsin L/metabolism , Cathepsin L/therapeutic use , Cattle , Cattle Diseases/immunology , Cattle Diseases/parasitology , Fasciola hepatica/enzymology , Fascioliasis/immunology , Fascioliasis/parasitology , Fascioliasis/prevention & control , Helminth Proteins/metabolism , Helminth Proteins/therapeutic use , Humans , Immunomodulation , Peroxiredoxins/immunology , Peroxiredoxins/metabolism , Peroxiredoxins/therapeutic use , Sheep , Sheep Diseases/immunology , Sheep Diseases/parasitology , Vaccines
7.
J UOEH ; 34(1): 27-39, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22428456

ABSTRACT

We review the critical roles of peroxiredoxin (PRDX) 4 in inflammatory diseases. The PRDX family, a new family of proteins with an antioxidative function, is ubiquitously synthesized and abundantly identified in various organisms. The function of these enzymes, which include at least six distinct PRDX genes expressed in mammals, still remains unclear. Especially, in contrast to the intracellular localization of other family members, PRDX4 is the only known secretory form located in the extracellular space and exerts its protective function against oxidative damage by scavenging reactive oxygen species in the vascular vessels. To date, however, it is not clear whether or how PRDX4 expression affects various diseases in vivo. More recently, we generated human PRDX4 (hPRDX4) transgenic (Tg) mice, and, for the first time, established a type 1 diabetes mellitus model induced by a single high dose of streptozotocin on Tg mice. Our published data demonstrate that streptozotocin-treated Tg mice, which overexpress hPRDX4 in pancreatic islets, can protect pancreatic beta-cells against streptozotocin-induced injury (insulitis) by suppressing increased oxidative stress and inflammatory signaling activation. These observations indicate that Tg mice could become a useful animal model to study the relevance of oxidative stress to inflammation, and that a specific accelerator of PRDX4 might prove to be a potential therapeutic agent for ameliorating various chronic inflammatory diseases.


Subject(s)
Inflammation/physiopathology , Oxidative Stress , Peroxiredoxins/physiology , Animals , Diabetes Mellitus, Experimental/physiopathology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Peroxiredoxins/therapeutic use
8.
J Neurosci ; 31(1): 247-61, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21209210

ABSTRACT

The peroxiredoxin (PRX) family of antioxidant enzymes helps maintain the intracellular reducing milieu and suppresses apoptosis in non-neuronal cells. However, whether PRX can inhibit neuronal apoptosis through specific signaling mechanisms remains poorly understood. Induction of PRX2, the most abundant neuronal PRX, occurs in Parkinson's disease (PD) patient brains, but its functional impact is unclear. In the present study, we used the dopaminergic (DA) toxin 6-hydroxydopamine (6-OHDA) to model PD and explore the protective effect and mechanisms of PRX on DA neurons. Of the 2-cysteine PRXs that were tested in MN9D DA neurons, endogenous PRX2 was most beneficial to cell survival. Lentivirus-mediated PRX2 overexpression conferred marked in vitro and in vivo neuroprotection against 6-OHDA toxicity in DA neurons, and preserved motor functions involving the dopamine system in mouse. In addition to its role as an antioxidant enzyme, PRX2 exhibited anti-apoptotic effects in DA neurons via suppression of apoptosis signal-regulating kinase (ASK1)-dependent activation of the c-Jun N-terminal kinase/c-Jun and p38 pro-death pathways, which are also activated in DA neurons of postmortem PD brains. PRX2 inhibited 6-OHDA-induced ASK1 activation by modulating the redox status of the endogenous ASK1 inhibitor thioredoxin (Trx). PRX2 overexpression maintained Trx in a reduced state by inhibiting the cysteine thiol-disulfide exchange, thereby preventing its dissociation from ASK1. This study describes a previously undefined mechanism by which redox-sensitive molecules signal via apoptotic pathways in response to PD-relevant toxic stress in DA neurons. Our results also suggest that PRX2 and ASK1 may be potential targets for neuroprotective intervention in PD.


Subject(s)
Dopamine/metabolism , MAP Kinase Kinase Kinase 5/drug effects , Neurodegenerative Diseases/prevention & control , Neuroprostanes/therapeutic use , Peroxiredoxins/therapeutic use , Signal Transduction/drug effects , Aged , Aged, 80 and over , Analysis of Variance , Animals , Apomorphine , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Humans , Indoles , Male , Mice , Mice, Inbred C57BL , Middle Aged , Motor Activity/drug effects , Neurodegenerative Diseases/chemically induced , Oxidopamine/adverse effects , Parkinson Disease/metabolism , Parkinson Disease/pathology , Postmortem Changes , RNA, Small Interfering/metabolism , Thioredoxins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...