Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
PLoS One ; 8(1): e53312, 2013.
Article in English | MEDLINE | ID: mdl-23326413

ABSTRACT

BACKGROUND: Cartilage degradation is a typical characteristic of arthritis. This study examined whether there was a subset of phagocytic chondrocytes that expressed the specific macrophage marker, CD163, and investigated their role in cartilage degradation. METHODS: Cartilage from the knee and temporomandibular joints of Sprague-Dawley rats was harvested. Cartilage degradation was experimentally-induced in rat temporomandibular joints, using published biomechanical dental methods. The expression levels of CD163 and inflammatory factors within cartilage, and the ability of CD163(+) chondrocytes to conduct phagocytosis were investigated. Cartilage from the knees of patients with osteoarthritis and normal cartilage from knee amputations was also investigated. RESULTS: In the experimentally-induced degrading cartilage from temporomandibular joints, phagocytes were capable of engulfing neighboring apoptotic and necrotic cells, and the levels of CD163, TNF-α and MMPs were all increased (P<0.05). However, the levels of ACP-1, NO and ROS, which relate to cellular digestion capability were unchanged (P>0.05). CD163(+) chondrocytes were found in the cartilage mid-zone of temporomandibular joints and knee from healthy, three-week old rats. Furthermore, an increased number of CD163(+) chondrocytes with enhanced phagocytic activity were present in Col-II(+) chondrocytes isolated from the degraded cartilage of temporomandibular joints in the eight-week experimental group compared with their age-matched controls. Increased number with enhanced phagocytic activity of CD163(+) chondrocytes were also found in isolated Col-II(+) chondrocytes stimulated with TNF-α (P<0.05). Mid-zone distribution of CD163(+) cells accompanied with increased expression of CD163 and TNF-α were further confirmed in the isolated Col-II(+) chondrocytes from the knee cartilage of human patients with osteoarthritis, in contrast to the controls (both P<0.05). CONCLUSIONS: An increased number of CD163(+) chondrocytes with enhanced phagocytic activity were discovered within degraded joint cartilage, indicating a role in eliminating degraded tissues. Targeting these cells provides a new strategy for the treatment of arthritis.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Phagocytosis , Receptors, Cell Surface/metabolism , Adult , Aged , Animals , Cartilage, Articular/metabolism , Cell Count , Cell Movement/drug effects , Cell Separation , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/pathology , Female , Humans , Knee Joint/drug effects , Knee Joint/metabolism , Knee Joint/pathology , Male , Middle Aged , Osteoarthritis/metabolism , Osteoarthritis/pathology , Phagocytosis/drug effects , Rats , Rats, Sprague-Dawley , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
2.
Exp Ther Med ; 3(5): 776-780, 2012 May.
Article in English | MEDLINE | ID: mdl-22969967

ABSTRACT

In order to investigate the effect and mechanisms of interferon (IFN)-γ in combination with all-trans-retinoic acid (ATRA) on NB4 cells [ATRA-sensitive acute promyelocytic leukemia (APL) cell line] and NB4-R1 cells (ATRA-resistant APL cell line) and to search for a novel approach to solve the problem of ATRA resistance in APL, we initially treated NB4 and NB4-R1 cells with IFN-γ, ATRA and IFN-γ in combination with ATRA, respectively. The cell proliferation was then tested by MTT assay, and the cell differentiation was tested through light microscopy, by NBT test and flow cytometry (FCM). The expression of promyelocytic leukemia (PML) protein was observed by indirect immune fluorescent test. Results showed that ATRA inhibited the growth of NB4 cells, however, it could not inhibit the growth of NB4-R1 cells. IFN-γ inhibited the growth of both NB4 and NB4-R1 cells. Meanwhile, the growth inhibition effect of IFN-γ in combination with ATRA on both NB4 and NB4-R1 cells was significantly stronger than that of any single drug treatment. The results of the NBT reduction test and CD11b antigen detection by FCM indicated that IFN-γ induces the differentiation of NB4 and NB4-R1 cells to some extent. Moreover, the maturation degree of both NB4 and NB4-R1 cells induced by IFN-γ in combination with ATRA was more significant than that of IFN-γ or ATRA alone. After treatment with IFN-γ, the number of fluorescent particles in NB4 and NB4-R1 cell nuclei was higher than those in the control group, which indicated that IFN-γ may induce the expression of PML protein. Together, IFN-γ augments the proliferation inhibition effect of ATRA on NB4 and NB4-R1 cells through enhancing the expression of PML protein. IFN-γ in combination with ATRA not only strengthens the induction differentiation effect of ATRA on NB4 cells, but also can partially induce the maturation of NB4-R1 cells with ATRA resistance.

3.
PLoS One ; 7(7): e40639, 2012.
Article in English | MEDLINE | ID: mdl-22829880

ABSTRACT

Perforin-1 (PRF), a cytotoxic lymphocyte pore-forming protein, plays an important role in the action of cytotoxic T cells and natural killer cells in that it causes the lysis of abnormal body cells and the elimination of virus-infected cells and tumors. Upon degranulation, PRF inserts itself into the target cell's plasma membrane, forming a pore. The subsequent translocation of pro-apoptotic granzymes (including granzyme B, A, M et al.) into the cytoplasm provides the proteases with access to numerous protein substrates that promote apoptosis after cleavage. These proteases are believed to be the main executioners of target cell apoptosis. Although the PRF and granzyme components are both critical to this process and in some way involved in inducing cell death in target cells, the inhibition of tumor growth could still be efficient in granzyme-deficient mice. It is unclear whether PRF alone can suppress tumors. In this study, we discovered that forced ectopic expression of PRF alone, in the absence of granzymes, could mediate cell death in cancer cells. Notably, transient expression of both full-length and truncated active-form PRF in human Hep G2, SK-BR-3, and HeLa cells was found to induce apparent cell growth inhibition and cell death, as evidenced by chromosome condensation and DNA fragmentation, increased caspase-3 activity, and the release of apoptosis inducing factor (AIF) and cytochrome c from the mitochondria. This PRF-induced cell death could be abrogated by pan-caspase inhibitor (Z-VAD) and mitochondria protector (TAT-BH4). The implication of these results is that ectopically expressed PRF has apoptosis-inducing abilities, and PRF alone is sufficient to induce apoptotic cell death in cells with ectopic expression. Taking this into consideration, our results suggest the possibility of using PRF as a pro-apoptotic gene for tumor therapeutics.


Subject(s)
Apoptosis/physiology , Caspase 3/metabolism , Cytochromes c/metabolism , Perforin/metabolism , Apoptosis/genetics , Apoptosis Inducing Factor/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Fluorescent Antibody Technique , HeLa Cells , Hep G2 Cells , Humans , In Situ Nick-End Labeling , Perforin/genetics , Phosphatidylserines/pharmacology
4.
Ann Saudi Med ; 32(3): 269-75, 2012.
Article in English | MEDLINE | ID: mdl-22588438

ABSTRACT

BACKGROUND AND OBJECTIVES: Radiotherapy is frequently applied in the treatment of malignant gliomas, but it is unclear if radiotherapy exerts its effects via induction of apoptosis. The present study was designed to determine whether a single-fraction γ-60Co radiation can induce apoptosis. DESIGN AND SETTING: In vitro cytological controlled study performed at a military medical university from October 2006 to June 2008. METHODS: C6 cells were treated with a single fraction of ?-60Co radiation at various doses (0, 4, 16, and 64 Gy). The 3-(4,5)-dimethylthiazol-2)-2,5-diphenyl tetrazolium bromide (MTT) assay, apoptosis assays using Annexin V-fluorescein isothiocyanate /propidium iodide or Hoechst 33258 staining, and the cell cycle assay were performed, and the expression of p53 and p21 proteins was evaluated. RESULTS: The C6 cell numbers in the 16 Gy and 64 Gy groups were much lower than in the control group at 48, 96, and 144 hours after irradiation. The irradiated cells underwent apoptosis in a dose-dependent manner. Irradiation also impacted cell cycle progression, arresting cells in the G1 phase. The p53 protein expression was shown in both the nucleus and the cytoplasm of irradiated cells, whereas p53 was only expressed in the nucleus of control (untreated) cells. The p21 protein was expressed in irradiated cells but not in control cells. CONCLUSIONS: Single-fraction ?-60Co radiation inhibited C6 cell growth by inducing apoptosis and G1 arrest, which correlated with the up-regulation of the p53-p21 pathway. The extent of apoptosis and G1 arrest was positively correlated with the dose of radiation. Better understanding of apoptosis induced by radiation therapy will help design optimal dosing schedules for radiation therapy, especially in combination with chemotherapy.


Subject(s)
Apoptosis/radiation effects , Cell Cycle Checkpoints/radiation effects , Cell Proliferation/radiation effects , Gamma Rays , Gene Expression Regulation, Neoplastic/radiation effects , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Radiation , G1 Phase Cell Cycle Checkpoints/radiation effects , Glioma/genetics , Glioma/radiotherapy , Rats , Tumor Suppressor Protein p53/metabolism
5.
Toxicol Sci ; 119(1): 169-77, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20937724

ABSTRACT

Manganese has long been known to induce neurological degenerative disorders. Emerging evidence indicates that hyperphosphorylated tau is associated with neurodegenerative diseases, but whether such hyperphosphorylation plays a role in manganese-induced neurotoxicity remains unclear. To fill this gap, we investigated the effects of manganese on tau phosphorylation in PC12 cells. In our present research, treatment of cells with manganese increased the phosphorylation of tau at Ser199, Ser202, Ser396, and Ser404 as detected by Western blot. Moreover, this manganese-induced tau phosphorylation paralleled the activation of extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK). The mitogen-activated protein kinase kinase-1 (MEK1) inhibitor PD98059, which inhibits the activation of ERK MAPK, partially attenuated manganese-induced tau hyperphosphorylation and cytotoxicity. Moreover, the activation of ERK MAPK was involved in the activation of glycogen synthase kinase-3ß (GSK-3ß) kinase, which also contributed to the hyperphosphorylation of tau and the cytotoxicity in PC12 cells induced by manganese. Taken together, we found for the first time that the exposure to manganese can cause the hyperphosphorylation of tau, which may be connected with the activation of ERK MAPK.


Subject(s)
Chlorides/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , tau Proteins/metabolism , Animals , Cell Culture Techniques , Cell Survival/drug effects , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Manganese Compounds , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/metabolism , PC12 Cells , Phosphorylation , Rats , Signal Transduction
6.
Cytotherapy ; 12(8): 1022-34, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20849360

ABSTRACT

BACKGROUND AIMS: Circulating monocytes have been exploited as an important progenitor cell resource for hepatocytes in vitro and are instrumental in the removal of fibrosis. We investigated the significance of monocytes in peripheral blood stem cells (PBSC) for the treatment of liver cirrhosis. METHODS: Rat CD14+ monocytes in PBSC were mobilized with granulocyte-colony-stimulating factor (G-CSF) and harvested by magnetic cell sorting (MACS). Female rats with carbon tetrachloride (CCl4-induced liver cirrhosis were injected CM-DiI-labeled monocytes, CD14⁻ cells (1 x 107 cells/rat) or saline via the portal vein. RESULTS: Rat CD14+ and CD11b+ monocytes in PBSC were partly positive for CD34, CD45, CD44, Oct3/4 and Sox2, suggesting monocytes with progenitor capacity. Compared with CD14⁻ cell-infused and saline-injected rats, rats undergoing monocyte transplantation showed a gradually increased serum albumin level and decreased portal vein pressure, resulting in a significantly improved survival rate. Meanwhile, monocyte transplantation apparently attenuated liver fibrosis by analysis for fibronectin, α2-(1)-procollagen, α-smooth muscle aorta (SMA) and transforming growth factor (TGF)-ß. Transplanted monocytes mainly clustered in periportal areas of liver, in which 1.8% cells expressed hepatocyte marker albumin and CK18. The expression level of hepatocyte growth factor (HGF), TGF-α, extracellular matrix (EGF) and vascular endothelial growth factor (VEGF) increased, while monocyte transplantation enhanced hepatocyte proliferation. On the other hand, the activities and expression of matrix metalloproteinases (MMP) increased while tissue inhibitor of metalloproteinase (TIMP)-1 expression significantly reduced in monocyte-transplanted livers. Some transplanted monocytes expressed MMP-9 and -13. CONCLUSIONS: The data suggest that CD14+ monocytes in PBSC contribute to hepatocyte regeneration and extracellular matrix (ECM) remodeling in rat liver cirrhosis much more than CD14⁻ cells, and might offer a therapeutic alternative for patients with liver cirrhosis.


Subject(s)
Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hepatocytes/metabolism , Liver Cirrhosis/therapy , Monocyte-Macrophage Precursor Cells/metabolism , Animals , Biomarkers/metabolism , Carbon Tetrachloride/administration & dosage , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Lipopolysaccharide Receptors/biosynthesis , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Liver Cirrhosis/prevention & control , Male , Monocyte-Macrophage Precursor Cells/drug effects , Monocyte-Macrophage Precursor Cells/immunology , Monocyte-Macrophage Precursor Cells/pathology , Rats , Rats, Inbred Strains
7.
Oncol Rep ; 24(5): 1297-305, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20878124

ABSTRACT

Docetaxel is a member of the taxane anti-microtubule class of chemotherapeutic agents, which are currently widely used in clinical cancer therapy. However, the anti-tumor mechanisms of docetaxel are not fully understood. Herein we show that docetaxel induces dose-dependent apoptosis in non-small cell lung cancer A549 cells, as detected by Annexin-V positive cells and PARP cleavage, which is via mitochondrial pathway and dependent on caspase-3 activation. Our study on the mechanisms confirms that docetaxel induces dose-dependent accumulation of cells in M phase and acetylation of α-tubulin, marker of tubulin stablization. Furthermore, docetaxel induces replication-dependent γ-H2AX formation which plays a crucial role in docetaxel-triggered apoptosis. The DNA polymerase inhibitor aphidicolin dose-dependently prevents docetaxel-induced γ-H2AX formation, as well as apoptosis. Notably, 0.6 µM APC almost completely blocked docetaxel-induced γ-H2AX formation and apoptosis. In addition, wortmannin pretreatment caused elevated γ-H2AX level, which was accompanied with increased apoptosis. This effect was due to the inhibition of DNA repair process by wortmannin, as down regulation of p21Waf1/Cip1 and DNA repair proteins such as Ku70, Ku80, DNA-PKcs and Rad50, were detected. These data show, for the first time, that the induction of apoptosis by docetaxel requires DNA replication, and replication-mediated DSBs are critical triggers of docetaxel-induced apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Histones/biosynthesis , Lung Neoplasms/drug therapy , Taxoids/pharmacology , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Division/drug effects , DNA Breaks, Double-Stranded , DNA Repair/drug effects , DNA Replication/drug effects , Docetaxel , Histones/genetics , Histones/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology
8.
Brain Res ; 1359: 201-7, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-20735995

ABSTRACT

Manganese has been known to induce neurological disorders. In the present study, we determined the effect of manganese on the expression of α-synuclein in PC12 cells and its role in manganese-induced cytotoxicity. We also investigated the relationship between α-synuclein expression and the change of ERK1/2 MAPK activity. In our research, manganese exposure induced the overexpression of α-synuclein, while siRNA knockdown of α-synuclein reversed manganese-induced cytotoxicity. Furthermore, manganese induced the activation of ERK1/2 MAPK. The MEK1 inhibitor PD98059, which inhibits the activation of ERK MAPK, attenuated the overexpression of α-synuclein and the cytotoxicity induced by manganese. In conclusion, our studies show that manganese may induce the overexpression of α-synuclein via ERK1/2 activation, which may play a role in manganese-induced cytotoxicity.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/drug effects , Manganese/toxicity , Trace Elements/toxicity , alpha-Synuclein/biosynthesis , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cell Separation , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , PC12 Cells , RNA Interference , Rats , Signal Transduction/drug effects , Signal Transduction/physiology
9.
Med Oncol ; 27(4): 1114-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19908170

ABSTRACT

The medicinal properties of curcumin are well documented in Indian and Chinese systems of medicine, which refer to its wide use in the treatment of some diseases. It has shown to have anti-carcinogenic properties and is known to prevent tumor development in some cancers. In our study, we confirmed that the expression of miR-15a and miR-16 was upregulated and that of Bcl-2 was downregulated in curcumin-treated MCF-7 cells. Silencing miR-15a and miR-16 by specific inhibitors restored the expression of Bcl-2. Thus, we concluded that curcumin can reduce the expression of Bcl-2 by upregulating the expression of miR-15a and miR-16 in MCF-7 cells.


Subject(s)
Adenocarcinoma/genetics , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Curcumin/pharmacology , MicroRNAs/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Female , Humans , MicroRNAs/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Up-Regulation
10.
J Cancer Res Clin Oncol ; 136(2): 267-74, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19693537

ABSTRACT

PURPOSE: Systemic combination chemotherapy is the only option for patients with unresectable hepatocellular carcinoma (HCC) not suitable for intra-arterial treatment. However, no systemic chemotherapy has been able to provide durable remission. The search for a new combination of drugs for HCC is significant. The combination of doxorubicin and paclitaxel shows promise in breast cancer therapy. This study was carried out to determine the synergistic effect of combined doxorubicin and paclitaxel in the two HCC cell lines: HepG2 and Huh7 in vitro and murine HCC H22-bearing BALB/c mice in vivo. METHODS: The morphology of the two cell lines treated with drugs was photomicrographed. The 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide assay was used to determine the number of surviving cells. Cell cycle was evaluated by flow cytometry. Cell viability was measured by the ability of single cells to form colonies in vitro. Anti-tumor activities against subcutaneoulsy implanted solid tumor induced by H22 cells in mice were evaluated. RESULTS: Our data demonstrated that the cytotoxicity produced by doxorubicin and paclitaxel was additive in HCC cells, while it was mainly held in the G2/M phase of the cell cycle by paclitaxel. In vivo anti-tumor activity assay also showed that the combination of the two drugs resulted in more significant tumor regression, compared to the single one. CONCLUSION: The study may provide a new combination of cytotoxic drugs for HCC chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Carcinoma, Hepatocellular/pathology , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/administration & dosage , Flow Cytometry , G2 Phase/drug effects , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Paclitaxel/administration & dosage , Xenograft Model Antitumor Assays
11.
Appl Microbiol Biotechnol ; 86(4): 1067-75, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20012277

ABSTRACT

Hantaan virus (HTNV) is the type of Hantavirus causing hemorrhagic fever with renal syndrome, for which no specific therapeutics are available so far. Cell type-specific internalizing antibodies can be used to deliver therapeutics intracellularly to target cell and thus, have potential application in anti-HTNV infection. To achieve intracellular delivery of therapeutics, it is necessary to obtain antibodies that demonstrate sufficient cell type-specific binding, internalizing, and desired cellular trafficking. Here, we describe the prokaryotic expression, affinity purification, and functional testing of a single-chain Fv antibody fragment (scFv) against HTNV envelop glycoprotein (GP), an HTNV-specific antigen normally located on the membranes of HTNV-infected cells. This HTNV GP-targeting antibody, scFv3G1, was produced in the cytoplasm of Escherichia coli cells as a soluble protein and was purified by immobilized metal affinity chromatography. The purified scFv possessed a high specific antigen-binding activity to HTNV GP and HTNV-infected Vero E6 cells and could be internalized into HTNV-infected cells probably through the clathrin-dependent endocytosis pathways similar to that observed with transferrin. Our results showed that the E. coli-produced scFv had potential applications in targeted and intracellular delivery of therapeutics against HTNV infections.


Subject(s)
Hantaan virus/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Base Sequence , Chlorocebus aethiops , Chromatography, Affinity , Endocytosis , Escherichia coli/genetics , Molecular Sequence Data , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Single-Chain Antibodies/metabolism , Vero Cells
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 25(10): 866-9, 2009 Oct.
Article in Chinese | MEDLINE | ID: mdl-19811728

ABSTRACT

AIM: To construct the replicative deficient adenovirus Ad-Runx3 expressing Runx3, and to express it in U251 malignant glioblastoma cells. METHODS: The runx3 gene with a flag tag was amplified by PCR using pCMV5-AML2 as a template, and was confirmed by DNA sequencing. The adenovirus shuttle vector pShuttle-CMV-Runx3 was constructed by introducing runx3 DNA fragment into the sites of Kpn I and Xho I of pShuttle-CMV vector. This recombinant plasmid was linearized by PmeI and electronically transfected into BJ5183 cells to get the recombinant adenovirus vector Ad-Runx3. The recombinant adenovirus expressing Runx3 was infected into U251 malignant glioblastoma cells. The expression of exogenous Runx3 was observed by immonoblotting and its localization was detected by immunostaining using anti-Flag tag antibody. RESULTS: The recombinant adenovirus expressing Runx3 with a Flag tag was constructed and infected into U251 glioblastoma cells. The exogenous Runx3 protein was detected only in the nuclei. CONCLUSION: The recombinant adenovirus expressing Runx3 with a Flag tag is constructed successfully, and the Runx3 protein expressed in the nuclei of infected cells. The study laid a foundation for further research of the function of Runx3 in gliocarcinogenesis.


Subject(s)
Adenoviridae/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Glioblastoma/pathology , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cloning, Molecular , Core Binding Factor Alpha 3 Subunit/biosynthesis , Core Binding Factor Alpha 3 Subunit/isolation & purification , Gene Expression , Humans , Mice , Plasmids/genetics , Protein Transport , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 25(10): 900-2, 2009 Oct.
Article in Chinese | MEDLINE | ID: mdl-19811737

ABSTRACT

AIM: To explore the different effect and mechanism of arsenic sulfide on telomerase activity and hTERT-mRNA expression in CML cell lines-K562 and APL cell lines-NB4. METHODS: Telomerase activity was determined by polymerase chain reaction enzyme-linked immunoassay (PCR-ELISA). The expression of hTERT-mRNA was analyzed by semi-quantitative RT-PCR. Flow cytometry was used to analyze the cell cycle and apoptosis. RESULTS: 0.15-0.6 mg/L arsenic sulfide (72 h)can induce apoptosis and inhibit telomerase activity and hTERT-mRNA expression in NB4 cell. The concentration of arsenic sulfide with the same effect on K562 cell was 0.3-3 mg/L. 0.3 mg/L arsenic sulfide (72 h) can cause the proportion of the NB4 cell in G2/M phase increased, but for K562 cell, The concentration of arsenic sulfide was 1.5 mg/L. CONCLUSION: Telomerase system may be one of the pathway for arsenic sulfide inducing apoptosis of NB4 and K562 cell; G2/M phrase arrest may have correlation with decrease of telomerase activity; The sensitivity of NB4 and K562 cell for arsenic sulfide is different, the mechanism of it need to study more.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenicals/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Leukemia/pathology , Sulfides/pharmacology , Telomerase/genetics , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Leukemia/enzymology , Leukemia/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Telomerase/metabolism
14.
Biol Res ; 42(2): 249-60, 2009.
Article in English | MEDLINE | ID: mdl-19746271

ABSTRACT

Apoptosis-inducing factor (AIF) is a mitochondrial flavoprotein that mediates both NADH-oxidizing and caspase-independent apoptosis. Further, the proapoptotic activity of AIF is located in the C-terminus of AIF, although the precise minimum sequence responsible for apoptosis induction remains to be investigated. In the present study, we generated two truncated AIFs, AIFDelta1-480-FLAG, which is a FLAG-tagged C-terminal peptide comprising amino acids from 481 to 613, and AIF360-480 containing amino acids from 360 to 480 of AIF. We used confocal microscopy to demonstrate that both the truncated proteins are expressed and located in the cytoplasm of transfected cells. AIFDelta1-480 but not AIF360-480 induces apoptosis in transfected cells. We also found that the expression of AIFDelta1-480 could initiate the release of cytochrome c from the mitochondria. The suppression of caspase-9 via siRNA blocked the proapoptotic activity of AIFDelta1-480. Therefore, AIFDelta1-480 is sufficient for inducing caspase-9-dependent apoptotic signaling, probably by promoting the release of cytochrome c. At last, we generated a chimeric immuno-AIFDelta1-480 protein, which comprised an HER2 antibody, a Pseudomonas exotoxin A translocation domain and AIFDelta1-480. Human Jurkat cells transfected with the immuno-AIFDeltal-480 gene could express and secrete the chimeric protein, which selectively recognize and kill HER2-overexpressing tumor cells. Our study demonstrates the feasibility of the immuno-AIFDeltal-480 gene as a novel approach to treating HER2-overexpressing cancers.


Subject(s)
Alcohol Oxidoreductases/drug effects , Apoptosis Inducing Factor/pharmacology , Apoptosis/drug effects , DNA, Complementary/drug effects , DNA-Binding Proteins/drug effects , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Apoptosis/genetics , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Blotting, Western , DNA, Complementary/genetics , DNA, Complementary/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Jurkat Cells , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Transfection
15.
Biol. Res ; 42(2): 249-260, 2009. ilus, tab
Article in English | LILACS | ID: lil-524896

ABSTRACT

Apoptosis-inducing factor (AIF) is a mitochondrial flavoprotein that mediates both NADH-oxidizing and caspase-independent apoptosis. Further, the proapoptotic activity of AIF is located in the C-terminus of AIF, although the precise minimum sequence responsible for apoptosis induction remains to be investigated. In the present study, we generated two truncated AIFs, AIFΔ1-480-FLAG, which is a FLAG-tagged C-terminal peptide comprising amino acids from 481 to 613, and AIF360-480 containing amino acids from 360 to 480 of AIF. We used confocal microscopy to demonstrate that both the truncated proteins are expressed and located in the cytoplasm of transfected cells. AIFΔ1-480 but not AIF360-480 induces apoptosis in transfected cells. We also found that the expression of AIFΔ1-480 could initiate the release of cytochrome c from the mitochondria. The suppression of caspase-9 via siRNA blocked the proapoptotic activity of AIFΔ1-480. Therefore, AIFΔ 1-480 is sufficient for inducing caspase-9-dependent apoptotic signaling, probably by promoting the release of cytochrome c. At last, we generated a chimeric immuno-AIFΔ 1-480 protein, which comprised an HER2 antibody, a Pseudomonas exotoxin A translocation domain and AIFΔ 1-480. Human Jurkat cells transfected with the immuno-AIFΔl-480 gene could express and secrete the chimeric protein, which selectively recognize and kill HER2-overexpressing tumor cells. Our study demonstrates the feasibility of the immuno-AIFΔl-480 gene as a novel approach to treating HER2-overexpressing cancers.


Subject(s)
Humans , Alcohol Oxidoreductases/drug effects , Apoptosis Inducing Factor/pharmacology , Apoptosis/drug effects , DNA, Complementary/drug effects , DNA-Binding Proteins/drug effects , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Apoptosis/genetics , Blotting, Western , DNA, Complementary/genetics , DNA, Complementary/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique, Indirect , Jurkat Cells , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Transfection
16.
Adv Ther ; 25(12): 1331-41, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19034397

ABSTRACT

INTRODUCTION: The action of cortistatin (CST), a novel cyclic neuropeptide, as an anti-inflammatory factor has been studied, but few investigations have explored the immunomodulatory role of CST in transplantation. In the present study, we examined whether CST affects the alloimmune response in a mouse model of skin transplantation and the effects of CST on T lymphocytes. METHODS: BALB/c (H-2K(d)) recipient mice (n=70) were divided into seven groups (n=10 per group) and given an intraperitoneal injection of CST or a somatostatin analog, SMS 201-995 (octreotide), on the day of skin transplantation from C57BL/6 (B6) (H-2K(b)) donors. Injections were continued for 7 consecutive days. Groups 1-3 received CST at doses of 0.02, 0.2, or 2 mg/kg, respectively. Groups 4-6 received SMS 201-995 at the same doses. Group 7 was a control group and received injections of phosphate buffered saline. Survival of the allografts was recorded. A semiquantitative reverse transcriptase polymerase chain reaction study of Foxp3 expression and a flow cytometry study of CD4 and CD25 markers of T lymphocytes were conducted to determine whether CD4(+)CD25(+) Foxp3(high) regulatory T cells (T(reg)) were generated in vivo. RESULTS: BALB/c mice given CST (0.2 or 2 mg/kg) had prolonged graft survival (median survival time [MST], 13 and 14 days, respectively; P<0.05 compared with controls). SMS 201-995 at the same concentrations did not have a significant effect on allograft survival (MST, 8 days for both groups). We found more than a twofold increase of CD4(+)CD25(+) T(reg) cells in the CD4(+) T-cell population and the expression of Foxp3 was up-regulated in the CST treatment groups, compared with control and SMS 201-995 treatment groups. CONCLUSION: In our study, CST induced a significant prolongation in survival time of allogeneic skin grafts and increased the generation of CD4(+)CD25(+) Foxp 3(high) T(reg) cells. These results suggest that CST may become a new modality in controlling allograft rejection.


Subject(s)
Graft Rejection/prevention & control , Immunologic Factors/therapeutic use , Neuropeptides/therapeutic use , Octreotide/therapeutic use , Skin Transplantation/immunology , Animals , Flow Cytometry , Forkhead Transcription Factors/biosynthesis , Graft Rejection/immunology , Graft Rejection/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
17.
Mol Immunol ; 45(14): 3818-26, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18617268

ABSTRACT

Dendritic cells (DC) are specialized cells that capture and present antigen to T cells. Recent advances have been made in understanding their origin, heterogeneity, and the signals that induce their migration and maturation resident microglia are antigen-presenting cells (APC) involved in stimulation or reactivation of CNS-targeted T cells. Generation of DC from microglia, as demonstrated ex vivo, may support GM-CSF-driven differentiation of brain DC from local, likely, microglial progenitors. Here, we report the establishment of long-term cultures of rat ecto-mesenchymal stem cells (EMSCs) using specific supplemented media for induction. These EMSCs share some morphological characteristics and the allostimulatory capacity of classical DCs, and when transplanted into the brain using a rat glioma model survive within the cortex, and are morphologically and phenotypically similar to microglia over 7 days. Our findings related to the development and differentiation of microglial progenitors support the view that microglia are derived prenatally from mesodermal progenitors that are distinct from monocytes.


Subject(s)
Antigen-Presenting Cells/physiology , Cell Differentiation/physiology , Dendritic Cells/immunology , Mesenchymal Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Bone Marrow Cells/cytology , Cell Culture Techniques , Cell Line, Tumor , Cells, Cultured , Culture Media/chemistry , Dendritic Cells/ultrastructure , Embryo, Mammalian/cytology , Female , Glioma/pathology , Immunohistochemistry , Mesenchymal Stem Cell Transplantation , Neoplasm Transplantation , Pregnancy , Rats , Rats, Sprague-Dawley , Transplantation, Homologous
18.
Biochem Biophys Res Commun ; 358(4): 1041-6, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17524364

ABSTRACT

Human Apr3 was first cloned from HL-60 cells treated by ATRA. In this study, we further demonstrated that Apr3 could be obviously upregulated by ATRA in many other ATRA sensitive cells, suggesting a common role of Apr3 in ATRA effects. Indirect immunofluorescence assay indicates that Apr3 is a membrane protein, while its truncated form without the predicted transmembrane and intracellular domain, was likely a secreted one. Furthermore, FACS analysis showed that Apr3 overexpression could cause an obvious G1/S phase arrest which might be induced by dramatic reduction of cyclin D1 expression. Strikingly, the truncated Apr3 antagonized the negative role of Apr3 on cell cycle and cyclin D1. Taken together, our data suggest that Apr3 should play an important role in ATRA signal pathway and the predicted transmembrane and/or the intracellular domain mediates Apr3 membrane localization and is vital for the negative regulation on cell cycle and cyclin D1.


Subject(s)
Actin-Related Protein 3/metabolism , Cell Cycle/physiology , Cyclins/metabolism , Gene Expression Regulation/physiology , Tretinoin/administration & dosage , Cell Cycle/drug effects , Cyclin D , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Gene Expression Regulation/drug effects , HL-60 Cells , Humans , Up-Regulation/drug effects , Up-Regulation/physiology
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 23(3): 205-8, 2007 Mar.
Article in Chinese | MEDLINE | ID: mdl-17343783

ABSTRACT

AIM: To explore the possibility and the possible mechanism of reversing ATRA-resistance in MR2 cells by using IFN-alpha and IFN-gamma in combination with all-trans retinoic acid (ATRA). METHODS: After MR2 cells(ATRA-resistance cell line) were treated with IFN-alpha, IFN-gamma and ATRA alone or IFN-alpha and IFN-gamma in combination with ATRA respectively, the cell proliferation was tested by MTT colorimetry, the cell differentiation was tested through light microscope, by NBT test and flow cytometry (FCM). The expression of promyelocytic leukemia (PML) protein was observed by indirect immunofluorescence staining. RESULTS: Both IFN-alpha and IFN-gamma could inhibit the proliferation of MR2 cells. The effects were more obviously in both IFN-alpha+ATRA group and IFN-gamma+ATRA group. But there were no significant difference between either IFN-alpha group and IFN-gamma group or IFN-alpha+ATRA group and IFN-gamma+ATRA group (P>0.05). Both IFN could also induce the differentiation of MR2 cells. The effects of IFN-alpha+ATRA group and IFN-gamma+ATRA group were more obvious. However, the differentiation of MR2 cells induced by IFN-gamma+ATRA group was more higher than that by IFN-alpha+ATRA group (P<0.05). Both IFN could induce the expression of PML protein. CONCLUSION: The reversing effcet of IFN-gamma+ATRA group on ATRA-resistence in MR2 cells are more powerful than that of IFN-alpha+ATRA group, which may be related to the different signal transduction pathway of IFN-alpha and IFN-gamma.


Subject(s)
Drug Resistance/drug effects , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Tretinoin/physiology , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Microscopy , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Transcription Factors/metabolism , Tretinoin/pharmacology , Tumor Suppressor Proteins/metabolism
20.
Clin Immunol ; 119(3): 245-51, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16616877

ABSTRACT

The expression of the apoptosis-inducing ligands, TNF-alpha, FasL and TRAIL on peripheral blood mononuclear cells (PBMC) and the levels of their soluble form (TNF-alpha, sFasL and sTRAIL) in plasma from 40 hemorrhagic fever with renal syndrome (HFRS) patients as well as 26 healthy blood donors were determined by flow cytometry (FCM) analysis and sandwich ELISA, respectively. The status of Th1, Th2, Tc1 and Tc2 subsets in PBMC was evaluated by intracellular cytokine staining and FCM. Compared to controls, the expression of membrane bound FasL and TRAIL was up-regulated on surface of PBMC isolated from the HFRS patients, particularly on CD8+ T lymphocytes. The levels of TNF-alpha, sFasL and sTRAIL in plasma from the HFRS patients in the acute phase increase 4.7-fold, 6.0-fold and 1.8-fold, respectively, over those from the healthy donors. The percentage of Th1, Tc1 and Tc2 subsets in PBMC from the patients also increased significantly compared with those from healthy donors. These results indicate that dynamic changes occurred in both the membrane bound and soluble forms of apoptosis-inducing ligands (FasL, TRAIL and TNF-alpha) and proportions of Th1 and CTL in HFRS patients increased. Both factors may play an important role in the etiology of Hantaan virus infection in humans.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Apoptosis/immunology , Hemorrhagic Fever with Renal Syndrome/immunology , Membrane Glycoproteins/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factors/immunology , Adolescent , Adult , Aged , Apoptosis Regulatory Proteins/blood , Child , Fas Ligand Protein , Female , Hantaan virus , Hemorrhagic Fever with Renal Syndrome/blood , Humans , Male , Membrane Glycoproteins/blood , Middle Aged , T-Lymphocytes/immunology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factors/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...