Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add filters








Year range
1.
Cancer Research and Treatment ; : 797-811, 2019.
Article in English | WPRIM | ID: wpr-763116

ABSTRACT

PURPOSE: In the present study, human neural stem cells (hNSCs) with tumor-tropic behavior were used as drug delivery vehicle to selectively target melanoma. A hNSC line (HB1.F3) was transduced into two types: one expressed only the cytosine deaminase (CD) gene (HB1.F3. CD) and the other expressed both CD and human interferon-β (IFN-β) genes (HB1.F3.CD. IFN-β). MATERIALS AND METHODS: This study verified the tumor-tropic migratory competence of engineered hNSCs on melanoma (A375SM) using a modified Boyden chamber assay in vitro and CM-DiI staining in vivo. The antitumor effect of HB1.F3.CD and HB1.F3.CD.IFN-β on melanoma was also confirmed using an MTT assay in vitro and xenograft mouse models. RESULTS: A secreted form of IFN-β from the HB1.F3.CD.IFN-β cells modified the epithelial-mesenchymal transition (EMT) process and metastasis of melanoma. 5-Fluorouracil treatment also accelerated the expression of the pro-apoptotic protein BAX and decelerated the expression of the anti-apoptotic protein Bcl-xL on melanoma cell line. CONCLUSION: Our results illustrate that engineered hNSCs prevented malignant melanoma cells from proliferating in the presence of the prodrug, and the form that secreted IFN-β intervened in the EMT process and melanoma metastasis. Hence, neural stem cell-directed enzyme/prodrug therapy is a plausible treatment for malignant melanoma.


Subject(s)
Animals , Humans , Mice , Cell Line , Cytosine Deaminase , Epithelial-Mesenchymal Transition , Flucytosine , Fluorouracil , Heterografts , In Vitro Techniques , Melanoma , Mental Competency , Neoplasm Metastasis , Neural Stem Cells , Stem Cells
2.
Cancer Research and Treatment ; : 79-91, 2017.
Article in English | WPRIM | ID: wpr-127967

ABSTRACT

PURPOSE: Genetically engineered stem cells may be advantageous for gene therapy against various human cancers due to their inherent tumor-tropic properties. In this study, genetically engineered human neural stem cells (HB1.F3) expressing Escherichia coli cytosine deaminase (CD) (HB1.F3.CD) and human interferon-β (IFN-β) (HB1.F3.CD.IFN-β) were employed against lymph node–derived metastatic colorectal adenocarcinoma. MATERIALS AND METHODS: CD can convert a prodrug, 5-fluorocytosine (5-FC), to active 5-fluorouracil, which inhibits tumor growth through the inhibition of DNA synthesis,while IFN-β also strongly inhibits tumor growth by inducing the apoptotic process. In reverse transcription polymerase chain reaction analysis, we confirmed that HB1.F3.CD cells expressed the CD gene and HB1.F3.CD.IFN-β cells expressed both CD and IFN-β genes. RESULTS: In results of a modified trans-well migration assay, HB1.F3.CD and HB1.F3.CD.IFN-β cells selectively migrated toward SW-620, human lymph node–derived metastatic colorectal adenocarcinoma cells. The viability of SW-620 cells was significantly reduced when co-cultured with HB1.F3.CD or HB1.F3.CD.IFN-β cells in the presence of 5-FC. In addition, it was found that the tumor-tropic properties of these engineered human neural stem cells (hNSCs) were attributed to chemoattractant molecules including stromal cell-derived factor 1, c-Kit, urokinase receptor, urokinase-type plasminogen activator, and C-C chemokine receptor type 2 secreted by SW-620 cells. In a xenograft mouse model, treatment with hNSC resulted in significantly inhibited growth of the tumor mass without virulent effects on the animals. CONCLUSION: The current results indicate that engineered hNSCs and a prodrug treatment inhibited the growth of SW-620 cells. Therefore, hNSC therapy may be a clinically effective tool for the treatment of lymph node metastatic colorectal cancer.


Subject(s)
Animals , Humans , Mice , Adenocarcinoma , Chemokine CXCL12 , Colorectal Neoplasms , Cytosine Deaminase , Cytosine , DNA , Escherichia coli , Flucytosine , Fluorouracil , Genetic Therapy , Heterografts , Interferon-beta , Lymph Nodes , Lymphatic Metastasis , Neural Stem Cells , Polymerase Chain Reaction , Reverse Transcription , Stem Cells , Urokinase-Type Plasminogen Activator
3.
Experimental Neurobiology ; : 45-52, 2014.
Article in English | WPRIM | ID: wpr-187155

ABSTRACT

The loss of neuronal cells in the central nervous system may occur in many neurodegenerative diseases. Alzheimer's disease is a common senile disease in people over 65 years, and it causes impairment characterized by the decline of mental function, including memory loss and cognitive impairment, and affects the quality of life of patients. However, the current therapeutic strategies against AD are only to relieve symptoms, but not to cure it. Because there are only a few therapeutic strategies against Alzheimer's disease, we need to understand the pathogenesis of this disease. Cell therapy may be a powerful tool for the treatment of Alzheimer's disease. This review will discuss the characteristics of Alzheimer's disease and various available therapeutic strategies.


Subject(s)
Humans , Alzheimer Disease , Cell- and Tissue-Based Therapy , Central Nervous System , Memory Disorders , Neurodegenerative Diseases , Neurons , Quality of Life , Stem Cells , Transplantation
4.
Clinical and Experimental Otorhinolaryngology ; : 176-183, 2013.
Article in English | WPRIM | ID: wpr-214429

ABSTRACT

OBJECTIVES: Based on studies of the extensive tropism of neural stem cells (NSCs) toward malignant brain tumor, we hypothesized that NSCs could also target head and neck squamous cell carcinoma (HNSCC) and could be used as a cellular therapeutic delivery system. METHODS: To apply this strategy to the treatment of HNSCC, we used a human NSC line expressing cytosine deaminase (HB1.F3-CD), an enzyme that converts 5-fluorocytosine (5-FC) into 5-fluorouracil (5-FU), an anticancer agent. HB1. F3-CD in combination with 5-FC were cocultured with the HNSCC (SNU-1041) to examine the cytotoxicity on target tumor cells in vitro. For in vivo studies, an HNSCC mouse model was created by subcutaneous implantation of human HNSCC cells into athymic nude mice. HB1.F3-CD cells were injected into mice using tumoral, peritumoral, or intravenous injections, followed by systemic 5-FC administration. RESULTS: In vitro, the HB1.F3-CD cells significantly inhibited the growth of an HNSCC cell line in the presence of the 5-FC. Independent of the method of injection, the HB1.F3-CD cells migrated to the HNSCC tumor, causing a significant reduction in tumor volume. In comparison to 5-FU administration, HB1.F3-CD cell injection followed by 5-FC administration reduced systemic toxicity, but achieved the same level of therapeutic efficacy. CONCLUSION: Transplantation of human NSCs that express the suicide enzyme cytosine deaminase combined with systemic administration of the prodrug 5-FC may be an effective regimen for the treatment of HNSCC.


Subject(s)
Animals , Humans , Mice , Brain Neoplasms , Carcinoma, Squamous Cell , Cell Line , Cytosine Deaminase , Flucytosine , Fluorouracil , Head , Head and Neck Neoplasms , Injections, Intravenous , Mice, Nude , Molecular Targeted Therapy , Neck , Neural Stem Cells , Suicide , Transplants , Tropism , Tumor Burden
5.
Laboratory Animal Research ; : 131-137, 2013.
Article in English | WPRIM | ID: wpr-226197

ABSTRACT

Stem cells derived from adult tissues or the inner cell mass (ICM) of embryos in the mammalian blastocyst (BL) stage are capable of self-renewal and have remarkable potential for undergoing lineage-specific differentiation under in vitro culturing conditions. In particular, neural stem cells (NSCs) that self-renew and differentiate into major cell types of the brain exist in the developing and adult central nervous system (CNS). The exact function and distribution of NSCs has been assessed, and they represent an interesting population that includes astrocytes, oligodendrocytes, and neurons. Many researchers have demonstrated functional recovery in animal models of various neurological diseases such as stroke, Parkinson's disease (PD), brain tumors, and metastatic tumors. The safety and efficacy of stem cell-based therapies (SCTs) are also being evaluated in humans. The therapeutic efficacy of NSCs has been shown in the brain disorder-induced animal models, and animal models may be well established to perform the test before clinical stage. Taken together, data from the literature have indicated that therapeutic NSCs may be useful for selectively treating diverse types of human brain diseases without incurring adverse effects.


Subject(s)
Adult , Animals , Humans , Astrocytes , Blastocyst , Brain , Brain Diseases , Brain Neoplasms , Central Nervous System , Embryonic Structures , Models, Animal , Neural Stem Cells , Neurons , Oligodendroglia , Parkinson Disease , Stem Cells , Stroke
6.
International Journal of Stem Cells ; : 79-83, 2012.
Article in English | WPRIM | ID: wpr-25518

ABSTRACT

BACKGROUND AND OBJECTIVES: Ischemic stroke caused by middle cerebral artery occlusion (MCAo) is the major type of stroke, but there are currently very limited options for cure. It has been shown that neural stem cells (NSCs) or neural precursor cells (NPCs) can survive and improve neurological deficits when they are engrafted in animal models of various neurological diseases. However, how the transplanted NSCs or NPCs are act in vivo in the injured or diseased brain is largely unknown. In this study, we utilized magnetic resonance imaging (MRI) techniques in order to understand the fates of human NSCs (HB1.F3) following transplantation into a rodent model of MCAo. METHODS AND RESULTS: HB1.F3 human NSCs were pre-labeled with ferumoxides (Feridex(R))-protamine sulfate complexes, which were visualized and examined by MRI up to 9 weeks after transplantation. Migration of the transplanted cells to the infarct area was further confirmed by histological methods. CONCLUSIONS: Based on these observations, we speculate that the transplanted NSCs have the extensive migratory ability to the injured site, which will in turn contribute to functional recovery in stroke.


Subject(s)
Humans , Brain , Dextrans , Infarction, Middle Cerebral Artery , Magnetic Resonance Imaging , Magnetite Nanoparticles , Models, Animal , Neural Stem Cells , Rodentia , Stroke , Track and Field , Transplants
7.
Laboratory Animal Research ; : 77-84, 2011.
Article in English | WPRIM | ID: wpr-77857

ABSTRACT

Periventricular leukomalacia, specifically characterized as white matter injury, in neonates is strongly associated with the damage of pre-myelinating oligodendrocytes. Clinical data suggest that hypoxia-ischemia during delivery and intrauterine or neonatal infection-inflammation are important factors in the etiology of periventricular leukomalacia including cerebral palsy, a serious case exhibiting neurobehavioral deficits of periventricular leukomalacia. In order to explore the pathophysiological mechanisms of white matter injury and to better understand how infectious agents may affect the vulnerability of the immature brain to injury, novel animal models have been developed using hypoperfusion, microbes or bacterial products (lipopolysaccharide) and excitotoxins. Such efforts have developed rat models that produce predominantly white matter lesions by adopting combined hypoxia-ischemia technique on postnatal days 1-7, in which unilateral or bilateral carotid arteries of animals are occluded (ischemia) followed by 1-2 hour exposure to 6-8% oxygen environment (hypoxia). Furthermore, low doses of lipopolysaccharide that by themselves have no adverse-effects in 7-day-old rats, dramatically increase brain injury to hypoxic-ischemic challenge, implying that inflammation sensitizes the immature central nervous system. Therefore, among numerous models of periventricular leukomalacia, combination of hypoxia-ischemia-lipopolysaccharide might be one of the most-acceptable rodent models to induce extensive white matter injury and ensuing neurobehavioral deficits for the evaluation of candidate therapeutics.


Subject(s)
Animals , Humans , Infant, Newborn , Rats , Brain , Brain Injuries , Carotid Arteries , Central Nervous System , Cerebral Palsy , Inflammation , Leukomalacia, Periventricular , Models, Animal , Neurotoxins , Oligodendroglia , Oxygen , Rodentia
8.
Experimental & Molecular Medicine ; : 24-34, 2011.
Article in English | WPRIM | ID: wpr-48417

ABSTRACT

Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) induces apoptosis selectively in cancer cells while sparing normal cells. However, many cancer cells are resistant to TRAIL-induced cell death. Here, we report that paxilline, an indole alkaloid from Penicillium paxilli, can sensitize various glioma cells to TRAIL-mediated apoptosis. While treatment with TRAIL alone caused partial processing of caspase-3 to its p20 intermediate in TRAIL-resistant glioma cell lines, co-treatment with TRAIL and subtoxic doses of paxilline caused complete processing of caspase-3 into its active subunits. Paxilline treatment markedly upregulated DR5, a receptor of TRAIL, through a CHOP/GADD153-mediated process. In addition, paxilline treatment markedly downregulated the protein levels of the short form of the cellular FLICE-inhibitory protein (c-FLIPS) and the caspase inhibitor, survivin, through proteasome-mediated degradation. Taken together, these results show that paxilline effectively sensitizes glioma cells to TRAIL-mediated apoptosis by modulating multiple components of the death receptor-mediated apoptotic pathway. Interestingly, paxilline/TRAIL co-treatment did not induce apoptosis in normal astrocytes, nor did it affect the protein levels of CHOP, DR5 or survivin in these cells. Thus, combined treatment regimens involving paxilline and TRAIL may offer an attractive strategy for safely treating resistant gliomas.


Subject(s)
Humans , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Astrocytes/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 3/metabolism , Cell Line, Tumor , Drug Discovery , Flow Cytometry , Glioma/metabolism , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , RNA, Small Interfering , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription Factor CHOP/analysis
9.
Experimental Neurobiology ; : 155-164, 2010.
Article in English | WPRIM | ID: wpr-122581

ABSTRACT

Dysfunctions of ubiquitin-proteasome system and toxicity of dopamine have been known as the key mechanisms in the pathogenesis of Parkinson's disease (PD) and proteasome inhibitors are widely used in experimental models of PD to reproduce cell death of dopaminergic neurons. In the present study, immortalized human neural stem cells (HB1.F3, F3) and those transfected with human aromatic acid decarboxylase gene (F3.AADC), were used to investigate the mechanism of selective dopaminergic neuronal cell death mediated by dopamine or proteasome inhibitors. Flow cytometric analysis revealed that F3.AADC was more susceptible to dopamine than parental F3 cell which does not carry dopaminergic phenotype. The dopamine-induced apoptosis was mediated by activation of caspases 3 and 9 and cleavage of PARP. Proteasome inhibitors also induced apoptosis in dose-dependent manner but there was no difference between cell types. Prolonged exposure to subtoxic dose of proteasome inhibitors further enhanced dopamine-induced apoptosis in the F3.AADC, and increased presence of alpha-synuclein and ubiquitin-positive inclusions was noted in the cytoplasm of apoptotic cells by immunocytochemistry. These findings indicate that dopaminergic cells are selectively susceptible to dopamine toxicity and prolonged suppression of proteasome system further enhances selective sensitivity to dopamine toxicity. Chronic subtoxic proteasomal dysfunction of dopaminergic cells might contribute to selective cell death of dopaminergic neurons during the pathogenesis of Parkinson's disease.


Subject(s)
Humans , alpha-Synuclein , Apoptosis , Caspases , Cell Death , Cytoplasm , Dopamine , Dopaminergic Neurons , Immunohistochemistry , Models, Theoretical , Neural Stem Cells , Parents , Parkinson Disease , Phenotype , Proteasome Endopeptidase Complex , Proteasome Inhibitors
10.
Experimental Neurobiology ; : 106-113, 2010.
Article in English | WPRIM | ID: wpr-162258

ABSTRACT

The molecular microenvironment of the injured spinal cord does not support survival and differentiation of either grafted or endogenous NSCs, restricting the effectiveness of the NSC-based cell replacement strategy. Studying the biology of NSCs in in vivo usually requires a considerable amount of time and cost, and the complexity of the in vivo system makes it difficult to identify individual environmental factors. The present study sought to establish the organotypic spinal cord slice culture that closely mimics the in vivo environment. The cultured spinal cord slices preserved the cytoarchitecture consisting of neurons in the gray matter and interspersed glial cells. The majority of focally applied exogenous NSCs survived up to 4 weeks. Pre-exposure of the cultured slices to a hypoxic chamber markedly reduced the survival of seeded NSCs on the slices. Differentiation into mature neurons was severely limited in this co-culture system. Endogenous neural progenitor cells were marked by BrdU incorporation, and applying an inflammatory cytokine IL-1beta significantly increased the extent of endogenous neural progenitors with the oligodendrocytic lineage. The present study shows that the organotypic spinal cord slice culture can be properly utilized to study molecular factors from the post-injury microenvironment affecting NSCs in the injured spinal cord.


Subject(s)
Hypoxia , Biology , Bromodeoxyuridine , Cellular Microenvironment , Coculture Techniques , Neural Stem Cells , Neuroglia , Neurons , Seeds , Spinal Cord , Spinal Cord Injuries , Stem Cells , Transplants
11.
Experimental & Molecular Medicine ; : 11-18, 2008.
Article in English | WPRIM | ID: wpr-219398

ABSTRACT

Recent studies have reported that the "cholinergic anti-inflammatory pathway" regulates peripheral inflammatory responses via alpha7 nicotinic acetylcholine receptors (alpha7 nAChRs) and that acetylcholine and nicotine regulate the expression of proinflammatory mediators such as TNF-alpha and prostaglandin E2 in microglial cultures. In a previous study we showed that ATP released by beta-amyloid-stimulated microglia induced reactive oxygen species (ROS) production, in a process involving the P2X7 receptor (P2X7R), in an autocrine fashion. These observations led us to investigate whether stimulation by nicotine could regulate fibrillar beta amyloid peptide (1-42) (fA beta(1-42))-induced ROS production by modulating ATP efflux-mediated Ca2+ influx through P2X7R. Nicotine inhibited ROS generation in fA beta(1-42)-stimulated microglial cells, and this inhibition was blocked by mecamylamine, a non-selective nAChR antagonist, and a-bungarotoxin, a selective alpha7 nAChR antagonist. Nicotine inhibited NADPH oxidase activation and completely blocked Ca2+ influx in fA beta(1-42)-stimulated microglia. Moreover, ATP release from fA beta(1-42)-stimulated microglia was significantly suppressed by nicotine treatment. In contrast, nicotine did not inhibit 2',3'-O-(4-benzoyl)-benzoyl ATP (BzATP)-induced Ca2+ influx, but inhibited ROS generation in BzATP-stimulated microglia, indicating an inhibitory effect of nicotine on a signaling process downstream of P2X7R. Taken together, these results suggest that the inhibitory effect of nicotine on ROS production in fA beta(1-42)-stimulated microglia is mediated by indirect blockage of ATP release and by directly altering the signaling process downstream from P2X7R.


Subject(s)
Animals , Rats , Adenosine Triphosphate/analogs & derivatives , Amyloid/metabolism , Amyloid beta-Peptides/pharmacology , Calcium/metabolism , Enzyme Activation/drug effects , Microglia/cytology , NADPH Oxidases/metabolism , Nicotine/pharmacology , Nicotinic Antagonists/pharmacology , Peptide Fragments/pharmacology , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, Nicotinic/metabolism , Receptors, Purinergic P2/metabolism
12.
Experimental & Molecular Medicine ; : 19-26, 2008.
Article in English | WPRIM | ID: wpr-219397

ABSTRACT

Previously we demonstrated that ATP released from LPS-activated microglia induced IL-10 expression in a process involving P2 receptors, in an autocrine fashion. Therefore, in the present study we sought to determine which subtype of P2 receptor was responsible for the modulation of IL-10 expression in ATP-stimulated microglia. We found that the patterns of IL-10 production were dose-dependent (1, 10, 100, 1,000 micrometer) and bell-shaped. The concentrations of ATP, ATP-gammaS, ADP, and ADP-beta S that showed maximal IL-10 release were 100, 10, 100, and 100 micrometer respectively. The rank order of agonist potency for IL-10 production was 2'-3'-O-(4-benzoyl)-benzoyl ATP (BzATP) = dATP > 2-methylthio-ADP (2-meSADP). On the other hand, 2-methylthio-ATP (2-meSATP), alpha,beta-methylene ATP (alpha,beta-meATP), UTP, and UDP did not induce the release of IL-10 from microglia. Further, we obtained evidence of crosstalk between P2 receptors, in a situation where intracellular Ca2+ release and/or cAMP-activated PKA were the main contributors to extracellular ATP-(or ADP)-mediated IL-10 expression, and IL-10 production was down- regulated by either MRS2179 (a P2Y1 antagonist) or 5'-AMPS (a P2Y11 antagonist), indicating that both the P2Y1 and P2Y11 receptors are major receptors involved in IL-10 expression. In addition, we found that inhibition of IL-10 production by high concentrations of ATP-gammaS (100 micrometer) was restored by TNP-ATP (an antagonist of the P2X1, P2X3, and P2X4 receptors), and that IL-10 production by 2-meSADP was restored by 2meSAMP (a P2Y12 receptor antagonist) or pertusis toxin (PTX; a Gi protein inhibitor), indicating that the P2X1, P2X3, P2X4 receptor group, or the P2Y12 receptor, negatively modulate the P2Y11 receptor or the P2Y1 receptor, respectively.


Subject(s)
Animals , Rats , Adenosine Diphosphate/analogs & derivatives , Adenosine Triphosphate/analogs & derivatives , Adenylyl Cyclases/antagonists & inhibitors , Calcium/metabolism , Chelating Agents/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Extracellular Space/drug effects , Gene Expression Regulation/drug effects , Interleukin-10/biosynthesis , Microglia/drug effects , RNA, Messenger/genetics , Rats, Sprague-Dawley , Receptor Cross-Talk/drug effects , Receptors, Purinergic P2/agonists , Thionucleotides/pharmacology
13.
Experimental & Molecular Medicine ; : 38-46, 2007.
Article in English | WPRIM | ID: wpr-37557

ABSTRACT

Huntington's disease is caused by CAG trinucleotide expansions in the gene encoding huntingtin. N- terminal fragments of huntingtin with polyglutamine produce aggregates in the endosome-lysosomal system, where proteolytic fragments of huntingtin is generated. Heat shock protein 70 (HSP70) prevents the formation of protein aggregates, but the effect of HSP70 on the huntingtin in the endosome-lysosomal system is unknown. This study was to determine whether HSP70 alters the distribution of huntingtin in endosome-lysosomal system. HSP70 expressing stable cells (NIH/3T3 or cerebral hybrid cell line A1) were generated, and mutant [(CAG)100] huntingtin was transiently overexpressed. Analysis of subcellular distribution by immnuocytochemistry or proteolysis cleavage by Western blotting was performed. 18 CAG repeat wild type [WT; (CAG)18] huntingtin was used as a control. Cells with huntingtin showed patterns of endosome- lysosomal accumulation, from a 'dispersed vacuole (DV)' type into a coalescent 'perinuclear vacuole (PV)' type over time. In WT huntingtin, HSP70 increased the cells with the PV types that enhanced the proteolytic cleavage of huntingtin. However, HSP70 reduced cells of the DV and PV types expressing mutant huntingtin, that result in less proteolysis than that of control. In addition, intranuclear inclusions were formed only in mutant cells, which was not affected by HSP70. These results suggest that HSP70 alters the distribution of huntingtin in the endosome-lysosomal system, and that this contributes to huntingtin proteolysis.


Subject(s)
Mice , Animals , Peptide Hydrolases/metabolism , Nuclear Proteins/genetics , Nerve Tissue Proteins/genetics , NIH 3T3 Cells , Lysosomes/metabolism , HSP70 Heat-Shock Proteins/genetics , Endosomes/metabolism , Cytoplasm/metabolism , Cell Survival
14.
Journal of the Korean Neurological Association ; : 183-190, 2003.
Article in Korean | WPRIM | ID: wpr-109684

ABSTRACT

BACKGROUND: Intracerebral hemorrhage (ICH) is associated with a considerable proportion of stroke and head injuries, but except for supportive care, there is no medical therapy available. Transplantation of human neural stem cells (NSCs) can be used to reduce behavioral deficit in experimental ischemic infarct model. However, effect of stem cell transplantation in experimental intracerebral hemorrhage (ICH) is unknown. We hypothesized that NSCs could migrate and differentiate into neurons or glial cells, and improve functional outcome in ICH. METHODS: Experimental ICH was made by intrastriatal administration of bacterial collagenase in adult rats. Animals were randomized to receive intravenously either immortalized Lac-Z positive human NSCs (5x1 06 in 500microL, n=15) or same volume of saline (n=12) on the following day. Animals were evaluated for 8 weeks after surgery with behavioral test battery. After 8 weeks, animals were sacrificed and the brains were sectioned. Transplanted NSCs were detected by X-gal histochemistry or beta-gal immunohistochemistry, and differentiation of grafted NSCs were evaluated by double labeling of GFAP, NeuN, or neurofilament. RESULTS: Transplanted NSCs migrated to the side of peri-hematomal areas, and differentiated into neurons and astrocytes. NSCs injection group showed improved performances on rotarod test after 2 weeks and on limb placing test after 5 weeks compared with control group (p<0.05) and these effect persisted up to 8 weeks. CONCLUSIONS: Intravenously injected NSCs enter rat brain with ICH, and differentiate into astrocytes or neuronal cell, which lead to functional recovery. These findings show the possibility that NSCs can be used to reduce neurological deficits in the experimental ICH.


Subject(s)
Adult , Animals , Humans , Rats , Astrocytes , Brain , Cerebral Hemorrhage , Collagenases , Craniocerebral Trauma , Extremities , Immunohistochemistry , Neural Stem Cells , Neuroglia , Neurons , Rotarod Performance Test , Stem Cell Transplantation , Stroke , Transplants
15.
Journal of the Korean Neurological Association ; : 634-640, 2002.
Article in Korean | WPRIM | ID: wpr-124509

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) is an angiogenic peptide that enhances microvascular perfusion. Recently, VEGF is known to have neurotrophic effect and rescues neurons from cell death induced by serum deprivation. To investigate the serial changes in VEGF expression and neuroprotective properties of VEGF during acute ischemia. METHODS: Human cortical-neuroblastoma hybrid cell line (A1G11), human neuroglioma cell line (H4), and human vascular endothelial cell line (ECV304) were placed in the glucose/serum free media and incubated in the hypoxic chamber (94% N2/5% CO2/1% room air) at 37 degrees C. Cell viability was determined by MTT assay. Western blot analysis was performed to detect VEGF and its receptor (VEGFR) expression. To test the protective effect of VEGF, human recombinant VEGF165 was used. RESULTS: Morphological changes and the decrease of cell viability were observed following 6 hr ischemia. In A1G11 cells, VEGF expression was not noted until 3 hr ischemia, but was induced after 6hr and continued to 12 hr and then diminished. In H4 and ECV304, the change of VEGF expression was not observed. VEGFR-2/Flk-1 expression was induced from 6 hr (peak level) to 12 hr in A1G11, and induced after 3 hr and continued to 12hr in ECV304. Administration of VEGF increased cell viability in A1G11 cells at 6 hr, 12 hr and 18 hr ischemia (p=0.009, p=0.01 p=0.013), but not in H4 or ECV304 cells ( p>0.05). CONCLUSION: Ischemia induces VEGF production in neurons and VEGF may exert a direct neuron-specific protective effect through VEGFR-2/Flk receptors during the acute phase of ischemic neuronal injury.


Subject(s)
Humans , Blotting, Western , Cell Death , Cell Line , Cell Survival , Endothelial Cells , Hybrid Cells , Ischemia , Neurons , Perfusion , Vascular Endothelial Growth Factor A
16.
Korean Journal of Pathology ; : 400-405, 2002.
Article in Korean | WPRIM | ID: wpr-164137

ABSTRACT

BACKGROUND: GM1 ganglioside-bound amyloid beta-protein (GM1/A) has been reported to be involved with senile plaque formation in Alzheimer disease. METHODS: To investigate the binding of major gangliosides on senile plaques and neurofibrillary tangles of Alzheimer disease-specific pathology, we developed four monoclonal antibodies -- GM1, GD1a, GD1b, and GT1b -- employing the hydridoma technique, and applied them for immunohistochemical staining at the frontotemporal neocortex and hippocampus of Alzheimer disease brains and age-matched control brains. RESULTS: Moderate immunopositivity for GM1 and GD1a was noted on the senile plaques and neurofibrillary tangles. Mild immunopositivity for GD1b and GT1b on neurofibrillary tangles was noted. Strong GD1b immunopositivity was observed on a few neurons and neurites. Strong immunopositivity for GT1b, and moderate immunopositivity for GM1 and GD1a were noted on reactive astrocytes. CONCLUSIONS: These observations suggest that GM1 and GD1a may be involved in the formation of senile plaques as well as neurofibrillary tangles in Alzheimer disease brains.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Antibodies, Monoclonal , Astrocytes , Brain , Gangliosides , Hippocampus , Immunohistochemistry , Neocortex , Neurites , Neurofibrillary Tangles , Neurons , Pathology , Plaque, Amyloid
17.
Korean Journal of Anatomy ; : 95-105, 2001.
Article in Korean | WPRIM | ID: wpr-646648

ABSTRACT

Parkinson's disease animal model was developed by the destruction of the striatonigral dopaminergic system. The morphological changes in the dopamine depleted striatum after the transplantation of the fetal mesencephalic dopaminergic neurons or tyrosine hydroxylase cDNA transfected human neural stem cells (C4-TH cells) were studied. Male Sprague-Dawley rats, weighting 250~300 gm, were used. To make unilateral lesion of nigrostriatal tract, 6-OHDA (6 microgram/microliter) was injected into the medial forebrain bundle. Two weeks after the lesion surgery, the effect of the 6-OHDA lesion was assessed by monitoring apomorphine (0.5 mg/kg, s.c)-induced turning behavior and confirmed by the lack of TH-immunoreactivity on tissue sections. Either cell suspension from ventral mesencephalic tissue obtained from embryonic day 14 fetus or C4-TH cells was grafted into the rostral striatum. After grafting, rats were tested with apomorphine every 2 weeks for 6 weeks. The grafted rats showing behavioral recovery were sacrificed and analysed by TH, neuropeptide Y (NPY), and parvalbumin (PV) immuno- histochemistry. TH-immunoreactive (ir) neurons were located around the graft and their processes extended into the striatum. The TH-ir axon terminals made a symmetrical synapse with the dendrites of the striatal neuron. Cell bodies either NPY- or PV-ir striatal neuron were observed around the graft and extended their processes into the graft. TH-ir C4-TH cells were also distributed along the needle track such as the transplanted fetal dopaminergic neurons, but had smaller soma and fewer processes than those. It is concluded that the grafted dopaminergic cells are survived in the dopamine depleted striatum and recovered the rotational behavior of Parkinson's disease animal model.


Subject(s)
Animals , Humans , Male , Rats , Apomorphine , Carisoprodol , Cell Transplantation , Dendrites , DNA, Complementary , Dopamine , Dopaminergic Neurons , Fetus , Medial Forebrain Bundle , Models, Animal , Needles , Neural Stem Cells , Neurons , Neuropeptide Y , Oxidopamine , Parkinson Disease , Presynaptic Terminals , Rats, Sprague-Dawley , Synapses , Transplantation , Transplants , Tyrosine 3-Monooxygenase
18.
Journal of Korean Medical Science ; : 643-648, 2001.
Article in English | WPRIM | ID: wpr-53143

ABSTRACT

This study investigated calcium/calmodulin kinase II (CaMKII) activity related to long-standing neuronal injury of the hippocampus in kainate (KA)-induced experimental temporal lobe epilepsy. Epileptic seizure was induced by injection of KA (1 g/L) dissolved in phosphate buffer (0.1 M, pH 7.4) into the left amygdala. Clinical seizures, histopathologic changes and CaMKII activity of the hippocampus were evaluated. Characteristic early limbic and late seizures were developed. Hippocampal CaMKII activity increased significantly 4 and 8 weeks after intra-amygdaloid injection of KA, when late seizures developed. The histopathologic changes of the hippocampus included swelling of neuronal cytoplasm with nuclear pyknosis and loss of neurons in CA3 during this period. The increased activity of CaMKII may correlate with appearance of distant damage in the hippocampus. The above results indicate that intra-amygdaloid injection of KA produces excitatory signals for ipsilateral CA3 neurons in the hippocampus and that subsequently increased levels of CaMKII in postsynaptic neurons induce neuronal injury via phosphorylation of N-methyl-D-aspartate type glutamate receptor.


Subject(s)
Male , Rats , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Epilepsy, Temporal Lobe/chemically induced , Hippocampus/enzymology , Kainic Acid/toxicity , Long-Term Potentiation/drug effects , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL