Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add filters








Year range
1.
Biomolecules & Therapeutics ; : 321-330, 2021.
Article in English | WPRIM | ID: wpr-897318

ABSTRACT

Oxidative stress plays a crucial role in the development of neuronal disorders including brain ischemic injury. Thioredoxin 1 (Trx1), a 12 kDa oxidoreductase, has anti-oxidant and anti-apoptotic functions in various cells. It has been highly implicated in brain ischemic injury. However, the protective mechanism of Trx1 against hippocampal neuronal cell death is not identified yet. Using a cell permeable Tat-Trx1 protein, protective mechanism of Trx1 against hydrogen peroxide-induced cell death was examined using HT-22 cells and an ischemic animal model. Transduced Tat-Trx1 markedly inhibited intracellular ROS levels, DNA fragmentation, and cell death in H 2O 2-treatment HT-22 cells. Tat-Trx1 also significantly inhibited phosphorylation of ASK1 and MAPKs in signaling pathways of HT-22 cells. In addition, Tat-Trx1 regulated expression levels of Akt, NF-κB, and apoptosis related proteins. In an ischemia animal model, Tat-Trx1 markedly protected hippocampal neuronal cell death and reduced astrocytes and microglia activation. These findings indicate that transduced Tat-Trx1 might be a potential therapeutic agent for treating ischemic injury.

2.
Biomolecules & Therapeutics ; : 321-330, 2021.
Article in English | WPRIM | ID: wpr-889614

ABSTRACT

Oxidative stress plays a crucial role in the development of neuronal disorders including brain ischemic injury. Thioredoxin 1 (Trx1), a 12 kDa oxidoreductase, has anti-oxidant and anti-apoptotic functions in various cells. It has been highly implicated in brain ischemic injury. However, the protective mechanism of Trx1 against hippocampal neuronal cell death is not identified yet. Using a cell permeable Tat-Trx1 protein, protective mechanism of Trx1 against hydrogen peroxide-induced cell death was examined using HT-22 cells and an ischemic animal model. Transduced Tat-Trx1 markedly inhibited intracellular ROS levels, DNA fragmentation, and cell death in H 2O 2-treatment HT-22 cells. Tat-Trx1 also significantly inhibited phosphorylation of ASK1 and MAPKs in signaling pathways of HT-22 cells. In addition, Tat-Trx1 regulated expression levels of Akt, NF-κB, and apoptosis related proteins. In an ischemia animal model, Tat-Trx1 markedly protected hippocampal neuronal cell death and reduced astrocytes and microglia activation. These findings indicate that transduced Tat-Trx1 might be a potential therapeutic agent for treating ischemic injury.

3.
Experimental Neurobiology ; : 612-627, 2019.
Article in English | WPRIM | ID: wpr-763785

ABSTRACT

Aldose reductase (AR) protein, a member of the NADPH-dependent aldo-keto reductase family, reduces a wide range of aldehydes and enhances cell survival by inhibition of oxidative stress. Oxidative stress is known as one of the major pathological factor in ischemia. Since the precise function of AR protein in ischemic injury is fully unclear, we examined the function of AR protein in hippocampal neuronal (HT-22) cells and in an animal model of ischemia in this study. Cell permeable Tat-AR protein was produced by fusion of protein transduction domain in Tat for delivery into the cells. Tat-AR protein transduced into HT-22 cells and significantly inhibited cell death and regulated the mitogen-activate protein kinases (MAPKs), Bcl-2, Bax, and Caspase-3 under oxidative stress condition. In an ischemic animal model, Tat-AR protein transduced into the brain tissues through the blood-brain barrier (BBB) and drastically decreased neuronal cell death in hippocampal CA1 region. These results indicate that transduced Tat-AR protein has protective effects against oxidative stress-induced neuronal cell death in vitro and in vivo, suggesting that Tat-AR protein could be used as potential therapeutic agent in ischemic injury.


Subject(s)
Humans , Aldehyde Reductase , Aldehydes , Blood-Brain Barrier , Brain , CA1 Region, Hippocampal , Caspase 3 , Cell Death , Cell Survival , In Vitro Techniques , Ischemia , Models, Animal , Neurons , Oxidative Stress , Oxidoreductases , Protein Kinases
4.
Chinese Medical Journal ; (24): 3758-3763, 2014.
Article in English | WPRIM | ID: wpr-240690

ABSTRACT

<p><b>BACKGROUND</b>Oenanthe javanica is an aquatic perennial herb originated from East Asia. Nowadays, the effects of Oenanthe javanica have been proven in various disease models. Studies regarding the antioxidant effect of Oenanthe javanica in the kidney are still unclear.</p><p><b>METHODS</b>This study was therefore performed to investigate the effect of the Oenanthe javanica extract (OJE) in the rat kidney using immunohistochemistry for antioxidant enzymes, copper, zinc-superoxide dismutase (SOD1), manganese superoxide dismutase (SOD2), catalase (CAT) and glutathione peroxidase (GPx). Sprague-Dawley rats were randomly assigned to three groups: (1) normal diet fed-group (normal-group), (2) diet containing ascorbic acid (AA)-fed group (AA-group) as a positive control, (3) diet containing OJE-fed group (OJE-group). AA and OJE were supplied during 28 days.</p><p><b>RESULTS</b>The side-effects were not observed in all the groups. Immunoreactivities of SOD1, SOD2, CAT and GPx were easily detected in the distal tubules of the kidney, and their immunoreactivities in the AA-and OJE-groups were increased to about 1.4-1.5 times and 2 times, respectively, compared with those in the normal-group.</p><p><b>CONCLUSION</b>OJE significantly increased expressions of SOD1 & 2, CAT and GPx immunoreactivities in the distal tubules of the rat kidney, and this finding suggests that significant enhancements of endogenous enzymatic antioxidants by OJE treatment may be a legitimate strategy for decreasing oxidative stresses in the kidney.</p>


Subject(s)
Animals , Male , Rats , Antioxidants , Metabolism , Catalase , Metabolism , Glutathione Peroxidase , Metabolism , Kidney , Metabolism , Oenanthe , Chemistry , Oxidative Stress , Plant Extracts , Chemistry , Pharmacology , Rats, Sprague-Dawley , Superoxide Dismutase , Metabolism
6.
Experimental Neurobiology ; : 172-172, 2012.
Article in English | WPRIM | ID: wpr-110122

ABSTRACT

On page 173, the incorrect image which was not submitted by the author was mistakenly printed for Fig. 5 by a system error of the editing company.

7.
Experimental & Molecular Medicine ; : 586-593, 2012.
Article in English | WPRIM | ID: wpr-14964

ABSTRACT

Reactive oxygen species (ROS) contribute to the development of a number of neuronal diseases including ischemia. DJ-1, also known to PARK7, plays an important role in transcriptional regulation, acting as molecular chaperone and antioxidant. In the present study, we investigated whether DJ-1 protein shows a protective effect against oxidative stress-induced neuronal cell death in vitro and in ischemic animal models in vivo. To explore DJ-1 protein's potential role in protecting against ischemic cell death, we constructed cell permeable Tat-DJ-1 fusion proteins. Tat-DJ-1 protein efficiently transduced into neuronal cells in a dose- and time-dependent manner. Transduced Tat-DJ-1 protein increased cell survival against hydrogen peroxide (H2O2) toxicity and also reduced intracellular ROS. In addition, Tat-DJ-1 protein inhibited DNA fragmentation induced by H2O2. Furthermore, in animal models, immunohistochemical analysis revealed that Tat-DJ-1 protein prevented neuronal cell death induced by transient forebrain ischemia in the CA1 region of the hippocampus. These results demonstrate that transduced Tat-DJ-1 protein protects against cell death in vitro and in vivo, suggesting that the transduction of Tat-DJ-1 may be useful as a therapeutic agent for ischemic injuries related to oxidative stress.


Subject(s)
Animals , Mice , Rats , Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , CA1 Region, Hippocampal/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Gerbillinae , Intracellular Signaling Peptides and Proteins/administration & dosage , Lipid Peroxidation , Malondialdehyde/metabolism , Neuroprotective Agents/administration & dosage , Oncogene Proteins/administration & dosage , Oxidative Stress , Prosencephalon/drug effects , Recombinant Fusion Proteins/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
8.
Experimental Neurobiology ; : 169-175, 2011.
Article in English | WPRIM | ID: wpr-73126

ABSTRACT

Chlorogenic acid (CGA) possesses various biological activities such as anti-oxidant, anti-inflammatory, and anti-diabetic activities. In the present study, we examined the effect of CGA on the transduction efficiency of PEP-1-ribosomal protein S3 (PEP-1-rpS3) into cells and brain tissues, and its neuroprotective potential against ischemia/reperfusion. We found that, in the presence of CGA, the transduction efficiency of PEP-1-rpS3 into astrocytes and the CA1 region of the hippocampus was enhanced, compared to its transduction in the absence of CGA. Also, cell viability data demonstrated that the sample treated with CGA + PEP-1-rpS3 exhibited improved cell viability against hydrogen peroxide (H2O2)-induced toxicity more significantly than the sample treated with PEP-1-rpS3 alone. Also, in a gerbil ischemia model, data demonstrated that following the ischemic insult, the group treated with PEP-1-rpS3 + CGA showed markedly enhanced protection of neuron cells in CA1 region of hippocampus, compared to those treated with CGA or PEP-1-rpS3 alone. Taken together, these results suggest that CGA may improve the transduction efficiency of protein transduction domain (PTD) fusion proteins into target cells or tissues, thereby enhancing their therapeutic potential against various diseases.


Subject(s)
Astrocytes , Brain , Cell Survival , Chlorogenic Acid , Gerbillinae , Hippocampus , Hydrogen Peroxide , Ischemia , Neurons , Neuroprotective Agents , Proteins
9.
Experimental & Molecular Medicine ; : 629-637, 2009.
Article in English | WPRIM | ID: wpr-10783

ABSTRACT

Inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2) have been known to be involved in various pathophysiological processes such as inflammation. This study was performed to determine the regulatory function of superoxide dismutase (SOD) on the LPS-induced expression of iNOS, and COX-2 in RAW 264.7 cells. When a cell-permeable SOD, Tat-SOD, was added to the culture medium of RAW 264.7 cells, it rapidly entered the cells in a dose-dependent manner. Treatment of RAW 264.7 cells with Tat-SOD led to decrease in LPS-induced ROS generation. Pretreatment with Tat-SOD significantly inhibited LPS-induced expression of iNOS and NO production but had no effect on the expression of COX-2 and PGE2 production in RAW 264.7 cells. Tat-SOD inhibited LPS-induced NF-kappaB DNA binding activity, IkappaBalpha degradation and activation of MAP kinases. These data suggest that SOD differentially regulate expression of iNOS and COX-2 in LPS-stimulated RAW 264.7 cells.


Subject(s)
Animals , Mice , Cell Line , Cyclooxygenase 2/genetics , Cytokines/immunology , Gene Expression Regulation , Lipopolysaccharides/immunology , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
10.
Experimental & Molecular Medicine ; : 86-93, 2009.
Article in English | WPRIM | ID: wpr-103080

ABSTRACT

The infiltration of monocytes into the CNS represents one of the early steps to inflammatory events in AIDS-related encephalitis and dementia. Increased activity of selected matrix metalloproteinases (MMPs) such as MMP-9 impairs the integrity of blood-brain barrier leading to enhanced monocyte infiltration into the CNS. In this study, we examined the effect of HIV-1 Tat on the expression of MMP-9 in CRT-MG human astroglioma cells. Treatment of CRT-MG cells with HIV-1 Tat protein significantly increased protein levels of MMP-9, as measured by Western blot analysis, zymography and an ELISA. Treatment of CRT-MG cells with HIV-1 Tat protein markedly increased mRNA levels of MMP-9, as analyzed by RT-PCR. Pretreatment of CRT-MG cells with NF-kappaB inhibitors led to decrease in Tat-induced protein and mRNA expression of MMP-9. Pretreatment of CRT-MG cells with MAPK inhibitors suppressed Tat-induced MMP-9 expression. Furthermore, HIV-1 Tat-induced expression of MMP-9 was significantly inhibited by neutralization of TNF-alpha, but not IL-1beta and IL-6. Taken together, our results indicate that HIV-1 Tat can up-regulate expression of MMP-9 via MAPK-NF-kappaB-dependent mechanisms as well as Tat-induced TNF-alpha production in astrocytes.


Subject(s)
Humans , AIDS Dementia Complex/metabolism , Astrocytes/drug effects , HIV Infections/complications , HIV-1 , Matrix Metalloproteinase 9/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/drug effects , tat Gene Products, Human Immunodeficiency Virus/metabolism
11.
Experimental & Molecular Medicine ; : 629-638, 2008.
Article in English | WPRIM | ID: wpr-59825

ABSTRACT

Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) is a tumor suppressor. Although it is well known to have various physiological roles in cancer, its inhibitory effect on inflammation remains poorly understood. In the present study, a human PTEN gene was fused with PEP-1 peptide in a bacterial expression vector to produce a genetic in-frame PEP-1-PTEN fusion protein. The expressed and purified PEP-1-PTEN fusion protein were transduced efficiently into macrophage Raw 264.7 cells in a time- and dose- dependent manner when added exogenously in culture media. Once inside the cells, the transduced PEP-1-PTEN protein was stable for 24 h. Transduced PEP-1-PTEN fusion protein inhibited the LPS-induced cyclooxygenase 2 (COX-2) and iNOS expression levels in a dose-dependent manner. Furthermore, transduced PEP-1-PTEN fusion protein inhibited the activation of NF-kappa B induced by LPS. These results suggest that the PEP-1-PTEN fusion protein can be used in protein therapy for inflammatory disorders.


Subject(s)
Animals , Humans , Mice , Cell Line , Cyclooxygenase 2/metabolism , Cysteamine/analogs & derivatives , Enzyme Activation , Lipopolysaccharides/pharmacology , Macrophages/metabolism , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , PTEN Phosphohydrolase/genetics , Peptides/genetics , Recombinant Fusion Proteins/biosynthesis , Signal Transduction
12.
Experimental & Molecular Medicine ; : 27-37, 2007.
Article in English | WPRIM | ID: wpr-37558

ABSTRACT

One of characteristic features of AIDS-related encephalitis and dementia is the infiltration of monocytes into the CNS. HIV-1 Tat was demonstrated to facilitate monocyte entry into the CNS. In this study, we examined the effect of HIV-1 Tat on the expression of adhesion molecules, generation of reactive oxygen species (ROS) and NF-kappaB activation in CRT-MG human astroglioma cells. Treatment of CRT-MG cells with HIV-1 Tat protein significantly increased protein and mRNA levels of ICAM-1 and VCAM-1, as measured by Western blot analysis and RT-PCR, indicating that Tat increases these protein levels at an mRNA level. In addition, Tat induced the activation of NF-kappaB in astrocytes. Treatment of CRT-MG with NF-kappaB inhibitors led to decrease in Tat-induced protein and mRNA expression of ICAM-1 and VCAM-1. Furthermore, HIV-1 Tat protein increased ROS generation. Inhibition of Tat-induced ROS generation by N-acetyl cysteine, vitamin C and diphenyl iodonium suppressed Tat-induced NF-kappaB activation, ICAM-1 and VCAM-1 expression, and monocyte adhesion in CRT-MG. These data indicate that HIV-1 Tat can modulate monocyte adhesiveness by increasing expression of adhesion molecules such as ICAM-1 and VCAM-1 via ROS- and NF-kappaB-dependent mechanisms in astrocytes.


Subject(s)
Humans , Vascular Cell Adhesion Molecule-1/genetics , Up-Regulation/drug effects , Transcription, Genetic/genetics , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Monocytes/cytology , Intercellular Adhesion Molecule-1/genetics , HIV-1 , Gene Products, tat/pharmacology , Cell Line , Cell Adhesion/drug effects , Astrocytes/cytology
13.
Experimental & Molecular Medicine ; : 778-786, 2007.
Article in English | WPRIM | ID: wpr-21101

ABSTRACT

HIV-1 Tat is considered to be one of key players to facilitate monocyte entry into the CNS, which is characteristic feature of AIDS-related encephalitis and dementia. This study was performed to determine the regulatory function of superoxide dismutase (SOD) on the HIV-1 Tat-induced signaling pathways leading to NF-kappaB activation, expression of adhesion molecules, and monocyte adhesion in CRT-MG human astroglioma cells by using cell-permeable SOD. When cell-permeable SOD was added to the culture medium of CRT-MG cells, it rapidly entered the cells in dose- and time-dependent manners. Treatment of astrocytes with cell-permeable SOD led to decrease in Tat-induced ROS generation as well as NF-kappaB activation. Cell-permeable SOD inhibited the activation of MAP kinases including ERK, JNK and p38 by HIV-1 Tat. Treatment of CRT-MG cells with cell-permeable SOD significantly inhibited protein and mRNA levels of ICAM-1 and VCAM-1 up-regulated by HIV-1 Tat, as measured by Western blot analysis and RT-PCR. Furthermore, enhanced adhesiveness of monocyte to astrocyte by HIV-1 Tat was significantly abrogated by pretreatment with cell-permeable SOD fusion proteins. These data indicate that SOD has a regulatory function for HIV-1 Tat-induced NF-kappaB activation in astrocytes and suggest that cell-permeable SOD can be used as a feasible therapeutic agent for regulation of ROS-related neurological diseases.


Subject(s)
Humans , Astrocytes/enzymology , Cell Adhesion/physiology , Cell Membrane Permeability , Gene Products, tat/pharmacology , HIV Infections/metabolism , HIV-1/chemistry , Monocytes/cytology , Signal Transduction , Superoxide Dismutase/genetics
14.
Experimental & Molecular Medicine ; : 575-587, 2005.
Article in English | WPRIM | ID: wpr-191493

ABSTRACT

Rac1 and Rac2 are essential for the control of oxidative burst catalyzed by NADPH oxidase. It was also documented that Rho is associated with the superoxide burst reaction during phagocytosis of serum- (SOZ) and IgG-opsonized zymosan particles (IOZ). In this study, we attempted to reveal the signal pathway components in the superoxide formation regulated by Rho GTPase. Tat-C3 blocked superoxide production, suggesting that RhoA is essentially involved in superoxide formation during phagocytosis of SOZ. Conversely SOZ activated both RhoA and Rac1/2. Inhibition of RhoA-activated kinase (ROCK), an important downstream effector of RhoA, by Y27632 and myosin light chain kinase (MLCK) by ML-7 abrogated superoxide production by SOZ. Extracellular signaling-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) were activated during phagocytosis of SOZ, and Tat-C3 and SB203580 reduced ERK1/2 and p38 MAPK activation, suggesting that RhoA and p38 MAPK may be upstream regulators of ERK1/2. Inhibition of ERK1/2, p38 MAPK, phosphatidyl inositol 3-kinase did not block translocation of RhoA to membranes, suggesting that RhoA is upstream to these kinases. Inhibition of RhoA by Tat-C3 blocked phosphorylation of p47 PHOX. Taken together, RhoA, ROCK, p38MAPK, ERK1/2, and p47 PHOX may be subsequently activated, leading to activation of NADPH oxidase to produce superoxide.


Subject(s)
Animals , Mice , Cell Line , Cell Membrane , Cytosol , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Macrophage-1 Antigen/pharmacology , Macrophages/drug effects , Myosin-Light-Chain Kinase/metabolism , Opsonin Proteins/blood , Phagocytosis , Protein Transport , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Superoxides/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Zymosan/blood , p38 Mitogen-Activated Protein Kinases/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors
15.
Experimental & Molecular Medicine ; : 43-51, 2004.
Article in English | WPRIM | ID: wpr-190975

ABSTRACT

Enzyme/prodrug approach is one of the actively developing areas for cancer therapy. In an effort to develop more effective enzyme/prodrug systems, cell-permeable cytosine deaminase was produced by fusing yeast cytosine deaminase (yCD) in frame with RKKRRQRRR domain of HIV-1 Tat which is an efficient delivery peptide of the foreign proteins into cells. The purified Tat-yCD fusion protein expressed in Escherichia coli was readily transduced into mammalian cells in a time- and dose-dependent manner. A significant level of the transduced Tat-yCD protein was recovered in the cell and was stable for 24 h as indicated by both results of the enzymatic assay of 5-fluorocytosine (5-FC) conversion to 5-fluorouracil (5-FU) and Western blot analysis. The cells transduced with Tat-yCD become highly sensitive to the cytotoxicity of 5-FC, while cells treated with yCD are unaffected by 5-FC. In addition, a strong bystander effect was observed with conditioned media from cells transduced with Tat-yCD added to non-transduced cells. Tat-yCD fusion protein demonstrated here for its ability to transduce into cells and convert nontoxic prodrug 5-FC to the toxic antimetabolite 5-FU, may be a useful approach for cancer therapy.


Subject(s)
Animals , Humans , Antimetabolites/metabolism , Bystander Effect , Cytosine Deaminase/genetics , Flucytosine/metabolism , Gene Products, tat/chemistry , Genetic Vectors/genetics , HIV-1/metabolism , HeLa Cells/drug effects , Prodrugs/metabolism , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Transduction, Genetic
16.
The Korean Journal of Physiology and Pharmacology ; : 163-168, 2003.
Article in English | WPRIM | ID: wpr-727908

ABSTRACT

The reactive oxygen species (ROS) are considered to be an important mediator in pancreatic beta cell destruction, thereby triggering the development of insulin-dependent diabetes mellitus. In the present study, HIV-1 Tat-mediated transduction of Cu, Zn-superoxide dismutase (SOD) was investigated to evaluate its protective potential against streptozotocin (STZ) -induced cytotoxicity in insulin-producing MIN6N cells. Tat-SOD fusion protein was successfully delivered into MIN6N cells in a dose-dependent manner and the transduced fusion protein was enzymatically active for 48 h. The STZ induced-cell destruction, superoxide anion radical production, and DNA fragmentation of MIN6N cells were significantly decreased in the cells pretreated with Tat-SOD for 1 h. Furthermore, the transduction of Tat-SOD increased Bcl-2 and heat shock protein 70 (hsp70) expressions in cells exposed to STZ, which might be partly responsible for the effect of Tat-SOD. These results suggest that an increased of free radical scavenging activity by transduction of Tat-SOD enhanced the tolerance of the cell against oxidative stress in STZ-treated MIN6N cells. Therefore, this Tat-SOD transduction technique may provide a new strategy to protect the pancreatic beta cell destruction in ROS-mediated diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , DNA Fragmentation , HIV-1 , HSP70 Heat-Shock Proteins , Insulin-Secreting Cells , Oxidative Stress , Reactive Oxygen Species , Streptozocin , Superoxide Dismutase , Superoxides
17.
Experimental & Molecular Medicine ; : 249-256, 2003.
Article in English | WPRIM | ID: wpr-13860

ABSTRACT

Five monoclonal antibodies (mAbs) that recognize human glutamate dehydrogenase (GDH) have been selected and designated as monoclonal antibodies hGDH60-6, hGDH60-8, hGDH63-10, hGDH63-11, and hGDH91-14. A total of five mAbs recognizing different epitopes of the enzyme were obtained, two of which inhibited human GDH activity. When total proteins of human homogenate separated by SDS- PAGE, were probed with mAbs, a single reactive protein band of 55 kDa, which co-migrated with purified recombinant human GDH was detected. When the purified GDH was incubated with each of the mAbs, its enzyme activity was inhibited by up to 58%. Epitope mapping analysis identified, two subgroups of mAbs recognizing different peptide fragments. Using the individual anti-GDH antibodies as probes, the cross reactivities of brain GDH obtained from human and other animal brain tissues were investigated. For the human and animal tissues tested, immunoreactive bands on Western blots appeared to have the same molecular mass of 55 kDa when hGHD60-6, hGHD60-8, or hGHD91-14 mAbs were used as probes. However, the anti-human GDH mAbs immunoreactive to bands on Western blots reacted differently on the immunoblots of the other animal brains tested, i.e., the two monoclonal antibodies hGDH63-10 and hGDH63-11 only produced positive results for human. These results suggest that human brain GDH is immunologically distinct from those of other mammalian brains. Thorough characterization of these anti-human GDH mAbs could provide potentially valuable tool as immunodiagnostic reagents for the detection, identification and characterization of the various neurological diseases related to the GDH enzyme.


Subject(s)
Animals , Humans , Mice , Rats , Antibodies, Monoclonal/immunology , Cross Reactions , Electrophoresis, Polyacrylamide Gel , Glutamate Dehydrogenase/classification , Organ Specificity
SELECTION OF CITATIONS
SEARCH DETAIL