Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Mol Ther ; 25(9): 2028-2037, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28705346

ABSTRACT

Generation of functional dopamine (DA) neurons is an essential step for the development of effective cell therapy for Parkinson's disease (PD). The generation of DA neurons can be accomplished by overexpression of DA-inducible genes using virus- or DNA-based gene delivery methods. However, these gene delivery methods often cause chromosomal anomalies. In contrast, mRNA-based gene delivery avoids this problem and therefore is considered safe to use in the development of cell-based therapy. Thus, we used mRNA-based gene delivery method to generate safe DA neurons. In this study, we generated transformation-free DA neurons by transfection of mRNA encoding DA-inducible genes Nurr1 and FoxA2. The delivery of mRNA encoding dopaminergic fate inducing genes proved sufficient to induce naive rat forebrain precursor cells to differentiate into neurons exhibiting the biochemical, electrophysiological, and functional properties of DA neurons in vitro. Additionally, the generation efficiency of DA neurons was improved by the addition of small molecules, db-cAMP, and the adjustment of transfection timing. The successful generation of DA neurons using an mRNA-based method offers the possibility of developing clinical-grade cell sources for neuronal cell replacement treatment for PD.


Subject(s)
Dopaminergic Neurons/metabolism , RNA, Messenger/chemical synthesis , RNA, Messenger/genetics , Transcription Factors/genetics , Animals , Cell Line , Dopaminergic Neurons/cytology , Gene Expression , Gene Expression Regulation , Gene Order , Genes, Reporter , Genetic Vectors/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Rats , Transfection , Tyrosine 3-Monooxygenase/genetics
2.
Molecules ; 23(4)2018 Mar 22.
Article in English | MEDLINE | ID: mdl-29565818

ABSTRACT

Otitis media (OM) is a group of inflammatory diseases of the middle ear (ME), regardless of cause or pathological mechanism. Among the molecular biological studies assessing the pathology of OM are investigations into the expression of C-type lectin receptors (CLR) in the ME and Eustachian tube (ET). To date, nine studies have evaluated CLR expression in the ME and ET. The expression of individual CLRs in mammalian ME and ET varies by species and model of OM. Assessments have shown that the patterns of CLR expression in the ME and ET vary; that CLR expression may vary by type of OM; and that the distribution and levels of expression of CLRs may depend on the presence or absence of inflammation, with variations even within the same species and same tissue. Infection of the ME and ET with various pathogens is a common cause of all types of OM, with host responses to pathogens mediated initially by the innate immune system. CLRs are important factors in the innate immune system because they act as both adhesion molecules and as pathogen recognition receptors. The expression of CLRs in OM tissues suggests that CLRs are associated with the pathogenesis of various types of OM.


Subject(s)
Ear, Middle/metabolism , Eustachian Tube/metabolism , Lectins, C-Type/metabolism , Otitis Media/metabolism , Animals , Humans
3.
Bioeng Transl Med ; 8(2): e10381, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36925687

ABSTRACT

Antibiotic resistance ranks among the top threats to humanity. Due to the frequent use of antibiotics, society is facing a high prevalence of multidrug resistant pathogens, which have managed to evolve mechanisms that help them evade the last line of therapeutics. An alternative to antibiotics could involve the use of bacteriophages (phages), which are the natural predators of bacterial cells. In earlier times, phages were implemented as therapeutic agents for a century but were mainly replaced with antibiotics, and considering the menace of antimicrobial resistance, it might again become of interest due to the increasing threat of antibiotic resistance among pathogens. The current understanding of phage biology and clustered regularly interspaced short palindromic repeats (CRISPR) assisted phage genome engineering techniques have facilitated to generate phage variants with unique therapeutic values. In this review, we briefly explain strategies to engineer bacteriophages. Next, we highlight the literature supporting CRISPR-Cas9-assisted phage engineering for effective and more specific targeting of bacterial pathogens. Lastly, we discuss techniques that either help to increase the fitness, specificity, or lytic ability of bacteriophages to control an infection.

4.
Biotechnol J ; 17(7): e2100468, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35157790

ABSTRACT

BACKGROUND: The recent emergence of gene editing using Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated system (Cas) tools and advances in genomics and proteomics has revolutionized drug discovery and personalized medicine. PURPOSE AND SCOPE: The CRISPR-Cas system has enabled gene and cell-based therapies, screening for novel drug targets, a new generation of disease models, elucidation of drug resistance mechanisms, and drug efficacy testing. Here, we summarized recent investigations and strategies involved in cancer-related drug discovery using the CRISPR-Cas system. CONCLUSION: CRISPR-Cas-mediated gene editing has shown great potential in the development of next generation drugs for treatment of Mendelian disorders and various cancer types. In this review, we focused on the impact of the CRISPR-Cas system in drug discovery and its application to biomarker identification and validation, high-end target genes, and breakthrough anticancer cell therapies. We also highlighted the role of CRISPR-Cas in precision disease modeling and functional drug screening.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CRISPR-Cas Systems/genetics , Drug Discovery , Gene Editing , Genomics , Neoplasms/drug therapy , Neoplasms/genetics
5.
Mol Cancer Res ; 7(3): 371-82, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19276188

ABSTRACT

Functional suppression of spindle checkpoint protein activity results in apoptotic cell death arising from mitotic failure, including defective spindle formation, chromosome missegregation, and premature mitotic exit. The recently identified p31(comet) protein acts as a spindle checkpoint silencer via communication with the transient Mad2 complex. In the present study, we found that p31(comet) overexpression led to two distinct phenotypic changes, cellular apoptosis and senescence. Because of a paucity of direct molecular link of spindle checkpoint to cellular senescence, however, the present report focuses on the relationship between abnormal spindle checkpoint formation and p31(comet)-induced senescence by using susceptible tumor cell lines. p31(comet)-induced senescence was accompanied by mitotic catastrophe with massive nuclear and chromosomal abnormalities. The progression of the senescence was completely inhibited by the depletion of p21(Waf1/Cip1) and partly inhibited by the depletion of the tumor suppressor protein p53. Notably, p21(Waf1/Cip1) depletion caused a dramatic phenotypic conversion of p31(comet)-induced senescence into cell death through mitotic catastrophe, indicating that p21(Waf1/Cip1) is a major mediator of p31(comet)-induced cellular senescence. In contrast to wild-type p31(comet), overexpression of a p31 mutant lacking the Mad2 binding region did not cause senescence. Moreover, depletion of Mad2 by small interfering RNA induced senescence. Here, we show that p31(comet) induces tumor cell senescence by mediating p21(Waf1/Cip1) accumulation and Mad2 disruption and that these effects are dependent on a direct interaction of p31(comet) with Mad2. Our results could be used to control tumor growth.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/metabolism , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Nuclear Proteins/biosynthesis , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis/drug effects , Apoptosis/genetics , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Clone Cells , DNA/metabolism , Humans , Mad2 Proteins , Mitosis/physiology , Nuclear Proteins/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/biosynthesis
6.
J Neurosci Res ; 88(11): 2494-503, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20623531

ABSTRACT

Mutations in the superoxide dismutase 1 (SOD1) gene are linked to glutamate excitotoxicity in familial amyotrophic lateral sclerosis (fALS), but the underlying mechanism remains unclear. We investigated whether nuclear factor-kappaB (NF-kappaB) activation is involved in glutamate excitotoxicity by using motor neuron-neuroblastoma hybrid cells that expressed a mutant (G93A) SOD1 (mtSOD1) or wild-type SOD1 (wtSOD1). MtSOD1 cells were more vulnerable to glutamate excitotoxicity than wtSOD1 cells and showed higher NF-kappaB activity, higher nuclear cRel expression, and lower nuclear RelA expression under basal conditions. Glutamate treatment increased NF-kappaB activation along with nuclear expressions of RelA and cRel in wtSOD1 cells but induced only weak nuclear RelA expression in mtSOD1 cells. Suppression of NF-kappaB activation using transfection of the superrepressive mutant form of IkappaBalpha (mIkappaBalpha) inhibited nuclear RelA expression in both types of SOD1 cells, which increased glutamate excitotoxicity in wtSOD1 cells but not in mtSOD1 cells. Furthermore, immunohistochemistry confirmed stronger RelA immunoreactivity in the nuclei of motor neurons of spinal cord in wild-type SOD1 transgenic mice than in those in SOD1 G93A transgenic mice. In addition, we found that glutamate treatment decreased XIAP expression and increased caspase-3 activity in mtSOD1 cells and mIkappaBalpha-overexpressing wtSOD1 cells. Our results suggest that glutamate excitotoxicity in motor neurons of SOD1-linked fALS is attributable, at least in part, to the impairment of IkappaBalpha-dependent RelA activation and subsequent apoptosis mediated by XIAP inhibition and caspase-3 activation.


Subject(s)
Glutamic Acid/toxicity , Motor Neurons/metabolism , NF-kappa B/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Animals , Biotransformation/physiology , Caspase 3/metabolism , Cell Death , Cell Line , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cell Survival/drug effects , Cytoplasm/enzymology , Cytoplasm/metabolism , Fluorescent Dyes , Genetic Vectors , Humans , Hybrid Cells , Immunohistochemistry , Indoles , Luciferases/metabolism , Mice , Mice, Transgenic , Motor Neurons/drug effects , Mutation/genetics , Mutation/physiology , Neuroblastoma/metabolism , Receptors, Glutamate/metabolism , Retroviridae/genetics
7.
Metabolites ; 10(11)2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33114596

ABSTRACT

Although many studies have been conducted on leukemia, only a few have analyzed the metabolomic profiles of various leukemic cells. In this study, the metabolomes of THP-1, U937, KG-1 (acute myelogenous leukemia, AML), K562 (chronic myelogenous leukemia, CML), and cord blood-derived CD34-positive hematopoietic stem cells (HSC) were analyzed using gas chromatography-mass spectrometry, and specific metabolic alterations were found using multivariate statistical analysis. Compared to HSCs, leukemia cell metabolomes were found to have significant alterations, among which three were related to amino acids, three to sugars, and five to fatty acids. Compared to CML, four metabolomes were observed specifically in AML. Given that overall more metabolites are present in leukemia cells than in HSCs, we observed that the activation of glycolysis and oxidative phosphorylation (OXPHOS) metabolism facilitated the incidence of leukemia and the proliferation of leukemic cells. Analysis of metabolome profiles specifically present in HSCs and leukemia cells greatly increases our basic understanding of cellular metabolic characteristics, which is valuable fundamental knowledge for developing novel anticancer drugs targeting leukemia metabolism.

8.
Mol Ther Nucleic Acids ; 17: 245-255, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31272074

ABSTRACT

Glioblastoma multiforme (GBM), a particularly aggressive type of malignant brain tumor, has a high mortality rate. Bcl-w, an oncogene, is reported to enhance cell survival, proliferation, epithelial-mesenchymal transition (EMT), migratory and invasive abilities, and stemness maintenance in a variety of cancer cell types, including GBM. In this study, we confirmed that Bcl-w-induced conditional medium (CM) enhances tumorigenic phenotypes of migration, invasiveness, and stemness maintenance. Notably, platelet-derived growth factor-A (PDGF-A) expression, among other factors of the tumor environment, was increased by CM of Bcl-w-overexpressing cells, prompting investigation of the potential correlation between Bcl-w and PDGF-A and their effects on GBM malignancy. Bcl-w and PDGF-A levels were positively regulated and increased tumorigenicity by Sox2 activation in GBM cells. miR-340-5p was further identified as a direct inhibitor of Bcl-w and Sox2. Overexpression of miR-340-5p reduced mesenchymal traits, cell migration, invasion, and stemness in GBM through attenuating Bcl-w and Sox2 expression. Our novel findings highlight the potential utility of miR-340-5p as a therapeutic agent for glioblastoma multiforme through inhibitory effects on Bcl-w-induced PDGF-A and Sox2 activation.

9.
Sci Rep ; 9(1): 13659, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31541140

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive movement disturbances caused by the selective loss of dopamine (DA) neurons in the substantia nigra. Despite the identification of the causal mechanisms underlying the pathogenesis of PD, effective treatments remain elusive. In this study, we observed that a low level of fetal bovine serum (FBS) effectively induced DA neurons in rat neural precursor cells (NPCs) by enhancing nuclear receptor-related 1 protein (NURR1) expression. Among the various components of FBS, the thyroid hormones triiodothyronine (T3) and thyroxine (T4) were identified as key factors for the induction of DA neurons. Since an overdose of thyroid hormones can cause hyperthyroidism, we synthesized several thyroid hormone derivatives that can partially activate thyroid hormone receptors and induce the complete differentiation of NPCs into DA neurons. Two derivatives (#3 and #9) showed positive effects on the induction and maturation of DA neurons without showing significant affinity for the thyroid hormone receptor. They also effectively protected and restored DA neurons from neurotoxic insults. Taken together, these observations demonstrate that thyroid hormone derivatives can strongly induce DA neuron differentiation while avoiding excessive thyroid stimulation and might therefore be useful candidates for PD treatment.


Subject(s)
Dopaminergic Neurons/cytology , Neural Stem Cells/cytology , Thyroid Hormones/chemical synthesis , Thyroxine/pharmacology , Triiodothyronine/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Culture Media/chemistry , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, Thyroid Hormone/metabolism , Thyroid Hormones/chemistry , Thyroid Hormones/pharmacology
10.
Biomol Ther (Seoul) ; 27(1): 78-84, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30458601

ABSTRACT

Cell therapeutic agents for treating degenerative brain diseases using neural stem cells are actively being developed. However, few systems have been developed to monitor in real time whether the transplanted neural stem cells are actually differentiated into neurons. Therefore, it is necessary to develop a technology capable of specifically monitoring neuronal differentiation in vivo. In this study, we established a system that expresses cell membrane-targeting red fluorescent protein under control of the Synapsin promoter in order to specifically monitor differentiation from neural stem cells into neurons. In order to overcome the weak expression level of the tissue-specific promoter system, the partial 5' UTR sequence of Creb was added for efficient expression of the cell surface-specific antigen. This system was able to track functional neuronal differentiation of neural stem cells transplanted in vivo, which will help improve stem cell therapies.

11.
Carcinogenesis ; 29(1): 44-51, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17984117

ABSTRACT

Underlying mechanisms involved in the activation of hypoxia-inducible factor-1 (HIF-1) in cancer cells are diverse and cell type specific. Although both HIF-1alpha and AKT (protein kinase B) have been implicated in gastric tumor promotion and angiogenesis, it remains unclear whether HIF-1 mediates the role of AKT in terms of promoting vascular endothelial growth factor (VEGF) expression. The present study was performed to investigate the correlation between HIF-1alpha activation and AKT activation in gastric cancer using human gastric cancer specimens, in vitro cell experiments and in vivo animal experiments. Immunohistochemistry performed on tissue array slides containing 268 surgical specimens of gastric carcinomas showed immunoreactivity for HIF-1alpha in 29% of samples. Moreover, HIF-1alpha was positively associated with phosphorylated AKT (pAKT) (P = 0.002) or VEGF (P = 0.002), and the immunoreactivities of pAKT and VEGF were positively correlated (P < 0.001). Western blot analysis and reverse transcription-polymerase chain reaction in cell experiments revealed that the over-expression of constitutively active AKT (CA-AKT) promotes the expressions of HIF-1alpha protein and VEGF messenger ribonucleic acid in Seoul national university (SNU)-216 and SNU-668 gastric cancer cells under normoxic conditions, whereas kinase-dead mutant of AKT down-regulated these expressions under the same conditions. Xenografts in nude mice derived from stable gastric cancer cells over-expressing CA-AKT showed higher tumor incidence, larger tumor volumes, higher microvessel density and stronger HIF-1alpha immunoreactivity than those derived from vector control cells. Thus, we propose that the hypoxia-independent promotion of the AKT-HIF-1alpha-VEGF pathway contributes, at least in part, to gastric cancer tumorigenesis and angiogenesis.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Neovascularization, Pathologic , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/blood supply , Up-Regulation , Animals , Base Sequence , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , DNA Primers , Enzyme Activation , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Phosphorylation , RNA, Messenger/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/metabolism , Vascular Endothelial Growth Factor A/genetics
12.
Mol Cancer Ther ; 6(1): 82-92, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17237268

ABSTRACT

Resistance to anticancer drugs can sometimes be overcome by combination treatment with other therapeutic drugs. Here, we showed that phytosphingosine treatment in combination with arsenic trioxide (As(2)O(3)) enhanced cell death of naturally As(2)O(3)-resistant human myeloid leukemia cells. The combination treatment induced an increase in intracellular reactive oxygen species level, mitochondrial relocalization of Bax, poly(ADP-ribose) polymerase-1 (PARP-1) activation, and cytochrome c release from the mitochondria. N-acetyl-l-cysteine, a thiol-containing antioxidant, completely blocked Bax relocalization, PARP-1 activation, and cytochrome c release. Pretreatment of 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone, a PARP-1 inhibitor, or PARP-1/small interfering RNA partially attenuated cytochrome c release, whereas the same treatment did not affect Bax relocalization. The combination treatment induced selective activation of p38 mitogen-activated protein kinase (MAPK). Inhibition of p38 MAPK by treatment of SB203580 or expression of dominant-negative forms of p38 MAPK suppressed the combination treatment-induced Bax relocalization but did not affect PARP-1 activation. In addition, antioxidant N-acetyl-l-cysteine completely blocked p38 MAPK activation. These results indicate that phytosphingosine in combination with As(2)O(3) induces synergistic apoptosis in As(2)O(3)-resistant leukemia cells through the p38 MAPK-mediated mitochondrial translocation of Bax and the PARP-1 activation, and that p38 MAPK and PARP-1 activations are reactive oxygen species dependent. The molecular mechanism that we elucidated in this study may provide insight into the design of future combination cancer therapies to cells intrinsically less sensitive to As(2)O(3) treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Arsenicals/pharmacology , Drug Resistance, Neoplasm , Neoplasms/pathology , Oxides/pharmacology , Sphingosine/analogs & derivatives , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Arsenic Trioxide , Arsenicals/administration & dosage , Arsenicals/therapeutic use , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Humans , Mitochondria/drug effects , Models, Biological , Neoplasms/drug therapy , Oxides/administration & dosage , Oxides/therapeutic use , Poly(ADP-ribose) Polymerases/metabolism , Protein Transport/drug effects , Reactive Oxygen Species/metabolism , Sphingosine/administration & dosage , Sphingosine/pharmacology , Sphingosine/therapeutic use , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Oncol Rep ; 17(1): 175-84, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17143496

ABSTRACT

Sensitization of cancer cells to TRAIL could improve the effectiveness of TRAIL as an anticancer agent. We explored whether TRAIL in combination with phytosphingosine could sensitize cancer cells to TRAIL. The combined treatment enhanced synergistic apoptotic cell death of Jurkat T cells, compared to TRAIL or phytosphingosine alone. Enhanced apoptosis in response to the combination treatment was associated with caspase-8 activation-mediated Bax and Bak activation and mitochondrial dysfunction. The combination treatment also resulted in synergistic up-regulation of TRAIL receptor R1 (DR4) and R2 (DR5). siRNA targeting of DR5 significantly attenuated the combination treatment-induced caspase-8 activation, mitochondrial dysfunction, and apoptotic cell death. Upon stimulation of cells with the combination treatment, NF-kappaB was activated. Moreover, siRNA targeting of NF-kappaB significantly attenuated the combination treatment-induced DR4 and DR5 expression and receptor-mediated caspase-8 activation. These results indicate that phytosphingosine sensitizes cancer cells to TRAIL through the synergistic up-regulation of DR4 and DR5 in an NF-kappaB-dependent fashion resulting in caspase-8 activation and subsequent mitochondrial dysfunction. These findings support the potential application of combination treatment with TRAIL and phytosphingosine in the treatment of cancers that are less sensitive to TRAIL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Lymphoma, T-Cell/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , Sphingosine/analogs & derivatives , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 8/metabolism , Drug Synergism , Enzyme Activation/drug effects , Humans , Jurkat Cells , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Mitochondria/drug effects , Mitochondria/physiology , NF-kappa B/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Sphingosine/administration & dosage , Sphingosine/pharmacology , T-Lymphocytes/drug effects , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Up-Regulation/drug effects , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
14.
Oncogene ; 24(23): 3715-25, 2005 May 26.
Article in English | MEDLINE | ID: mdl-15806174

ABSTRACT

Since radiation-induced caspase-dependent apoptosis and ROS generation were partially prevented by HSP25 overexpression, similar to the treatment of control cells with antioxidant agents such as DPI and tiron, questions arise whether radiation-mediated ROS generation contributes to the apoptotic cell death, and also whether HSP25 overexpression can reduce ROS mediated apoptotic cell death. In the present study, radiation-induced cytochrome c release from mitochondria and activation of caspases accompanied by a decrease of mitochondrial membrane potential in Jurkat T cells were shown to be inhibited by mitochondrial complex I inhibitor rotenone, suggesting that mitochondrial ROS might be important in radiation-induced caspase-dependent apoptosis. When HSP25 was overexpressed, effects similar to the treatment of cells with the antioxidants were obtained, indicating that HSP25 suppressed radiation-induced mitochondrial alteration that resulted in apoptosis. Furthermore, activation of p38 MAP kinase by radiation was associated with radiation-induced cell death and ROS production and PKCdelta was an upstream molecule for p38 MAP kinase activation, ROS generation and subsequent caspase-dependent apoptotic events. However, in the HSP25 overexpressed cells, the above-described effects were blocked. In fact, radiation-induced membrane translocation of PKCdelta and tyrosine phosphorylation were inhibited by HSP25. Based on the above data, we suggest that HSP25 downregulates PKCdelta, which is a key molecule for radiation-induced ROS generation and mitochondrial-mediated caspase-dependent apoptotic events.


Subject(s)
Apoptosis/radiation effects , Heat-Shock Proteins/physiology , Neoplasm Proteins/physiology , Protein Kinase C/physiology , Reactive Oxygen Species/metabolism , Enzyme Activation , HSP27 Heat-Shock Proteins , Humans , Jurkat Cells , Mitochondria/physiology , Molecular Chaperones , Phosphorylation , Protein Kinase C-delta , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Invest Ophthalmol Vis Sci ; 47(3): 892-900, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16505021

ABSTRACT

PURPOSE: Side population (SP) cells are known to reside in the limbus as putative corneal epithelial stem cells. This study was performed to demonstrate the presence and the characteristics of SP cells in the rabbit limbal epithelium and explore their sensitivity in response to the central cornea wounding. METHODS: To sort out the SP cells, freshly isolated rabbit limbal and central corneal epithelial cells were subjected to Hoechst 33342 dye efflux assay. For characterization of the sorted SP cells, RT-PCR analysis, semi-dry three-dimensional (3-D) cell culture, and transplantation in nude mice were performed. To explore wound sensitivity of the limbal SP cells, the rabbit central cornea was wounded by direct contact of a 6-mm paper disk soaked with 1 N NaOH, and changes in the population size of the SP cells and the colony-forming efficiency (CFE) was monitored on days 1, 2, and 5 after wounding. RESULTS: The SP cells were present in the rabbit limbal epithelium with an incidence of 0.73% +/- 0.14% (n = 8) and were smaller in cell size than the major population (MP) cells, quiescent in the cell cycle, and in the undifferentiated state. The SP cells were able to regenerate the cornea-like structure with basal enrichment of p63-positive cells by in vitro 3-D culture and in vivo transplantation, all of which were best achieved by the whole population (WP) of cells comprising SP and MP cells. After central cornea wounding, this rare population of the limbal SP cells increased in size fivefold on day 1 and then decreased on day 2. The transient increase in the SP cells was subsequently followed by the propagation of an increase in CFE in the limbal MP cells on day 2 and then in the corneal MP cells on day 5. In the hematopoietic colony-forming assay, the limbal SP cells gave approximately eightfold higher CFU than the limbal MP cells. CONCLUSIONS: The SP cells identified in the rabbit limbus are an undifferentiated and noncycling rare epithelial cell population, which sensitively respond to the central cornea wounding by their transient increase in the population size.


Subject(s)
Burns, Chemical/metabolism , Epithelium, Corneal/cytology , Eye Burns/chemically induced , Limbus Corneae/cytology , Stem Cells/physiology , Animals , Benzimidazoles/pharmacology , Cell Culture Techniques , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Colony-Forming Units Assay , Epithelium, Corneal/drug effects , Eye Burns/metabolism , Fluorescent Dyes/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Sodium Hydroxide , Stem Cell Transplantation
16.
Clin Cancer Res ; 11(7): 2518-25, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15814628

ABSTRACT

PURPOSE: Because the biological significance of constitutive nuclear factor-kappaB (NF-kappaB) activation in human gastric cancer is unclear, we undertook this study to clarify the regulatory mechanism of NF-kappaB activation and its clinical significance. EXPERIMENTAL DESIGN: Immunohistochemistry for NF-kappaB/RelA was done on 290 human gastric carcinoma specimens placed on tissue array slides. The correlations between NF-kappaB activation and clinicopathologic features, prognosis, Akt activation, tumor suppressor gene expression, or Bcl-2 expression were analyzed. We also did luciferase reporter assay, Western blot analysis, and reverse transcription-PCR using the SNU-216 human gastric cancer cell line transduced with retroviral vectors containing constitutively active Akt or the NF-kappaB repressor mutant of IkappaBalpha. RESULTS: Nuclear expression of RelA was found in 18% of the gastric carcinomas and was higher in early-stage pathologic tumor-node-metastasis (P = 0.019). A negative correlation was observed between NF-kappaB activation and lymphatic invasion (P = 0.034) and a positive correlation between NF-kappaB activation and overall survival rate of gastric cancer patients (P = 0.0228). In addition, NF-kappaB activation was positively correlated with pAkt (P = 0.047), p16 (P = 0.004), adenomatous polyposis coli (P < 0.001), Smad4 (P = 0.002), and kangai 1 (P < 0.001) expression. An in vitro study showed that NF-kappaB activity in gastric cancer cells is controlled by and controls Akt. CONCLUSIONS: NF-kappaB activation was frequently observed in early-stage gastric carcinoma and was significantly correlated with better prognosis and Akt activation. These findings suggest that NF-kappaB activation is a valuable prognostic variable in gastric carcinoma.


Subject(s)
NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Stomach Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Androstadienes/pharmacology , Blotting, Western , Cell Line, Tumor , Child , Child, Preschool , Chromones/pharmacology , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic/drug effects , Genetic Vectors/genetics , Humans , Immunohistochemistry , Infant , Infant, Newborn , Luciferases/genetics , Luciferases/metabolism , Male , Middle Aged , Morpholines/pharmacology , NF-kappa B/analysis , NF-kappa B/genetics , Neoplasm Staging , Prognosis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Analysis , Tissue Array Analysis , Transcription Factor RelA , Transfection , Tumor Suppressor Proteins/analysis , Wortmannin
17.
J Radiat Res ; 47(1): 83-90, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16571921

ABSTRACT

Thermoresistant (TR) clone of radiation-induced fibrosarcoma (RIF) cells have been reported to show adaptive response to 1 cGy of low dose radiation, and hsp25 and inducible hsp70 are involved in this process. In the present study, to further elucidate the mechanism of how hsp25 and inducible hsp70 regulate the adaptive response, hsp25 or inducible hsp70 overexpressed RIF cells were irradiated with 1 cGy and cell cycle was analyzed. Hsp25 or inducible hsp70 overexpressed cells as well as TR cells showed increase of G1 phase population after gamma-irradiation at 1 cGy, while the parent RIF cells did not. [3H]-Thymidine and BrdU incorporation also indicated that both hsp25 and inducible hsp70 were involved in G1 arrest after 1 cGy irradiation. Molecular analysis revealed upregulation of p27Cip/Kip protein in hsp25 and inducible hsp70 overexpressed cells, and cotransfection of p27Cip/Kip antisense abolished the induction of adaptive response and 1 cGy-mediated G1 arrest. The above results indicate that induction of adaptive response by hsp25 and inducible hsp70 is mediated by upregulation of p27Cip/Kip protein, resulting in low dose radiation-induced G1 arrest.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Adaptation, Physiological/drug effects , Adaptation, Physiological/physiology , Animals , Cell Cycle/radiation effects , Cell Line, Tumor , Dose-Response Relationship, Radiation , Mice , Molecular Chaperones , Radiation Dosage
18.
Cancer Res ; 64(24): 8960-7, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15604259

ABSTRACT

Although mechanisms of arsenic trioxide (As(2)O(3))-induced cell death have been studied extensively in hematologic cancers, those in solid cancers have yet to be clearly defined. In this study, we showed that the translocation of apoptosis-inducing factor (AIF) from mitochondria to the nucleus is required for As(2)O(3)-induced cell death in human cervical cancer cells. We also showed that reactive oxygen species (ROS)-mediated poly(ADP-ribose) polymerase-1 (PARP-1) activation is necessary for AIF release from mitochondria. The treatment of human cervical cancer cells with As(2)O(3) induces dissipation of mitochondrial membrane potential (Deltapsi(m)), translocation of AIF from mitochondria to the nucleus, and subsequent cell death. Small interfering RNA targeting of AIF effectively protects cervical cancer cells against As(2)O(3)-induced cell death. As(2)O(3) also induces an increase of intracellular ROS level and a marked activation of PARP-1. N-acetyl-l-cystein, a thiol-containing antioxidant, completely blocks As(2)O(3)-induced PARP-1 activation, Deltapsi(m) loss, nuclear translocation of AIF from mitochondria, and the consequent cell death. Furthermore, pretreatment of 1,5-dihydroxyisoquinoline or 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone, PARP-1 inhibitors, effectively attenuates the loss of Deltapsi(m), AIF release, and cell death. These data support a notion that ROS-mediated PARP-1 activation signals AIF release from mitochondria, resulting in activation of a caspase-independent pathway of cell death in solid tumor cells by As(2)O(3) treatment.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Caspases/physiology , Chlorides/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Uterine Cervical Neoplasms/pathology , Acetylcysteine/pharmacology , Apoptosis/physiology , Apoptosis Inducing Factor , Arsenicals/antagonists & inhibitors , Cell Line, Tumor , Cell Nucleus/metabolism , Chlorides/antagonists & inhibitors , Enzyme Activation , Female , Flavoproteins/genetics , Flavoproteins/metabolism , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Poly(ADP-ribose) Polymerase Inhibitors , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism
19.
Oncogene ; 23(1): 9-20, 2004 Jan 08.
Article in English | MEDLINE | ID: mdl-14712206

ABSTRACT

It has been well known that Ras signaling is involved in various cellular processes, including proliferation, differentiation, and apoptosis. However, distinct cellular functions of Ras isozymes are not fully understood. Here we show the opposing roles of Ha-Ras and Ki-Ras genes in the modulation of cell sensitivity to ionizing radiation. Overexpression of active isoform of Ha-Ras (12V-Ha-Ras) in Rat2 cells increases resistance to the ionizing radiation. Constitutive activation of phosphoinositide-3-kinase (PI3K) and Akt is detected specifically in 12V-Ha-Ras-overexpressing cells. The specific PI3K inhibitor LY294002 inhibits PI3K/Akt signaling and potentiates the radiation-induced apoptosis, suggesting that activation of the PI3K/Akt signaling pathway is involved in the increased radio-resistance in cells overexpressing 12V-Ha-Ras. Overexpression of activated Ki-Ras (12V-Ki-Ras), on the other hand, markedly increases radiation sensitivity. The p38 mitogen-activated protein kinase (MAPK) activity is selectively enhanced by ionizing radiation in cells overexpressing 12V-Ki-Ras. The specific p38 MAPK inhibitor, PD169316, or dominant-negative p38 MAPK decreases radiation-induced cell death. We further show that the mechanism that underlies potentiation of cell death in cells overexpressing 12V-Ki-Ras involves Bax translocation to the mitochondrial membrane. Elevated Bax translocation following ionizing irradiation in 12V-Ki-Ras-overexpressing cells is completely inhibited by PD169316 or dominant-negative p38 MAPK. In addition, introduction of cells with RacN17, a dominant-negative mutant of Rac, resulted in a marked inhibition of radiation-induced Bax translocation and apoptotic cell death as well as p38 MAPK activation. Taken together, these findings explain the opposite effects of Ha-Ras and Ki-Ras on modulation of radiosensitivity, and suggest that differential activation of PI3K/Akt and Rac/p38 MAPK signaling by Ha-Ras and Ki-Ras may account for the opposing response to the ionizing radiation. These data provide an explanation for the diverse biological functions of Ras isozymes, and partly accounts for the differential response of transformed cells to anticancer treatments.


Subject(s)
Apoptosis/radiation effects , Mitogen-Activated Protein Kinases/physiology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins p21(ras)/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , rac GTP-Binding Proteins/physiology , Animals , Caspase 3 , Caspases/metabolism , Cell Line , Cytochromes c/metabolism , Enzyme Activation , Mice , Protein Transport , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Radiation Tolerance , Rats , bcl-2-Associated X Protein , p38 Mitogen-Activated Protein Kinases
20.
Exp Mol Med ; 37(6): 601-7, 2005 Dec 31.
Article in English | MEDLINE | ID: mdl-16391521

ABSTRACT

Nanog, a homeodomain (HD) transcription factor, plays a critical role in the maintenance of embryonic stem (ES) cell self-renewal. Here, we report the identification of an alternatively-spliced variant of nanog. This variant lacked a stretch of amino acids (residues 168-183) located between the HD and tryptophan-repeat (WR) of the previously-reported full length sequence, suggesting that the deleted sequence functions as a linker and possibly affects the flexibility of the C-terminal transactivation domain relative to the DNA binding domain. Expression of mRNA encoding the splice variant, designated as nanog-delta 48, was much lower than that of the full length version in human ES cells. The ratio of nanog-delta 48 transcript to full length transcript increased, however, in multipotent adult progenitor cells. EMSA analysis revealed that both forms of Nanog were able to bind a nanog binding sequence with roughly the same affinity. A reporter plasmid assay also showed that both variants of nanog modestly repressed transactivation of gata-4, whose expression is proposed to be inhibited by nanog, with comparable potency. We conclude that, despite the difference in primary structure and expression pattern in various stem cells, the alternatively-spliced variant of Nanog has similar activity to that of the full length version.


Subject(s)
Alternative Splicing/genetics , DNA-Binding Proteins/genetics , Exons/genetics , Homeodomain Proteins/genetics , Amino Acid Sequence , Base Sequence , Cell Nucleus , Cells, Cultured , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , GATA4 Transcription Factor/metabolism , Gene Expression Profiling , Genes, Reporter , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Humans , Introns/genetics , Molecular Sequence Data , Nanog Homeobox Protein , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL