Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Mol Neurobiol ; 55(7): 5879-5888, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29101647

ABSTRACT

Stroke causes death or long-term disabilities and threatens the general health of the population worldwide. Recent studies have suggested that miRNAs are dysregulated and can be used as biomarkers for diagnosis and prognosis in stroke. The intracerebral hemorrhage (ICH) accounts for 15% of all the stroke cases. However, at present, little is known regarding the functions and clinical implications of miRNAs in ICH. In the present study, we established the collagenase-induced rat ICH model to mimic human ICH syndrome. We profiled the expression of 728 rat miRNAs at different time points in rat brain tissues and plasma post-ICH and identified a set human brain-enriched miRNAs that had changed expression level in the plasma of rat ICH. Among them, the expression levels of miR-124 displayed significantly synchronous alterations in rat plasma and brain tissue during ICH progression. They were significantly elevated at the acute injury phase (day 1 and 2), gradually decreased during the delayed recovery phase (day 7, 14 and 30), and finally restored to normal levels at late recovery phase (day 60). We further determined the plasma expression profile of miR-124 from human ICH patients. Similar to the pattern observed in rat ICH model, our results indicated that immediately after patients reached the hospital, the average plasma concentrations of miR-124 increased more than 100-fold in 24 h, then decreased gradually on day 2, 7, 14 and to near normal level on day 30. Taken together, these results strongly suggested that plasma concentration of miR-124 is a promising candidate biomarker for the early detection and predictive prognosis of human ICH.


Subject(s)
Cerebral Hemorrhage/blood , Cerebral Hemorrhage/genetics , MicroRNAs/blood , Stroke/blood , Stroke/genetics , Animals , Biomarkers/blood , Case-Control Studies , Cerebral Hemorrhage/pathology , Disease Models, Animal , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Male , Middle Aged , Rats, Sprague-Dawley , Stroke/pathology
2.
PLoS One ; 10(12): e0144724, 2015.
Article in English | MEDLINE | ID: mdl-26675167

ABSTRACT

BACKGROUND: Aneurysmal subarachnoid hemorrhage (SAH) is a highly morbid and fatal condition with high rate of cognitive impairment and negative impact in quality of life among survivors. Delayed cerebral infarction (DCI) is one the major factors for these negative outcomes. In this study we compared the circulating microRNA profiles of SAH patients and healthy individuals, and the circulating microRNA profiles of SAH patients with and without DCI. METHODS: Peripheral blood samples on Day 7 after the onset of SAH were subjected to microarray analysis with Affymetrix miRNA 3.0 array and quantitative PCR analysis. SAH patients with (N = 20) and without DCI (N = 20) and Healthy controls (N = 20) were included for analyses. RESULTS: We demonstrated that 99 miRNAs were found to be dysregulated in the SAH patient group with DCI. 81 miRNAs were upregulated and 18 were downregulated. Findings from KEGG pathway analysis showed that miRNAs and target genes for axon guidance and TGF-beta signaling were involved, implying that the resulted differential miRNA expression pattern reflect the results of SAH instead of etiology of the disease. miR-132-3p and miR-324-3p showed distinctive upregulations in qPCR [miR-132: 9.5 fold (95%CI: 2.3 to 16.7) in DCI group and 3.4 fold (95%CI: 1.0 to 5.8) in Non-DCI group; miR-324: 4924 fold (95%CI: 2620 to 7228) in DCI group and 4545 fold (95%CI: 2408 to 6683) in non-DCI group]. However, there were no significant differences in fold changes between SAH patients with and without DCI [fold change ratios (mean+/-SD): 2.7+/-4.2 and 1.1+/-1.1 for miRNA-132 and miRNA-324]. CONCLUSION: Our study demonstrated that as compared to healthy control, miR-132 and miR-324 showed a upregulation in both SAH DCI and Non-DCI groups. However, the differences between the SAH DCI and non-DCI groups were not statistically significant.


Subject(s)
Intracranial Aneurysm/complications , MicroRNAs/genetics , Subarachnoid Hemorrhage/blood , Subarachnoid Hemorrhage/etiology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers , Case-Control Studies , Female , Gene Expression Profiling , Humans , Male , MicroRNAs/blood , Middle Aged , ROC Curve , Young Adult
3.
J Biol Chem ; 287(13): 9962-9971, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22253443

ABSTRACT

miR-124 is a brain-enriched microRNA that plays a crucial role in neural development and has been shown to be down-regulated in glioma and medulloblastoma, suggesting its possible involvement in brain tumor progression. Here, we show that miR-124 is down-regulated in a panel of different grades of glioma tissues and in all of the human glioma cell lines we examined. By integrated bioinformatics analysis and experimental confirmation, we identified SNAI2, which is often up-regulated in glioma, as a direct functional target of miR-124. Because SNAI2 has been shown to regulate stem cell functions, we examined the roles of miR-124 and SNAI2 in glioma cell stem-like traits. The results showed that overexpression of miR-124 and knockdown of SNAI2 reduced neurosphere formation, CD133(+) cell subpopulation, and stem cell marker (BMI1, Nanog, and Nestin) expression, and these effects could be rescued by re-expression of SNAI2. Furthermore, enhanced miR-124 expression significantly inhibited glioma cell invasion in vitro. Finally, stable overexpression of miR-124 and knockdown of SNAI2 inhibited the tumorigenicity and invasion of glioma cells in vivo. These findings reveal, for the first time, that the tumor suppressor activity of miR-124 could be partly due to its inhibitory effects on glioma stem-like traits and invasiveness through SNAI2.


Subject(s)
Antigens, Differentiation/metabolism , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Glioma/metabolism , MicroRNAs/biosynthesis , Neoplastic Stem Cells/metabolism , Animals , Antigens, Differentiation/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms , Cell Line, Tumor , Down-Regulation/genetics , Glioma/genetics , Glioma/pathology , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Snail Family Transcription Factors , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation/genetics
4.
Int J Pharm ; 426(1-2): 182-192, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22265912

ABSTRACT

In this study we describe a novel polymer, mPPS-FA, synthesized as a potential gene transfer vector. To complete mPPS-FA, folic acid was conjugated to a backbone (named mPPS) consisting of a copolymer of methyl PEG-2000, PEI-600, and sebacoyl chloride. (1)H NMR, FT-IR, and UV spectroscopy were used to characterize the structure of mPPS-FA. It was revealed that mPPS-FA holds the ability to bind plasmid DNA yielding positively charged particles (polyplexes). Dynamic light scattering (DLS) and TEM techniques were used to study the size and morphology of the formed mPPS-FA/DNA nanocomplexes. The mPPS-FA/DNA nanoparticles exhibited low cytotoxicity as transfection of B16-F0, U87MG, CHO-1, and Ho-8910 cells produced >80% viability indicating low cytotoxicity of the polymer. The ability of mPPS-FA to deliver EGFP plasmid to melanoma B16-F0, U87, CHO-1, Ho-8910, and A549 cells was investigated in vitro as compared to the lipid-based transfection agent Lipofectamine2000 and Linear PEI 22 kDa (L-PEI 22 kDa). We found that mPPS-FA/DNA complexes yielded the highest GFP transfection efficiency in B16-F0, U87, CHO-1, and Ho-8910 cells, which all highly express folate receptors (FR), at an mPPS-FA/DNA ratio (w/w) of 15. Furthermore, the transfection of mPPS-FA/DNA complexes in CHO-1 cells could be competitively blocked by free folic acid molecules. In contrast, in low FR expressing A549 cells, mPPS-FA showed similar low transfection efficiency as mPPS. Taken together, mPPS-FA showed the highest efficiency in vitro and the potential to be developed as a nonviral gene carrier.


Subject(s)
Alkanes/chemistry , DNA/metabolism , Folic Acid/metabolism , Imines/chemistry , Polyethylene Glycols/chemistry , Polyethylenes/chemistry , Transfection/methods , Alkanes/toxicity , Animals , Binding, Competitive , CHO Cells , Cell Survival/drug effects , Cricetinae , Cricetulus , DNA/chemistry , Folic Acid/chemistry , Folic Acid/toxicity , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Imines/toxicity , Light , Magnetic Resonance Spectroscopy , Methylation , Mice , Microscopy, Electron, Transmission , Nanoparticles , Nanotechnology , Particle Size , Polyethylene Glycols/toxicity , Polyethylenes/toxicity , Scattering, Radiation , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared
5.
J Biol Chem ; 286(46): 39703-14, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21903590

ABSTRACT

The human microRNA cluster MC-let-7a-1∼let-7d, with three members let-7a-1, let-7f-1, and let-7d, is an important cluster of the let-7 family. These microRNAs play critical roles in regulating development and carcinogenesis. Therefore, precise control of MC-let-7a-1∼let-7d level is critical for cellular functions. In this study, we first showed that the expression of these three members was significantly reduced in human hepatocellular carcinoma HepG2 cells as compared with the immortalized human liver L02 cells. We demonstrated that the MC-let-7a-1∼let-7d cluster was encoded by a single polycistronic transcript driven by a 10-kb upstream promoter, with two MYC-binding sites. Importantly, MYC inhibited MC-let-7a-1∼let-7d promoter activity via binding to the noncanonical E-box 3 downstream of the transcription start sites, whereas it enhanced promoter activity by binding to the canonical E-box 2 upstream of the transcription start sites. We found that although the binding affinity of MYC to E-box 2 was stronger than E-box 3, the binding quantum of MYC to E-box 3 was significantly higher in cancerous HepG2 cells as compared with the noncancerous L02 cells. In addition, forced expression of let-7 could reverse the MYC-mediated cell proliferation. These findings suggested that in L02 cells with a low level of MYC, MYC binds mainly to E-box 2 to enhance MC-let-7a-1∼let-7d expression. However, in HepG2 cells with an elevated MYC, the extra MYC could bind to E-box 3 to suppress the transcription of MC-let-7a-1∼let-7d and thus enable HepG2 cells to maintain a high level of MYC and a low level of let-7 microRNAs simultaneously.


Subject(s)
MicroRNAs/biosynthesis , Multigene Family , Proto-Oncogene Proteins c-myc/metabolism , Response Elements , Transcription, Genetic , HEK293 Cells , Hep G2 Cells , Humans , MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/genetics
6.
Mol Cancer Ther ; 10(6): 1082-92, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21518728

ABSTRACT

Interleukin-2 (IL-2) has been shown to possess antitumor activity in numerous preclinical and clinical studies. However, the short half-life of recombinant IL-2 protein in serum requires repeated high-dose injections, resulting in severe side effects. Although adenovirus-mediated IL-2 gene therapy has shown antitumor efficacy, the host antibody response to adenoviral particles and potential biosafety concerns still obstruct its clinical applications. Here we report a novel nanopolymer for IL-2 delivery, consisting of low molecular weight polyethylenimine (600 Da) linked by ß-cyclodextrin and conjugated with folate (named H1). H1 was mixed with IL-2 plasmid to form H1/pIL-2 polyplexes of around 100 nm in diameter. Peritumoral injection of these polyplexes suppressed the tumor growth and prolonged the survival of C57/BL6 mice bearing B16-F1 melanoma grafts. Importantly, the antitumor effects of H1/pIL-2 (50 µg DNA) were similar to those of recombinant adenoviruses expressing IL-2 (rAdv-IL-2; 2 × 10(8) pfu). Furthermore, we showed that H1/pIL-2 stimulated the activation and proliferation of CD8+, CD4+ T cell, and natural killer cells in peripheral blood and increased the infiltration of CD8+, CD4+ Tcells, and natural killer cells into the tumor environment. In conclusion, these results show that H1/pIL-2 is an effective and safe melanoma therapeutic with an efficacy comparable to that of rAdv-IL-2. This treatment represents an alternative gene therapy strategy for melanoma.


Subject(s)
Immunotherapy/methods , Interleukin-2/administration & dosage , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Nanoparticles/administration & dosage , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Drug Delivery Systems , Female , Folic Acid/chemistry , Humans , Interleukin-2/chemistry , Interleukin-2/genetics , Killer Cells, Natural/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Polyethyleneimine/chemistry , Polymers/chemistry , Polymers/therapeutic use , T-Lymphocytes, Helper-Inducer/metabolism , Transgenes , beta-Cyclodextrins/chemistry
7.
J Neurooncol ; 103(2): 187-95, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20838853

ABSTRACT

Interferon-induced transmembrane protein 1 (IFITM1) has recently been identified as a new molecular marker in human colorectal cancer. However, its role in glioma carcinogenesis is not known. In this study, we demonstrated that suppression of IFITM1 expression significantly inhibited proliferation of glioma cells in a time-dependent manner. The growth inhibitory effect was mediated by cell cycle arrest. Furthermore, IFITM1 knockdown significantly inhibited migration and invasion of glioma cells, which could be attributed to decreased expression and enzymatic activity of matrix metalloproteinase 9. Taken together, these results suggest that IFITM1 is a potential therapeutic target for gliomas.


Subject(s)
Antigens, Differentiation/metabolism , Cell Movement/genetics , Cell Proliferation , Glioma/metabolism , Antigens, Differentiation/genetics , Blotting, Western , Cell Line, Tumor , Gene Knockdown Techniques , Glioma/genetics , Glioma/pathology , Humans , Neoplasm Invasiveness/genetics , Reverse Transcriptase Polymerase Chain Reaction
8.
J Biol Chem ; 285(47): 36995-7004, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20826811

ABSTRACT

The emerging concept of generating cancer stem cells from epithelial-mesenchymal transition has attracted great interest; however, the factors and molecular mechanisms that govern this putative tumor-initiating process remain largely elusive. We report here that miR-200a not only regulates epithelial-mesenchymal transition but also stem-like transition in nasopharyngeal carcinoma cells. We first showed that stable knockdown of miR-200a promotes the transition of epithelium-like CNE-1 cells to the mesenchymal phenotype. More importantly, it also induced several stem cell-like traits, including CD133(+) side population, sphere formation capacity, in vivo tumorigenicity in nude mice, and stem cell marker expression. Consistently, stable overexpression of miR-200a switched mesenchyme-like C666-1 cells to the epithelial state, accompanied by a significant reduction of stem-like cell features. Furthermore, in vitro differentiation of the C666-1 tumor sphere resulted in diminished stem-like cell population and miR-200a induction. To investigate the molecular mechanism, we demonstrated that miR-200a controls epithelial-mesenchymal transition by targeting ZEB2, although it regulates the stem-like transition differentially and specifically by ß-catenin signaling. Our findings reveal for the first time the function of miR-200a in shifting nasopharyngeal carcinoma cell states via a reversible process coined as epithelial-mesenchymal to stem-like transition through differential and specific mechanisms.


Subject(s)
Epithelial-Mesenchymal Transition , Homeodomain Proteins/metabolism , MicroRNAs/physiology , Nasopharyngeal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Repressor Proteins/metabolism , beta Catenin/metabolism , Animals , Blotting, Western , Cell Differentiation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zinc Finger E-box Binding Homeobox 2 , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
9.
Eur J Cancer ; 46(9): 1752-61, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20466538

ABSTRACT

Cell cycle-related kinase (CCRK) is a newly identified protein kinase homologous to Cdk7. We have previously shown that CCRK is a candidate oncogene in human glioblastoma. However, whether CCRK is a bona fide oncogene remains to be tested. The aim of this study was to investigate the role of CCRK in human colorectal cancer carcinogenesis. By Western blotting, we analysed the expression profile of CCRK protein in 10 colorectal cancer tissue samples and their adjacent normal colon tissues and in seven colorectal cancer cell lines. CCRK protein expression was also investigated by immunohistochemistry in a colorectal tissue microarray, which contained 120 cases of primary colorectal cancer and adjacent normal colorectal mucosa. The effects of CCRK knock-down on cell cycle profile and proliferation of colorectal cancer cells were examined by transfecting LoVo and DLD1 human colorectal cancer cell lines by either short-hairpin RNA (shCCRK) or small interfering RNA targeting CCRK (siCCRK). We found that CCRK protein levels were elevated by more than 1.5-fold in 70% of colorectal cancer patient samples examined and CCRK was detectable in all seven colorectal cancer cell lines tested. Colorectal tissue microarray indicated that overexpression of CCRK was detected in 62/109 (56.9%) of informative colorectal cancer cases and was significantly associated with the tumour pT and pN status (p<0.05). Suppression of CCRK by siCCRK led to G1 phase cell cycle arrest and reduced cell growth. Consistently, stable clones of LoVo and DLD1 cells expressing shCCRK exhibited decreased cell proliferation rates. Furthermore, we showed that CCRK is required for the phosphorylation of Cdk2 (on Thr-160) and Rb (on Ser-795) and the expression of cyclin E. These results suggest for the first time that CCRK is involved in colorectal cancer carcinogenesis and G1/S cell cycle transition by regulating Cdk2, cyclin E and Rb.


Subject(s)
Colorectal Neoplasms/etiology , Cyclin-Dependent Kinases/physiology , Neoplasm Proteins/physiology , Adult , Aged , Aged, 80 and over , Blotting, Western , Cell Cycle/physiology , Cell Line, Tumor , Cyclins/metabolism , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Male , Microarray Analysis , Middle Aged , Phosphorylation , RNA, Small Interfering/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Cyclin-Dependent Kinase-Activating Kinase
10.
Biochem Biophys Res Commun ; 391(1): 535-41, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19931509

ABSTRACT

Nasopharyngeal carcinoma (NPC), a highly metastatic and invasive malignant tumor originating from the nasopharynx, is widely prevalent in Southeast Asia, the Middle East and North Africa. Although viral, dietary and genetic factors have been implicated in NPC, the molecular basis of its pathogenesis is not well defined. Based on a recent microRNA (miRNA) microarray study showing miR-200 downregulation in NPC, we further investigated the role of miR-200a in NPC carcinogenesis. We found that the endogenous miR-200a expression level increases with the degree of differentiation in a panel of NPC cell lines, namely undifferentiated C666-1, high-differentiated CNE-1, and low-differentiated CNE-2 and HNE1 cells. By a series of gain-of-function and loss-of-function studies, we showed that over-expression of miR-200a inhibits C666-1 cell growth, migration and invasion, whereas its knock-down stimulates these processes in CNE-1 cells. In addition, we further identified ZEB2 and CTNNB1 as the functional downstream targets of miR-200a. Interestingly, knock-down of ZEB2 solely impeded NPC cell migration and invasion, whereas CTNNB1 suppression only inhibited NPC cell growth, suggesting that the inhibitory effects of miR-200a on NPC cell growth, migration and invasion are mediated by distinct targets and pathways. Our results reveal the important role of miR-200a as a regulatory factor of NPC carcinogenesis and a potential candidate for miRNA-based therapy against NPC.


Subject(s)
Carcinoma/pathology , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , MicroRNAs/physiology , Nasopharyngeal Neoplasms/pathology , Repressor Proteins/genetics , beta Catenin/genetics , Base Sequence , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Down-Regulation , Humans , MicroRNAs/genetics , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Neoplasm Invasiveness , Zinc Finger E-box Binding Homeobox 2
11.
Int J Cancer ; 125(11): 2631-42, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19672860

ABSTRACT

Our previous study has suggested that the cell cycle-related kinase (CCRK) is a putative candidate oncogene in glioblastoma tumorigenesis. The potential oncogenic role of CCRK and its clinical/prognostic significance, however, in ovarian carcinoma are unclear. In this study, CCRK expression was examined by immunohistochemistry in a series of ovarian carcinoma tissues. Overexpression of CCRK was detected in 53% of the ovarian carcinomas, and it was positively correlated with an ascending histological grade and/or advanced clinical stage of the disease (p < 0.05). In addition, overexpression of CCRK in ovarian carcinoma was determined to be a strong and an independent predictor of short overall survival (p < 0.05). In ovarian carcinoma cells, CCRK knockdown by RNAi led to a G1 phase cell cycle arrest, while CCRK overexpression by stable transfection of CCRK-containing plasmid pcDNA-CCRK promoted cell proliferation in vitro and tumor growth in vivo. In addition, CCRK knockdown was found to reduce cyclin D1 expression. Consistently, CCRK overexpression increased cyclin D1 expression, and furthermore, a significant correlation between expression of CCRK and cyclin D1 in ovarian carcinomas was observed (p < 0.001). These findings suggest a potential important role of CCRK in the control of cell proliferation via regulation of cyclin D1 expression, and the overexpression of CCRK, as detected by immunohistochemistry, is an independent molecular marker for shortened survival time of patients with ovarian carcinoma.


Subject(s)
Cell Proliferation , Cyclin D1/metabolism , Cyclin-Dependent Kinases/metabolism , Cystadenocarcinoma, Serous/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/metabolism , Adult , Aged , Animals , Apoptosis , Blotting, Western , Case-Control Studies , Cohort Studies , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/secondary , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Middle Aged , Ovarian Neoplasms/genetics , Ovary/pathology , Phosphorylation , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tissue Array Analysis , Tumor Cells, Cultured , Cyclin-Dependent Kinase-Activating Kinase
12.
J Natl Cancer Inst ; 99(12): 936-48, 2007 Jun 20.
Article in English | MEDLINE | ID: mdl-17565152

ABSTRACT

BACKGROUND: Median survival for patients with glioblastoma multiforme, the most aggressive glioma, is only 12-15 months, despite multimodal treatment that includes surgery, chemotherapy, and radiotherapy. Thus, identification of genes that control the progression of glioblastoma multiforme is crucial for devising new therapies. We investigated the involvement of cell cycle-related kinase (CCRK), a novel protein kinase that is homologous to cyclin-dependent kinase 7, in glioblastoma multiforme carcinogenesis. METHODS: We analyzed the expression levels of CCRK in 26 glioma patient samples (19 high-grade and seven low-grade) and normal brain by semiquantitative reverse transcription-polymerase chain reaction assays. CCRK expression was knocked down in human glioma U-373 MG and U-87 MG cells with small-interfering RNAs and short hairpin RNAs (siCCRK and shCCRK, respectively), and cell proliferation, cell cycle distribution, and cyclin-dependent kinase 2 (CDK2) phosphorylation were examined. A subcutaneous nude mouse xenograft model (n = 4 mice per group) was used to study the effect of CCRK knockdown and overexpression on tumorigenicity and growth of glioblastoma multiforme cells in vivo. All statistical tests were two-sided. RESULTS: CCRK mRNA was elevated at least 1.5-fold and as much as 3.7-fold in 14 (74%) of 19 high-grade glioblastoma multiforme patient samples and in four (80%) of five glioma cell lines examined compared with normal brain tissue. Suppression of CCRK by siCCRK inhibited the proliferation of U-373 MG and U-87 MG glioblastoma cells in a time- and dose-dependent manner. The growth-inhibiting effect of siCCRK was mediated via G1- to S-phase cell cycle arrest and reduced CDK2 phosphorylation. CCRK knockdown statistically significantly suppressed glioma cell growth in vivo as indicated by the mean tumor volumes at week 6 after tumor cell injection (U-373-control = 1352 mm3, U-373-shCCRK = 294 mm3, difference = 1058 mm3, 95% confidence interval [CI] = 677 to 1439 mm3, P<.001; U-87-control = 1910 mm3, U-87-shCCRK = 552 mm3, difference = 1358 mm3, 95% CI = 977 to 1739 mm3, P<.001). CONCLUSIONS: CCRK is a candidate oncogene in glioblastoma multiforme tumorigenesis.


Subject(s)
Brain Neoplasms/genetics , Cyclin-Dependent Kinases/genetics , Glioblastoma/genetics , Oncogenes , Animals , Brain Neoplasms/enzymology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/biosynthesis , Cyclin-Dependent Kinases/metabolism , Female , Glioblastoma/enzymology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Phosphorylation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transplantation, Heterologous , Cyclin-Dependent Kinase-Activating Kinase
13.
World J Gastroenterol ; 10(1): 82-5, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14695774

ABSTRACT

AIM: To evaluate the covalently closed circle DNA (cccDNA) level of hepatitis B virus (HBV) in patients' liver and sera. METHODS: HBV DNA was isolated from patients' liver biopsies and sera. A sensitive real-time PCR method, which is capable of differentiation of HBV viral genomic DNA and cccDNA, was used to quantify the total HBV cccDNA. The total HBV viral DNA was quantitated by real-time PCR using a HBV diagnostic kit (PG Biotech, LTD, Shenzhen, China) described previously. RESULTS: For the first time, we measured the level of HBV DNA and cccDNA isolated from ten HBV patients' liver biopsies and sera. In the liver biopsies, cccDNA was detected from all the biopsy samples. The copy number of cccDNA ranged from from 0.03 to 173.1 per cell, the copy number of total HBV DNA ranged from 0.08 to 3 717 per cell. The ratio of total HBV DNA to cccDNA ranged from 1 to 3 406. In the sera, cccDNA was only detected from six samples whereas HBV viral DNA was detected from all ten samples. The ratio of cccDNA to total HBV DNA ranged from 0 to 1.77%. To further investigate the reason why cccDNA could only be detected in some patients' sera, we performed longitudinal studies. The cccDNA was detected from the patients' sera with HBV reactivation but not from the patients' sera without HBV reactivation. The level of cccDNA in the sera was correlated with ALT and viral load in the HBV reactivation patients. CONCLUSION: HBV cccDNA is actively transcribed and replicated in some patients' hepatocytes, which is reflected by a high ratio of HBV total DNA vs cccDNA. Detection of cccDNA in the liver biopsy will provide an end-point for the anti-HBV therapy. The occurrence of cccDNA in the sera is an early signal of liver damage, which may be another important clinical parameter.


Subject(s)
Hepatitis B virus/genetics , Hepatitis B, Chronic/pathology , Hepatitis B, Chronic/virology , Liver/pathology , Alanine Transaminase/blood , Biopsy , DNA, Circular/blood , DNA, Viral/blood , Hepatocytes/pathology , Hepatocytes/virology , Humans , Kinetics , Liver/virology , Viral Load
14.
FEMS Microbiol Lett ; 211(2): 259-64, 2002 Jun 04.
Article in English | MEDLINE | ID: mdl-12076822

ABSTRACT

2-Haloacid dehalogenases are hydrolytic enzymes that cleave the halogen-carbon bond(s) in haloalkanoic acids. We have previously isolated a cryptic haloacid dehalogenase gene from Burkholderia cepacia MBA4 and expressed it in Escherichia coli. This recombinant protein is unusual in having a long leader sequence, a property of periplasmic enzymes. In this paper, we report the functional role of this leader sequence. Western blot analyses showed that Chd1 is translocated to the periplasm. The results on the expression of Chd1 in the presence of sodium azide suggested the cleavage of the leader to be Sec-dependent. Chimeras of Chd1 and green fluorescent protein demonstrated that the leader sequence is fully functional in translocating the fusion protein to the periplasm. The expression of the chimeras in Sec mutants supported the Sec-dependent translocation. Surprisingly, recombinant Chd1 and a chimera with no leader sequence were also found in the periplasm.


Subject(s)
Adenosine Triphosphatases/physiology , Bacterial Proteins/metabolism , Burkholderia cepacia/enzymology , Escherichia coli Proteins/physiology , Escherichia coli/genetics , Hydrolases/metabolism , Membrane Transport Proteins/physiology , Adenosine Triphosphatases/genetics , Bacterial Proteins/genetics , Burkholderia cepacia/genetics , Cloning, Molecular , DNA, Bacterial/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Hydrolases/genetics , Membrane Transport Proteins/genetics , Plasmids , Protein Transport , Recombinant Proteins/biosynthesis , SEC Translocation Channels , SecA Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...