Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biochimie ; 156: 158-168, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30352250

ABSTRACT

Parkinson's disease (PD), a neurodegenerative disorder, is characterized by a loss of dopaminergic neurons in the substantia nigra (SN) of the brain and it is well known that the pathogenesis of PD is related to a number of risk factors including oxidative stress. Antioxidant 1 (ATOX1) protein plays a crucial role in various diseases as an antioxidant and chaperone. In this study, we determined whether Tat-ATOX1 could protect against 1-methyl-4-phenylpyridinium ion (MPP+)-induced SH-SY5Y cell death and in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced animal model of PD. In the MPP+ exposed SH-SY5Y cells, Tat-ATOX1 markedly inhibited cell death and toxicities. In addition, Tat-ATOX1 markedly suppressed the activation of Akt and mitogen activated protein kinases (MAPKs) as well as cleavage of caspase-3 and Bax expression levels. In a MPTP-induced animal model, Tat-ATOX1 transduced into brain and protected dopaminergic neuronal cell loss. Taken together, Tat-ATOX1 inhibits dopaminergic neuronal death through the suppression of MAPKs and apoptotic signal pathways. Thus, Tat-ATOX1 represents a potential therapeutic protein drug candidate for PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Cation Transport Proteins , MPTP Poisoning/prevention & control , Metallochaperones , Molecular Chaperones , Recombinant Fusion Proteins , Animals , Cation Transport Proteins/biosynthesis , Cation Transport Proteins/genetics , Cell Death/drug effects , Cell Line, Tumor , Copper Transport Proteins , Humans , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Male , Metallochaperones/biosynthesis , Metallochaperones/genetics , Mice , Molecular Chaperones/biosynthesis , Molecular Chaperones/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transduction, Genetic
2.
J Neurosurg Spine ; 29(5): 599-607, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30141762

ABSTRACT

The authors investigated the effects of a silk solution against laminectomy-induced dural adhesion formation and inflammation in a rat model. They found that it significantly reduced postlaminectomy dural adhesion formation and inflammation. Dural adhesion formation, thought to be an inevitable consequence of laminectomy, is one of the most common complications following spinal surgery, and the authors' results indicate that the silk solution might be a potential novel therapeutic agent for dural adhesion formation.


Subject(s)
Inflammation/complications , Laminectomy/adverse effects , Silk/adverse effects , Animals , Disease Models, Animal , Interleukin-1beta/metabolism , Nitric Oxide/metabolism , Rats
3.
Immunobiology ; 223(11): 709-717, 2018 11.
Article in English | MEDLINE | ID: mdl-30049418

ABSTRACT

Phosphoprotein enriched in astrocytes 15 (PEA15) plays a multi-functional role in neuronal cell survival, however the effects of PEA15 against inflammation have not been investigated yet. To examine the effects of PEP-1-PEA15 protein against lipopolysaccharide (LPS)-induced inflammatory responses in Raw 264.7 cells and in a 12-O-tetradecanoylphobol 13-acetate (TPA)-induced mouse model, we constructed and purified PEP-1-PEA15 protein, which can transduce into cells or tissues. PEP-1-PEA15 inhibited LPS-induced damage in cells including that caused by reactive oxygen species (ROS) production and DNA fragmentation. PEP-1-PEA15 also significantly suppressed activation of mitogen activated protein kinases (MAPKs), pro-inflammatory mediator proteins and various cytokines. In a TPA-induced mouse ear edema model, PEP-1-PEA15 significantly reduced ear weight and thickness as well as MAPK activation as well as the expression levels of COX-2, iNOS, IL-6, IL-1ß, and TNF-α. These results demonstrated that PEP-1-PEA15 showed anti-inflammatory effect in cells and animal model suggesting that this fusion protein protects cells or skin tissues from inflammatory response.


Subject(s)
Cysteamine/analogs & derivatives , Edema/immunology , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Peptides/metabolism , Phosphoproteins/metabolism , Animals , Apoptosis Regulatory Proteins , Cysteamine/metabolism , Cytokines/metabolism , DNA Fragmentation , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred ICR , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/immunology
4.
BMB Rep ; 51(7): 362-367, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29936932

ABSTRACT

A major feature of type 1 diabetes mellitus (T1DM) is hyperglycemia and dysfunction of pancreatic ß-cells. In a previous study, we have shown that Tat-DJ-1 protein inhibits pancreatic RINm5F ß-cell death caused by oxidative stress. In this study, we examined effects of Tat-DJ-1 protein on streptozotocin (STZ)-induced diabetic mice. Wild type (WT) Tat-DJ-1 protein transduced into pancreas where it markedly inhibited pancreatic ß-cell destruction and regulated levels of serum parameters including insulin, alkaline phosphatase (ALP), and free fatty acid (FFA) secretion. In addition, transduced WT Tat-DJ-1 protein significantly inhibited the activation of NF-κB and MAPK (ERK and p38) expression as well as expression of COX-2 and iNOS in STZ exposed pancreas. In contrast, treatment with C106A mutant Tat-DJ-1 protein showed no protective effects. Collectively, our results indicate that WT Tat-DJ-1 protein can significantly ameliorate pancreatic tissues in STZ-induced diabetes in mice. [BMB Reports 2018; 51(7): 362-367].


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Gene Products, tat/genetics , Protective Agents/therapeutic use , Protein Deglycase DJ-1/genetics , Recombinant Fusion Proteins/therapeutic use , Alkaline Phosphatase/blood , Animals , Cyclooxygenase 2/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fatty Acids, Nonesterified/blood , Insulin/blood , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Pancreas/metabolism , Protective Agents/pharmacology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Mol Med Rep ; 18(2): 2216-2228, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29916538

ABSTRACT

Oxidative stress is known to be a primary risk factor for neuronal diseases. Glutaredoxin (GLRX)­1, a redox­regulator of the thioredoxin superfamily, is known to exhibit an important role in cell survival via various cellular functions. However, the precise roles of GLRX1 in brain ischemia are still not fully understood. The present study investigated whether transduced PEP­1­GLRX1 protein has protective effects against oxidative stress in cells and in an animal model. Transduced PEP­1­GLRX1 protein increased HT­22 cell viability under oxidative stress and this fusion protein significantly reduced intracellular reactive oxygen species and levels of DNA damage. In addition, PEP­1­GLRX1 protein regulated RAC­a serine/threonine­protein kinase and mitogen­activated protein kinase signaling, in addition to apoptotic signaling including B cell lymphoma (Bcl)­2, Bcl­2 associated X, apoptosis regulator, pro­caspase­9 and p53 expression levels. In an ischemic animal model, it was verified that PEP­1­GLRX1 transduced into the Cornu Ammonis 1 region of the animal brain, where it markedly protected against ischemic injury. These results indicate that PEP­1­GLRX1 attenuates neuronal cell death resulting from oxidative stress in vitro and in vivo. Therefore, PEP­1­GLRX1 may exhibit a beneficial role in the treatment of neuronal disorders, including ischemic injury.


Subject(s)
Cysteamine/analogs & derivatives , Glutaredoxins/pharmacology , Hippocampus/metabolism , MAP Kinase Signaling System/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Peptides/pharmacology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Line , Cysteamine/pharmacology , Hippocampus/pathology , Mice , Neurons/pathology
6.
BMB Rep ; 50(9): 460-465, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28760196

ABSTRACT

Polycystic kidney disease (PKD) is one of the most common inherited disorders, involving progressive cyst formation in the kidney that leads to renal failure. FK506 binding protein 12 (FK506BP) is an immunophilin protein that performs multiple functions, including regulation of cell signaling pathways and survival. In this study, we determined the roles of PEP-1-FK506BP on cell proliferation and cyst formation in PKD cells. Purified PEP-1-FK506BP transduced into PKD cells markedly inhibited cell proliferation. Also, PEP-1-FK506BP drastically inhibited the expression levels of p-Akt, p-p70S6K, p-mTOR, and p-ERK in PKD cells. In a 3D-culture system, PEP-1-FK506BP significantly reduced cyst formation. Furthermore, the combined effects of rapamycin and PEP-1-FK506BP on cyst formation were markedly higher than the effects of individual treatments. These results suggest that PEP-1-FK506BP delayed cyst formation and could be a new therapeutic strategy for renal cyst formation in PKD. [BMB Reports 2017; 50(9): 460-465].


Subject(s)
Polycystic Kidney Diseases/metabolism , TOR Serine-Threonine Kinases/metabolism , Tacrolimus Binding Protein 1A/metabolism , Animals , Blotting, Western , Cell Proliferation/genetics , Cell Proliferation/physiology , Cysts/genetics , Cysts/metabolism , Disease Models, Animal , Humans , Microscopy, Confocal , Polycystic Kidney Diseases/genetics , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases/genetics , Tacrolimus Binding Protein 1A/genetics
7.
Biotechnol Lett ; 39(4): 511-521, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28074428

ABSTRACT

OBJECTIVES: To identify the protective effect of DJ-1 protein against oxidative stress-induced HepG2 cell death, we used cell-permeable wild type (WT) and a mutant (C106A Tat-DJ-1) protein. RESULTS: By using western blotting and fluorescence microscopy, we observed WT and C106A Tat-DJ-1 proteins were efficiently transduced into HepG2 cells. Transduced WT Tat-DJ-1 proteins increased cell survival and protected against DNA fragmentation and intracellular ROS generation levels in H2O2-exposed HepG2 cells. At the same time, transduced WT Tat-DJ-1 protein significantly inhibited NF-κB and MAPK (JNK and p38) activation as well as regulated the Bcl-2 and Bax expression levels. However, C106A Tat-DJ-1 protein did not show any protective effect against cell death responses in H2O2-exposed HepG2 cells. CONCLUSIONS: Oxidative stress-induced HepG2 cell death was significantly reduced by transduced WT Tat-DJ-1 protein, not by C106A Tat-DJ-1 protein. Thus, transduction of WT Tat-DJ-1 protein could be a novel strategy for promoting cell survival in situations of oxidative stress-induced HepG2 cell death.


Subject(s)
Cell Survival , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxidative Stress , Recombinant Fusion Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis , DNA Fragmentation , Hep G2 Cells , Humans , Hydrogen Peroxide , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/genetics , Transduction, Genetic , bcl-2-Associated X Protein/metabolism
8.
Mol Brain ; 10(1): 1, 2017 01 04.
Article in English | MEDLINE | ID: mdl-28052764

ABSTRACT

Oxidative stress plays an important role in the progression of various neuronal diseases including ischemia. Heat shock protein 22 (HSP22) is known to protect cells against oxidative stress. However, the protective effects and mechanisms of HSP22 in hippocampal neuronal cells under oxidative stress remain unknown. In this study, we determined whether HSP22 protects against hydrogen peroxide (H2O2)-induced oxidative stress in HT-22 using Tat-HSP22 fusion protein. We found that Tat-HSP22 transduced into HT-22 cells and that H2O2-induced cell death, oxidative stress, and DNA damage were significantly reduced by Tat-HSP22. In addition, Tat-HSP22 markedly inhibited H2O2-induced mitochondrial membrane potential, cytochrome c release, cleaved caspase-3, and Bax expression levels, while Bcl-2 expression levels were increased in HT-22 cells. Further, we showed that Tat-HSP22 transduced into animal brain and inhibited cleaved-caspase-3 expression levels as well as significantly inhibited hippocampal neuronal cell death in the CA1 region of animals in the ischemic animal model. In the present study, we demonstrated that transduced Tat-HSP22 attenuates oxidative stress-induced hippocampal neuronal cell death through the mitochondrial signaling pathway and plays a crucial role in inhibiting neuronal cell death, suggesting that Tat-HSP22 protein may be used to prevent oxidative stress-related brain diseases including ischemia.


Subject(s)
Gene Products, tat/pharmacology , Heat-Shock Proteins/pharmacology , Hippocampus/pathology , Mitochondria/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/pharmacology , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , Animals , Cell Death/drug effects , Cell Membrane Permeability/drug effects , Gerbillinae , Hydrogen Peroxide/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Molecular Chaperones , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Recombinant Fusion Proteins/isolation & purification , Transduction, Genetic
9.
Med Chem Res ; 25(11): 2589-2598, 2016.
Article in English | MEDLINE | ID: mdl-27818604

ABSTRACT

Oxidative stress is highly involved in the development of diabetes mellitus by destruction of pancreatic ß-cells. DJ-1 is an antioxidant protein and DJ-1 expression levels are known to be reduced in diabetes mellitus. Thus, we examined the effects of DJ-1 protein against oxidative stress-induced pancreatic ß-cell (RINm5F) death using cell permeable wild-type and mutant-type (C106A) Tat-DJ-1 proteins, which both efficiently transduced into RINm5F cells. Intracellular stability of wild-type Tat-DJ-1 persisted two times longer than C106A Tat-DJ-1. Wild-type Tat-DJ-1 protein markedly protected cells from hydrogen peroxide-induced toxicities such as cell death, reactive oxygen species generation, and DNA fragmentation. Further, wild-type Tat-DJ-1 protein significantly inhibited hydrogen peroxide-induced activation of mitogen-activated protein kinases and NF-κB signaling. On the other hand, C106A Tat-DJ-1 protein did not show the same protective effects. These results indicate that wild-type Tat-DJ-1 inhibits oxidative stress-induced cellular toxicity and activation of mitogen-activated protein kinases and NF-κB signals in RINm5F cells. These results suggest that wild-type Tat-DJ-1 protein may be a potential therapeutic agent against diabetes mellitus or toward the prevention of pancreatic ß-cell destruction.

10.
BMB Rep ; 49(11): 617-622, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27616357

ABSTRACT

Oxidative stress is closely associated with various diseases and is considered to be a major factor in ischemia. NAD(P)H:quinone oxidoreductase 1 (NQO1) protein is a known antioxidant protein that plays a protective role in various cells against oxidative stress. We therefore investigated the effects of cell permeable Tat-NQO1 protein on hippocampal HT-22 cells, and in an animal ischemia model. The Tat-NQO1 protein transduced into HT-22 cells, and significantly inhibited against hydrogen peroxide (H2O2)-induced cell death and cellular toxicities. Tat-NQO1 protein inhibited the Akt and mitogen activated protein kinases (MAPK) activation as well as caspase-3 expression levels, in H2O2 exposed HT-22 cells. Moreover, Tat-NQO1 protein transduced into the CA1 region of the hippocampus of the animal brain and drastically protected against ischemic injury. Our results indicate that Tat-NQO1 protein exerts protection against neuronal cell death induced by oxidative stress, suggesting that Tat-NQO1 protein may potentially provide a therapeutic agent for neuronal diseases. [BMB Reports 2016; 49(11): 617-622].


Subject(s)
Gene Products, tat/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , Oxidative Stress , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line , Disease Models, Animal , Gene Products, tat/metabolism , Gerbillinae , Hippocampus/cytology , Hippocampus/metabolism , Humans , Hydrogen Peroxide/toxicity , Ischemia/metabolism , Ischemia/pathology , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress/drug effects , Plasmids/genetics , Plasmids/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology
11.
Free Radic Biol Med ; 97: 250-262, 2016 08.
Article in English | MEDLINE | ID: mdl-27317854

ABSTRACT

Proline rich Akt substrate (PRAS40) is a component of mammalian target of rapamycin complex 1 (mTORC1) and is known to play an important role against reactive oxygen species-induced cell death. However, the precise function of PRAS40 in ischemia remains unclear. Thus, we investigated whether Tat-PRAS40, a cell-permeable fusion protein, has a protective function against oxidative stress-induced hippocampal neuronal (HT-22) cell death in an animal model of ischemia. We showed that Tat-PRAS40 transduced into HT-22 cells, and significantly protected against cell death by reducing the levels of H2O2 and derived reactive species, and DNA fragmentation as well as via the regulation of Bcl-2, Bax, and caspase 3 expression levels in H2O2 treated cells. Also, we showed that transduced Tat-PARS40 protein markedly increased phosphorylated RRAS40 expression levels and 14-3-3σ complex via the Akt signaling pathway. In an animal ischemia model, Tat-PRAS40 effectively transduced into the hippocampus in animal brain and significantly protected against neuronal cell death in the CA1 region. We showed that Tat-PRAS40 protein effectively transduced into hippocampal neuronal cells and markedly protected against neuronal cell damage. Therefore, we suggest that Tat-PRAS40 protein may be used as a therapeutic protein for ischemia and oxidative stress-induced brain disorders.


Subject(s)
Apoptosis/drug effects , Brain Ischemia/metabolism , Oxidative Stress , Phosphoproteins/pharmacology , Recombinant Fusion Proteins/pharmacology , 14-3-3 Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Brain Ischemia/drug therapy , CA1 Region, Hippocampal/pathology , Cell Line , DNA Fragmentation , Drug Evaluation, Preclinical , Gerbillinae , Male , Protein Processing, Post-Translational
12.
BMB Rep ; 49(7): 382-7, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27049109

ABSTRACT

Reactive oxygen species generated under oxidative stress are involved in neuronal diseases, including ischemia. Glutathione S-transferase pi (GSTpi) is a member of the GST family and is known to play important roles in cell survival. We investigated the effect of GSTpi against oxidative stress-induced hippocampal HT-22 cell death, and its effects in an animal model of ischemic injury, using a cell-permeable PEP-1-GSTpi protein. PEP-1-GSTpi was transduced into HT-22 cells and significantly protected against H2O2-treated cell death by reducing the intracellular toxicity and regulating the signal pathways, including MAPK, Akt, Bax, and Bcl-2. PEP-1-GSTpi transduced into the hippocampus in animal brains, and markedly protected against neuronal cell death in an ischemic injury animal model. These results indicate that PEP-1-GSTpi acts as a regulator or an antioxidant to protect against oxidative stressinduced cell death. Our study suggests that PEP-1-GSTpi may have potential as a therapeutic agent for the treatment of ischemia and a variety of oxidative stress-related neuronal diseases. [BMB Reports 2016; 49(7): 382-387].


Subject(s)
Glutathione S-Transferase pi/metabolism , Hippocampus/metabolism , Hydrogen Peroxide/toxicity , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line , Cell Survival/drug effects , Cysteamine/analogs & derivatives , Cysteamine/metabolism , Glutathione S-Transferase pi/genetics , Neuroprotective Agents/pharmacology , Peptides/genetics , Peptides/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism
13.
BMB Rep ; 49(5): 297-302, 2016 May.
Article in English | MEDLINE | ID: mdl-26996344

ABSTRACT

Loss of pancreatic ß-cells by oxidative stress or cytokines is associated with diabetes mellitus (DM). DJ-1 is known to as a multifunctional protein, which plays an important role in cell survival. We prepared cell permeable wild type (WT) and mutant type (M26I) Tat-DJ-1 proteins to investigate the effects of DJ-1 against combined cytokines (IL-1ß, IFN-γ and TNF-α)-induced RINm5F cell death. Both Tat-DJ-1 proteins were transduced into RINm5F cells. WT Tat-DJ-1 proteins significantly protected against cell death from cytokines by reducing intracellular toxicities. Also, WT Tat-DJ-1 proteins markedly regulated cytokines-induced pro- and anti-apoptosis proteins. However, M26I Tat-DJ-1 protein showed relatively low protective effects, as compared to WT Tat-DJ-1 protein. Our experiments demonstrated that WT Tat-DJ-1 protein protects against cytokine-induced RINm5F cell death by suppressing intracellular toxicities and regulating apoptosisrelated protein expression. Thus, WT Tat-DJ-1 protein could potentially serve as a therapeutic agent for DM and cytokine related diseases. [BMB Reports 2016; 49(5): 297-302].


Subject(s)
Cytokines/pharmacology , Pancreas/pathology , Protein Deglycase DJ-1/metabolism , Recombinant Fusion Proteins/metabolism , Transduction, Genetic , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Cell Death/drug effects , Cell Line , Humans , Rats , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects
14.
J Neurol Sci ; 358(1-2): 226-35, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26365288

ABSTRACT

Oxidative stress is considered a major factor in various neuronal diseases including ischemia-reperfusion injury. Proviral Integration Moloney 2 (PIM2) proteins, one of the families of PIM kinases, play crucial roles in cell survival. However, the functions of PIM2 protein against ischemia are not understood. Therefore, the protective effects of PIM2 against oxidative stress-induced hippocampal HT22 cell death and brain ischemic injury were evaluated using Tat-PIM2, a cell permeable fusion protein. Tat-PIM2 protein transduced into hippocampal HT22 cells. Low doses of transduced Tat-PIM2 protein protected against oxidative stress-induced cell death including DNA damage and markedly inhibited the activation of mitogen activated protein kinase (MAPKs), NF-κB and the expression levels of Bax protein. Furthermore, Tat-PIM2 protein transduced into the CA1 region of the hippocampus and significantly prevented neuronal cell death in an ischemic insult animal model. These results indicated that low doses of Tat-PIM2 protein protects against oxidative stress-induced neuronal cell death, suggesting low doses of Tat-PIM2 protein provides a potential therapeutic agent against oxidative stress-induced neuronal diseases including ischemia.


Subject(s)
Cell Survival/drug effects , Gene Products, tat/administration & dosage , Hippocampus/drug effects , Neurons/drug effects , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/administration & dosage , Proto-Oncogene Proteins/administration & dosage , Animals , Cell Line , Gerbillinae , Hippocampus/metabolism , Mice , Neurons/metabolism , Reactive Oxygen Species/metabolism , Transduction, Genetic
15.
Biomaterials ; 64: 45-56, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26117230

ABSTRACT

Parkinson's disease (PD) is an oxidative stress-mediated neurodegenerative disorder caused by selective dopaminergic neuronal death in the midbrain substantia nigra. Paraoxonase 1 (PON1) is a potent inhibitor of low-density lipoprotein (LDL) and high-density lipoprotein (HDL) against oxidation by destroying biologically active phospholipids with potential protective effects against oxidative stress-induced inflammatory disorders. In a previous study, we constructed protein transduction domain (PTD) fusion PEP-1-PON1 protein to transduce PON1 into cells and tissue. In this study, we examined the role of transduced PEP-1-PON1 protein in repressing oxidative stress-mediated inflammatory response in microglial BV2 cells after exposure to lipopolysaccharide (LPS). Moreover, we identified the functions of transduced PEP-1-PON1 proteins which include, mitigating mitochondrial damage, decreasing reactive oxidative species (ROS) production, matrix metalloproteinase-9 (MMP-9) expression and protecting against 1-methyl-4-phenylpyridinium (MPP(+))-induced neurotoxicity in SH-SY5Y cells. Furthermore, transduced PEP-1-PON1 protein reduced MMP-9 expression and protected against dopaminergic neuronal cell death in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice model. Taken together, these results suggest a promising therapeutic application of PEP-1-PON1 proteins against PD and other inflammation and oxidative stress-related neuronal diseases.


Subject(s)
Aryldialkylphosphatase/therapeutic use , Cell-Penetrating Peptides/therapeutic use , Dopaminergic Neurons/drug effects , Genetic Therapy , Microglia/drug effects , Parkinsonian Disorders/therapy , Recombinant Fusion Proteins/therapeutic use , Animals , Apoptosis/drug effects , Aryldialkylphosphatase/administration & dosage , Brain/pathology , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , Cells, Cultured , Dopaminergic Neurons/pathology , Enzyme Induction/drug effects , Genetic Vectors/therapeutic use , Humans , Lipopolysaccharides/toxicity , Matrix Metalloproteinase 9/biosynthesis , Membrane Potential, Mitochondrial/drug effects , Mice , Microglia/physiology , Neuroblastoma/pathology , Oxidative Stress , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , Protein Structure, Tertiary , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Transduction, Genetic
16.
BMB Rep ; 48(11): 618-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25817214

ABSTRACT

FK506 binding protein 12 (FK506BP) is a small peptide with a single FK506BP domain that is involved in suppression of immune response and reactive oxygen species. FK506BP has emerged as a potential drug target for several inflammatory diseases. Here, we examined the protective effects of directly applied cell permeable FK506BP (PEP-1-FK506BP) on corneal alkali burn injury (CAI). In the cornea, there was a significant decrease in the number of cells expressing pro-inflammation, apoptotic, and angiogenic factors such as TNF-α, COX-2, and VEGF. Both corneal opacity and corneal neovascularization (CNV) were significantly decreased in the PEP-1-FK506BP treated group. Our results showed that PEP-1-FK506BP can significantly inhibit alkali burn-induced corneal inflammation in rats, possibly by accelerating corneal wound healing and by reducing the production of angiogenic factors and inflammatory cytokines. These results suggest that PEP-1-FK506BP may be a potential therapeutic agent for CAI.


Subject(s)
Burns, Chemical/drug therapy , Cornea/drug effects , Corneal Injuries/prevention & control , Eye Burns/drug therapy , Tacrolimus Binding Proteins/pharmacology , Animals , Burns, Chemical/pathology , Cornea/pathology , Corneal Neovascularization/metabolism , Disease Models, Animal , Eye Burns/pathology , Inflammation/metabolism , Male , Peptides/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
17.
Mol Immunol ; 63(2): 355-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25239864

ABSTRACT

Reactive oxygen species (ROS) accumulation induces oxidative stress and cell damage, which then activates several signaling pathways and triggers inflammatory response. Biliverdin is a natural product of heme metabolism which is converted to bilirubin by the enzyme biliverdin reductase A (BLVRA) which also plays a role in antioxidant activity via the ROS scavenging activity of bilirubin. In this study, we examined the anti-inflammatory and anti-apoptotic effects of Tat-BLVRA protein on lipopolysaccharide (LPS)-induced inflammation in Raw 264.7 macrophage cells. Transduction of Tat-BLVRA protein into Raw 264.7 cells and mice ear tissue was tested by Western blot analysis and immunohistochemical analysis. Tat-BLVRA protein was effective in inhibiting mitogen activated protein kinases (MAPKs), Akt and NF-κB activation, intracellular ROS production and DNA fragmentation. Also, Tat-BLVRA protein significantly inhibited the expression of cytokines, COX-2, and iNOS. In a 12-O-tetradecanoylphobol 13-acetate (TPA)-induced mouse model, mice ears treated with Tat-BLVRA protein showed decreased ear thickness and weight, as well as inhibited MAPKs activation and cytokine expression. Thus we suggested that Tat-BLVRA protein may provide an effective therapeutic agent for inflammatory skin diseases.


Subject(s)
Edema/therapy , Inflammation/pathology , Macrophages/pathology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Animals , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Edema/pathology , Humans , Inflammation/enzymology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Male , Mice, Inbred ICR , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/isolation & purification , Signal Transduction , Tetradecanoylphorbol Acetate , Transduction, Genetic , tat Gene Products, Human Immunodeficiency Virus/metabolism
18.
Free Radic Biol Med ; 69: 181-96, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440593

ABSTRACT

Human carbonyl reductase 1 (CBR1) is a member of the NADPH-dependent short-chain dehydrogenase/reductase superfamily that is known to play an important role in neuronal cell survival via its antioxidant function. Oxidative stress is one of the major causes of degenerative disorders including ischemia. However, the role CBR1 plays with regard to ischemic injury is as yet poorly understood. Protein transduction domains such as PEP-1 are well known and now commonly used to deliver therapeutic proteins into cells. In this study, we prepared PEP-1-CBR1 protein and examined whether it protects against oxidative-stress-induced neuronal cell damage. PEP-1-CBR1 protein was efficiently transduced into hippocampal neuronal HT-22 cells and protected against hydrogen peroxide (H2O2)-induced neuronal cell death. Transduced PEP-1-CBR1 protein drastically inhibited H2O2-induced reactive oxygen species production, the oxidation of intracellular macromolecules, and the activation of mitogen-activated protein kinases, as well as cellular apoptosis. Furthermore, we demonstrated that transduced PEP-1-CBR1 protein markedly protected against neuronal cell death in the CA1 region of the hippocampus resulting from ischemic injury in an animal model. In addition, PEP-1-CBR1 protein drastically reduced activation of glial cells and lipid peroxidation in an animal model. These results indicate that PEP-1-CBR1 protein significantly protects against oxidative-stress-induced neuronal cell death in vitro and in vivo. Therefore, we suggest that PEP-1-CBR1 protein may be a therapeutic agent for the treatment of ischemic injuries as well as oxidative-stress-induced cell damage and death.


Subject(s)
Alcohol Oxidoreductases/genetics , Ischemia/genetics , Oxidative Stress , Recombinant Fusion Proteins/genetics , Alcohol Oxidoreductases/metabolism , Antioxidants/metabolism , Apoptosis/genetics , Cell Survival/genetics , Cysteamine/analogs & derivatives , Cysteamine/metabolism , Hippocampus/metabolism , Humans , Ischemia/etiology , Ischemia/pathology , Neurons/pathology , Peptides/genetics , Peptides/metabolism , Reactive Oxygen Species , Recombinant Fusion Proteins/metabolism
19.
Free Radic Biol Med ; 63: 432-45, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770196

ABSTRACT

Sirtuin 2 (SIRT2), a member of the sirtuin family of proteins, plays an important role in cell survival. However, the biological function of SIRT2 protein is unclear with respect to inflammation and oxidative stress. In this study, we examined the protective effects of SIRT2 on inflammation and oxidative stress-induced cell damage using a cell permeative PEP-1-SIRT2 protein. Purified PEP-1-SIRT2 was transduced into RAW 264.7 cells in a time- and dose-dependent manner and protected against lipopolysaccharide- and hydrogen peroxide (H2O2)-induced cell death and cytotoxicity. Also, transduced PEP-1-SIRT2 significantly inhibited the expression of cytokines as well as the activation of NF-κB and mitogen-activated protein kinases (MAPKs). In addition, PEP-1-SIRT2 decreased cellular levels of reactive oxygen species (ROS) and of cleaved caspase-3, whereas it elevated the expression of antioxidant enzymes such as MnSOD, catalase, and glutathione peroxidase. Furthermore, topical application of PEP-1-SIRT2 to 12-O-tetradecanoylphorbol 13-acetate-treated mouse ears markedly inhibited expression levels of COX-2 and proinflammatory cytokines as well as the activation of NF-κB and MAPKs. These results demonstrate that PEP-1-SIRT2 inhibits inflammation and oxidative stress by reducing the levels of expression of cytokines and ROS, suggesting that PEP-1-SIRT2 may be a potential therapeutic agent for various disorders related to ROS, including skin inflammation.


Subject(s)
Inflammation/metabolism , Oxidative Stress , Peptides/metabolism , Sirtuin 2/metabolism , Animals , Antioxidants/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Survival/drug effects , Cell Survival/genetics , Hydrogen Peroxide/toxicity , Inflammation/drug therapy , Inflammation/pathology , Lipopolysaccharides/toxicity , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Mice , Peptides/administration & dosage , Peptides/chemical synthesis , Peptides/genetics , Phorbol Esters/toxicity , Reactive Oxygen Species/metabolism , Sirtuin 2/administration & dosage , Sirtuin 2/genetics , Skin/drug effects , Skin/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...