Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Gene Ther ; 29(7-8): 407-417, 2022 08.
Article in English | MEDLINE | ID: mdl-33414522

ABSTRACT

Camptothecin has been used in tumor therapy for a long time but its antitumor effect is rather limited due to the side effect and the drug resistance. FEN1, a major component of DNA repair systems, plays important roles in maintaining genomic stability via DNA replication and repair. Here we found that FEN1 inhibitor greatly sensitizes cancer cells to low-dose camptothecin. The combinative treatment of FEN1 inhibitor and 1 nM camptothecin induced a synthetic lethal effect, which synergistically suppressed cancer cell proliferation and significantly mediated apoptosis both in vitro and in vivo. Our study suggested that targeting FEN1 could be a potent strategy for tumor-targeting cancer therapy.


Subject(s)
Camptothecin , Flap Endonucleases , Neoplasms , Apoptosis , Camptothecin/pharmacology , DNA Damage , Flap Endonucleases/antagonists & inhibitors , Humans , Mitochondria/metabolism
2.
J Pathol ; 235(5): 672-85, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25385233

ABSTRACT

G protein-regulated cell function is crucial for cardiomyocytes, and any deregulation of its gene expression or protein modification can lead to pathological cardiac hypertrophy. Herein, we report that protein prenylation, a lipidic modification of G proteins that facilitates their association with the cell membrane, might control the process of cardiomyocyte hypertrophy. We found that geranylgeranyl diphosphate synthase (GGPPS), a key enzyme involved in protein prenylation, played a critical role in postnatal heart growth by regulating cardiomyocyte size. Cardiac-specific knockout of GGPPS in mice led to spontaneous cardiac hypertrophy, beginning from week 4, accompanied by the persistent enlargement of cardiomyocytes. This hypertrophic effect occurred by altered prenylation of G proteins. Evaluation of the prenylation, membrane association and hydrophobicity showed that Rheb was hyperactivated and increased mTORC1 signalling pathway after GGPPS deletion. Protein farnesylation or mTORC1 inhibition blocked GGPPS knockdown-induced mTORC1 activation and suppressed the larger neonatal rat ventricle myocyte size and cardiomyocyte hypertrophy in vivo, demonstrating a central role of the FPP-Rheb-mTORC1 axis for GGPPS deficiency-induced cardiomyocyte hypertrophy. The sustained cardiomyocyte hypertrophy progressively provoked cardiac decompensation and dysfunction, ultimately causing heart failure and adult death. Importantly, GGPPS was down-regulated in the hypertrophic hearts of mice subjected to transverse aortic constriction (TAC) and in failing human hearts. Moreover, HPLC-MS/MS detection revealed that the myocardial farnesyl diphosphate (FPP):geranylgeranyl diphosphate (GGPP) ratio was enhanced after pressure overload. Our observations conclude that the alteration of protein prenylation promotes cardiomyocyte hypertrophic growth, which acts as a potential cause for pathogenesis of heart failure and may provide a new molecular target for hypertrophic heart disease clinical therapy.


Subject(s)
Cardiomegaly/enzymology , Farnesyltranstransferase/deficiency , Heart Failure/enzymology , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , Myocytes, Cardiac/enzymology , Neuropeptides/metabolism , Protein Prenylation , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Cardiomegaly/drug therapy , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Line , Disease Models, Animal , Disease Progression , Farnesyltranstransferase/genetics , Female , Heart Failure/drug therapy , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/genetics , Multiprotein Complexes/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Neuropeptides/genetics , Protein Kinase Inhibitors/pharmacology , Protein Prenylation/drug effects , RNA Interference , Ras Homolog Enriched in Brain Protein , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Time Factors , Transfection , Ventricular Function, Left
3.
EMBO J ; 30(18): 3754-65, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21829168

ABSTRACT

It is well known that insulin can activate both PI3K/Akt pathway, which is responsible for glucose uptake, and MAPK pathway, which is crucial for insulin resistance formation. But, it is unclear exactly how the two pathways coordinate to regulate insulin sensitivity upon hyperinsulinism stress of type 2 diabetes mellitus (T2DM). Here, we show that an early response transcription factor Egr-1 could tilt the signalling balance by blocking PI3K/Akt signalling through PTEN and augmenting Erk/MAPK signalling through GGPPS, resulting in insulin resistance in adipocytes. Egr-1, PTEN and GGPPS are upregulated in the fat tissue of T2DM patients and db/db mice. Egr-1 overexpression in epididymal fat induced systematic insulin resistance in wild-type mice, and loss of Egr-1 function improved whole-body insulin sensitivity in diabetic mice, which is mediated by Egr-1 controlled PI3K/Akt and Erk/MAPK signalling balance. Therefore, we have revealed, for the first time, the mechanism by which Egr-1 induces insulin resistance under hyperinsulinism stress, which provides an ideal pharmacological target since inhibiting Egr-1 can simultaneously block MAPK and augment PI3K/Akt activation during insulin stimulation.


Subject(s)
Adipocytes/metabolism , Early Growth Response Protein 1/metabolism , Farnesyltranstransferase/metabolism , Gene Expression Regulation , Insulin Resistance , Insulin/metabolism , Multienzyme Complexes/metabolism , PTEN Phosphohydrolase/metabolism , Animals , Cell Line , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
4.
Biochem Cell Biol ; 91(5): 287-94, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24032677

ABSTRACT

Polo-like kinase 1 (Plk1) is a conserved key regulator of the G2/M transition, but its upstream spatiotemporal regulators remain unknown. With the help of immunofluorescence, co-immunoprecipitation, and glutathione S-transferase (GST) pull-down assay, we found that calmodulin (CaM) is one such regulatory molecule that associates with Plk1 from G2 to metaphase. More importantly, this interaction results in considerable stimulation of Plk1 kinase activity leading to hyperphosphorylation of Cdc25C. Our results provide new insight into the role of CaM as an upstream regulator of Plk1 activation during mitotic entry.


Subject(s)
Calmodulin/metabolism , Cell Cycle Proteins/metabolism , Cell Division/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , cdc25 Phosphatases/metabolism , Cell Line , Centrosome/metabolism , Enzyme Activation , G2 Phase , HEK293 Cells , HeLa Cells , Humans , Mitosis , Phosphorylation , Signal Transduction/genetics , Polo-Like Kinase 1
5.
BMC Med Genomics ; 16(1): 111, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37217926

ABSTRACT

BACKGROUND: Xq22.1-q22.3 deletion is a rare chromosome aberration. The purpose of this study was to identify the correlation between the phenotype and genotype of chromosome Xq22.1-q22.3 deletions. METHODS: Chromosome aberrations were identified by copy number variation sequencing (CNV-seq) technology and karyotype analysis. Furthermore, we reviewed patients with Xq22.1-q22.3 deletions or a deletion partially overlapping this region to highlight the rare condition and analyse the genotype-phenotype correlations. RESULTS: We described a female foetus who is the "proband" of a Chinese pedigree and carries a heterozygous 5.29 Mb deletion (GRCh37: chrX: 100,460,000-105,740,000) in chromosome Xq22.1-q22.3, which may affect 98 genes from DRP2 to NAP1L4P2. This deletion encompasses 7 known morbid genes: TIMM8A, BTK, GLA, HNRNPH2, GPRASP2, PLP1, and SERPINA7. In addition, the parents have a normal phenotype and are of normal intelligence. The paternal genotype is normal. The mother carries the same deletion in the X chromosome. These results indicate that the foetus inherited this CNV from her mother. Moreover, two more healthy female family members were identified to carry the same CNV deletion through pedigree analysis according to the next-generation sequencing (NGS) results. To our knowledge, this family is the first pedigree to have the largest reported deletion of Xq22.1-q22.3 but to have a normal phenotype with normal intelligence. CONCLUSIONS: Our findings further improve the understanding of the genotype-phenotype correlations of chromosome Xq22.1-q22.3 deletions.This report may provide novel information for prenatal diagnosis and genetic counselling for patients who carry similar chromosome abnormalities.


Subject(s)
Chromosome Aberrations , DNA Copy Number Variations , Pregnancy , Female , Humans , Pedigree , Phenotype , Chromosome Deletion , Chromosomes , Mitochondrial Precursor Protein Import Complex Proteins
6.
World J Clin Cases ; 11(26): 6206-6212, 2023 Sep 16.
Article in English | MEDLINE | ID: mdl-37731580

ABSTRACT

BACKGROUND: Patients with trisomy 8 consistently present with myeloid neoplasms and/or auto-inflammatory syndrome. A possible link between myelodysplastic syndromes (MDS) with trisomy 8 (+8-MDS) and inflammatory disorders is well recognized, several cases having been reported. However, inflammatory disorders in patients without MDS have been largely overlooked. Generally, Behçet's disease is the most common type in +8-MDS. However, inflammatory disorders with pulmonary involvement are less frequent, and no effective treatment has been established. CASE SUMMARY: A 27-year-old man with recurrent fever, fatigue for > 2 mo, and unconsciousness for 1 day was admitted to our emergency department with a provisional diagnosis of severe pneumonia. Vancomycin and imipenem were administered and sputum collected for metagenomic next-generation sequencing. Epstein-Barr virus and Mycobacterium kansasii were detected. Additionally, chromosomal analysis showed duplications on chromosome 8. Two days later, repeat metagenomic next-generation sequencing was performed with blood culture. Cordyceps portugal, M. kansasii, and Candida portugal were detected, and duplications on chromosome 8 confirmed. Suspecting hematological disease, we aspirated a bone marrow sample from the iliac spine, examination of which showed evidence of infection. We added fluconazole as further antibiotic therapy. Seven days later, the patient's condition had not improved, prompting addition of methylprednisolone as an anti-inflammatory agent. Fortunately, this treatment was effective and the patient eventually recovered. CONCLUSION: Severe inflammatory disorders with pulmonary involvement can occur in patients with trisomy 8. Methylprednisolone may be an effective treatment.

7.
J Biol Chem ; 286(16): 14508-15, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21321112

ABSTRACT

One of the most important characteristics of type 2 diabetes is insulin resistance, during which the patients normally experienced hyperinsulinism stress that would alter insulin signal transduction in insulin target tissues. We have previously found that early growth responsive gene-1 (Egr-1), a zinc finger transcription factor, is highly expressed in db/db mice and in the fat tissue of individuals with type 2 diabetes. In this report, we found that chronic exposure to hyperinsulinism caused persistent Erk/MAPK activity in adipocytes and enhanced insulin resistance in an Egr-1-dependent manner. An elevation in Egr-1 augmented Erk1/2 activation via geranylgeranyl diphosphate synthase (GGPPS). Egr-1-promoted GGPPS transcription increased Ras prenylation and caused Erk1/2 activation. The sustained activation of Erk1/2 resulted in the phosphorylation of insulin receptor substrate-1 at Serine 612. Phosphorylation at this site impaired insulin signaling in adipocytes and reduced glucose uptake. The loss of Egr-1 function, knockdown of GGPPS, or inhibition of Erk1/2 activity in insulin-resistant adipocytes restored insulin receptor substrate-1 tyrosine phosphorylation and increased insulin sensitivity. Our results suggest a new mechanism by which the Egr-1/GGPPS/Erk1/2 pathway is responsible for insulin resistance during hyperinsulinism. This pathway provides a new therapeutic target for increasing insulin sensitivity: inhibiting the function of Egr-1.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Early Growth Response Protein 1/metabolism , Hyperinsulinism/metabolism , 3T3 Cells , Adipocytes/cytology , Animals , Dimethylallyltranstransferase/chemistry , Farnesyltranstransferase/chemistry , Geranyltranstransferase/chemistry , Humans , Insulin/metabolism , Insulin Resistance , MAP Kinase Signaling System , Male , Mice , Phosphorylation , Transcription, Genetic
8.
Am J Pathol ; 179(6): 2740-50, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21983072

ABSTRACT

Cigarette smoke activates the extracellular signal-regulated kinase (ERK) 1/2 mitogen activated-protein kinase pathway, which, in turn, is responsible for early growth response gene-1 (EGR-1) activation. Here we provide evidence that EGR-1 activation can also reactivate ERK 1/2 mitogen activated-protein kinase through a positive feedback loop through its target gene (geranylgeranyl diphosphate synthase) GGPPS. For the first time, the GGPPS gene is identified as a target of EGR-1, as EGR-1 can directly bind to the predicted consensus-binding site in the GGPPS promoter and regulate its transcription. Long-term observations show that there are two ERK 1/2 phosphorylation peaks after cigarette smoke extract stimulation in human lung epithelial Beas-2B cells. The first peak (at 10 minutes) is responsible for EGR-1 accumulation, and the second (at 4 hours) is diminished after the disruption of EGR-1 transcriptional activity. EGR-1 overexpression enhances Ras prenylation and membrane association in a GGPPS-dependent manner, and it augments ERK 1/2 activation. Likewise, a great reduction of the second peak of ERK 1/2 phosphorylation is observed during long-term cigarette smoke extract stimulation in cells where GGPPS is disrupted. Thus, we have uncovered an intricate positive feedback loop in which ERK 1/2-activated EGR-1 promotes ERK 1/2 reactivation through promoting GGPPS transcription, which might affect cigarette smoke-related lung pathological processes.


Subject(s)
Early Growth Response Protein 1/genetics , Farnesyltranstransferase/genetics , MAP Kinase Signaling System/genetics , Prenylation/genetics , Smoking/genetics , ras Proteins/metabolism , Animals , Cells, Cultured , Early Growth Response Protein 1/physiology , Feedback, Physiological , HEK293 Cells , Humans , Mice , Mice, Mutant Strains , Pneumonia/etiology , RNA, Small Interfering/pharmacology , Smoke , Smoking/adverse effects , Transcription, Genetic
9.
Am J Pathol ; 178(1): 110-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224049

ABSTRACT

Early growth response 1 (EGR-1) contributes to the development of chronic obstructive pulmonary disease in the lungs of smokers by mediating pulmonary inflammatory responses, but the direct downstream genes of EGR-1 that regulate this process remain unknown. We show that a new EGR-1 target gene, geranylgeranyl diphosphate synthase (GGPPS), which controls protein prenylation, can regulate the proinflammatory function of EGR-1 by activating MAPK signaling. When C57BL/6 mice were exposed to cigarette smoke, EGR-1 and GGPPS levels increased in their lungs, and the inflammatory responses were augmented, whereas these effects could be reversed by the down-regulation of EGR-1 transcription activity. The accumulation of EGR-1 and GGPPS was induced by MAPK/ERK pathway activation when Beas-2B human bronchial epithelial cells were exposed to cigarette smoke extract (CSE). Further examination showed that EGR-1 in turn regulated Erk1/2 activity because inhibition of EGR-1 transcription activity decreased CSE-induced Erk1/2 phosphorylation. Furthermore, EGR-1-promoted Erk1/2 activation was dependent on GGPPS transcription. Knockdown of GGPPS expression with small-interfering RNA abolished the EGR-1-activated Erk1/2 activity. Both EGR-1 transcription inhibition and GGPPS expression knockdown decreased the inflammatory response induced by CSE in Beas-2B cells. Our results reveal a new EGR-1/GGPPS/MAPK signaling pathway that controls cigarette smoke-induced pulmonary inflammation, and this may shed light on our understanding of the mechanism of cigarette smoke-related pulmonary diseases such as chronic obstructive pulmonary disease.


Subject(s)
Early Growth Response Protein 1/metabolism , Farnesyltranstransferase/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nicotiana/adverse effects , Pulmonary Disease, Chronic Obstructive/genetics , Smoking/adverse effects , Animals , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive/pathology , Signal Transduction
10.
Histochem Cell Biol ; 134(3): 285-95, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20677010

ABSTRACT

It is well accepted that cell scattering (dispersion of clustered cells into single cells) is the initial step of tumor metastasis, and the downregulation of E-cadherin is associated with metastatic potential of tumor cells; however, the molecular mechanisms underlying loss of E-cadherin during tumor development are still poorly understood. Here, we report that hepatocyte growth factor (HGF) induced E-cadherin downregulation and cell scattering are attributed to the activation of Wnt/beta-catenin signaling and transcriptional activation of matrix metalloproteinase MMP-7. Furthermore, the increased MMP-7 is secreted into the medium and cleaves the ectodomain of E-cadherin. Inhibition of HGF signal by siRNA of c-Met, blocking the beta-catenin transcriptional activity through a dominant negative form of TCF4, MMP-7 knockdown by siRNA or suppression of MMP-7 enzymatic activity with a neutralization antibody allowed inhibition of HGF-induced loss of E-cadherin and HepG2 scattering. Our data presented here revealed the intrinsic mechanism of HGF activated Wnt/beta-catenin signaling regulation of HepG2 cell scattering through MMP-7 transcription activation and E-cadherin degradation. The results suggest that the blocking of HGF/c-Met/beta-catenin/MMP-7/E-cadherin signaling pathway might present a practical therapeutic target for interference with hepatocellular carcinoma metastasis.


Subject(s)
Cadherins/biosynthesis , Hepatocyte Growth Factor/physiology , Matrix Metalloproteinase 7/metabolism , beta Catenin/physiology , Down-Regulation , Enzyme Activation , Hep G2 Cells , Humans , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Proto-Oncogene Proteins c-met/physiology , Signal Transduction/drug effects , Wnt Proteins/physiology
11.
BMC Med Genomics ; 13(1): 96, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32631433

ABSTRACT

BACKGROUND: Non-invasive prenatal testing (NIPT) has been established as a routine prenatal screening to assess the risk of common foetal aneuploidy disorder (trisomy 21, 18, and 13). NIPT has high sensitivity and high specificity, but false positive and false negative results still exist. False negative NIPT results involving Down syndrome are rare, but have a high clinical impact on families and society. CASE PRESENTATION: We described a case of a foetus that tested "negative" for trisomy 21 (Z-score was 0.664) by NIPT based on the semiconductor sequencing platform (SSP). The foetal fraction of cell-free DNA was 16.9%; this percentage was much larger than the threshold of 4% for obtaining accurate NIPT results. However, postnatally, the newborn was diagnosed with Down syndrome with the 46,XY,der(21;21)(q10;q10),+ 21 karyotype. CONCLUSIONS: We presented a case of false negative NIPT results, which may occur through biological mechanisms rather than poor quality, technical errors or negligence. It is imperative for clinical geneticists and their patients to understand that NIPT is still a screening test.


Subject(s)
Cell-Free Nucleic Acids/genetics , Chromosomes, Human, Pair 21/genetics , Down Syndrome/diagnosis , Fetus/metabolism , Gene Rearrangement , Prenatal Diagnosis/methods , Trisomy , Adult , Cell-Free Nucleic Acids/analysis , Down Syndrome/genetics , False Negative Reactions , Female , Humans , Infant, Newborn , Male , Pregnancy
12.
DNA Repair (Amst) ; 95: 102953, 2020 11.
Article in English | MEDLINE | ID: mdl-32861926

ABSTRACT

The up-regulation of PRMT1 is critical to the cell growth and cancer progression of lung cancer cells. In our research, we found that PRMT1 is important to the DNA repair ability and drug resistance of lung cancer cells. To demonstrate the functions of PRMT1, we identified Flap endonuclease 1 (FEN1) as a post-translationally modified downstream target protein of PRMT1. As a major component of Base Excision Repair pathway, FEN1 plays an important role in DNA replication and DNA damage repair. However, the detailed mechanism of FEN1 up-regulation in lung cancer cells remains unclear. In our study, we identified PRMT1 as a key factor that maintains the high expression levels of FEN1, which is critical to the DNA repair ability and the chemotherapeutic drug resistance of lung cancer cells.


Subject(s)
Drug Resistance, Neoplasm/genetics , Flap Endonucleases/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , A549 Cells , Apoptosis/genetics , Cell Proliferation/genetics , DNA Repair , Epigenesis, Genetic , Gene Knockdown Techniques , Humans , Protein-Arginine N-Methyltransferases/deficiency , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/deficiency , Repressor Proteins/genetics , Up-Regulation
13.
Mol Cancer Ther ; 18(12): 2407-2420, 2019 12.
Article in English | MEDLINE | ID: mdl-31467180

ABSTRACT

DNA repair mechanisms are crucial for cell survival. It increases the cancer cell's ability to resist DNA damage. FEN1 is involved in DNA replication and repair, specifically long-patch base excision repair. Although the gene function and post-translational modification of FEN1 are well studied, the regulatory mechanism of FEN1 by upstream signal pathways remains unclear. In this article, we have identified AKT as a regulator of FEN1 activity in lung cancer cells. Sustained activation of AKT can phosphorylate nuclear transcription factor NF-κB/p65. NF-κB/p65 directly binds to FEN1 promoter to promote a high transcription level of FEN1, revealing the contribution of the AKT signaling pathway to drug resistance of cancer cells. The combination of an AKT inhibitor and cisplatin efficiently suppressed lung cancer cell growth both in vitro and in vivo Our study illustrated an upstream regulatory mechanism of FEN1, which will contribute to the development of effective lung cancer therapies.These findings identified AKT as a regulator of FEN1 activity and revealed the AKT signaling pathway's contribution to drug resistance, which will contribute to the development of effective lung cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Flap Endonucleases/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Down-Regulation , Female , Humans , Mice , Mice, Nude
14.
FEBS Lett ; 580(15): 3624-30, 2006 Jun 26.
Article in English | MEDLINE | ID: mdl-16753148

ABSTRACT

Ultraviolet (UV) irradiation can result in cell cycle arrest. The reactivation of Polo-like kinase 1 (Plk1) is necessary for cell cycle reentry. But the mechanism of how Plk1 regulates p53 in UV-induced mitotic arrest cells remained elusive. Here we find that UV treatment leads HEK293 cells to inverse changes of Plk1 and p53. Over-expression of Plk1 rescue UV-induced mitotic arrest cells by inhibiting p53 activation. Plk1 could also inhibit p53 phosphorylation at Ser15, thus facilitates its nuclear export and degradation. Further examination shows that Plk1, p53 and Cdc25C can form a large complex. Plk1 could bind to the sequence-specific DNA-binding domain of p53 and active Cdc25C by hyperphosphorylation. These results hypothesize that Plk1 and Cdc25C participate in recovery the mitotic arrest through binding to the different domain of p53. Cdc25C may first be actived by Plk1, and then its phosphatase activity makes p53 dephosphorylated at Ser15.


Subject(s)
Cell Cycle Proteins/metabolism , Mitosis/radiation effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle Proteins/genetics , Cell Line , DNA Damage/genetics , Humans , Phosphorylation/radiation effects , Phosphoserine/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Protein Transport , Proto-Oncogene Proteins/genetics , cdc25 Phosphatases/metabolism , Polo-Like Kinase 1
15.
Mol Cancer Ther ; 4(10): 1577-84, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16227408

ABSTRACT

c-Met is highly expressed and constitutively activated in various human tumors. We employed adenovirus-mediated RNA interference technique to knock down c-Met expression in hepatocellular carcinoma cells and observed its effects on hepatocellular carcinoma cell growth in vitro and in vivo. Among the five hepatocellular carcinoma and one normal human liver cell lines we analyzed, c-Met was highly expressed and constitutively tyrosine phosphorylated in only MHCC97-L and HCCLM3 hepatocellular carcinoma cells. Knockdown of c-Met could inhibit MHCC97-L cells proliferation by arresting cells at G0-G1 phase. Soft agar colony formation assay indicated that the colony forming ability of MHCC97-L cells decreased by approximately 70% after adenovirus AdH1-small interfering RNA (siRNA)/met infection. In vivo experiments showed that adenovirus AdH1-siRNA/met inhibited the tumorigenicity of MHCC97-L cells and significantly suppressed tumor growth when injected directly into tumors. These results suggest that knockdown of c-Met by adenovirus-delivered siRNA may be a potential therapeutic strategy for treatment of hepatocellular carcinoma in which c-Met is overexpressed.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , RNA, Small Interfering/genetics , Adenoviridae/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line , Cell Line, Tumor , Flow Cytometry , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation , Proto-Oncogene Proteins c-met/biosynthesis , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Cell Res ; 15(7): 548-52, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16045818

ABSTRACT

A transient postanaphase repositioning of the centriole is found to control the completion of cytokinesis. Using a green fluorescent protein-calmodulin fusion protein as a living cell probe, we have previously found that calmodulin is associated with the initiation and progression of cytokinesis. In this study, we further studied the effect of calmodulin on the repositioning of the centriole and subsequent cell cycle progression. When activity of calmodulin is inhibited, the regression of the centriole from the intercellular bridge to the cell center is blocked, and thus the completion of cell division is repressed and two daughter cells are linked by longer cell bridge in perturbed cells. W7 treatment during cytokinesis also results in unfinished cytokinesis and stopped G1 phase. These results suggest that calmodulin activity is required for centriole repositioning and can affect the completion of cytokinesis and cell cycle progression.


Subject(s)
Anaphase/physiology , Calmodulin/pharmacology , Centrioles/physiology , Cytokinesis/physiology , Anaphase/drug effects , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Line , Centrioles/drug effects , Cytokinesis/drug effects , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/pharmacology , HeLa Cells , Humans , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Sulfonamides/pharmacology , Time Factors
17.
Anat Rec (Hoboken) ; 294(6): 1025-34, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21542139

ABSTRACT

Histone acetylation is important for gene transcription, which is controlled by the balance between two kinds of opposing enzymes: histone acetyltransferases and histone deacetylases (HDACs). HDACs repress gene transcription by decreasing histone acetylation levels. Our hypothesis was that shuttling of Class II HDACs, such as HDAC4, between the nucleus and cytoplasm is critical for its function. We constructed mutants of mammalian HDAC4 that had different cellular locations and checked their function during memory formation using Caenorhabditis elegans as a model. The deletion of hda4, a homolog of HDAC4, was able to enhance learning and long-term memory (LTM) in a thermotaxis model. Transgenic experiments showed that mammalian wild-type HDAC4 rescued the phenotype of hda4-deleted worms but impaired LTM formation in wild-type worms. The cytosol-localized HDAC4 mutant was not able to alter the phenotype of knock-out worms but led to enhanced LTM formation in wild-type worms similar to hda4-deletion mutants. Constitutive nuclear localization of HDAC4 rescued the phenotype of deletion worms similar to wild-type HDAC4 but had no effect on wild-type worms. These results support our hypothesis that HDAC4's biological function is regulated by its intracellular distribution.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Histone Deacetylases/metabolism , Intracellular Fluid/enzymology , Memory, Long-Term/physiology , Repressor Proteins/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Cell Nucleus/enzymology , Histone Deacetylases/physiology , Humans , Protein Transport/physiology , Repressor Proteins/physiology , Structural Homology, Protein
18.
Anat Rec (Hoboken) ; 293(3): 414-20, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20169563

ABSTRACT

The hypoxic environment of solid tumor causes the tumor cells survive and which could protect them from death by facilitating resistance to therapy. Here, we provide evidence that hypoxia can increase tumor cell viability and proliferation through an Egr-1-dependant pathway. Hypoxia protected the microtubules from disassembly, and Egr-1 was colocalized with microtubules in different cell cycle stages. Knockdown of Egr-1 with its siRNA overcame the protection effect of hypoxia and increased the sensitivity of tumor cells to vinblastine under hypoxic conditions. Our results suggest a novel approach for increasing the sensitivity of tumor cells to chemotherapeutics that target microtubule assembly.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Early Growth Response Protein 1/metabolism , Hypoxia/metabolism , Microtubules/metabolism , Vinblastine/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/genetics , Fluorescent Antibody Technique , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured
19.
Anat Rec (Hoboken) ; 292(6): 818-26, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19462446

ABSTRACT

beta-catenin functions as both a structural protein and a transcriptional activator. In this study, we examined the expression of beta-catenin in human cirrhotic livers, and administered adenoviruses carrying the beta-catenin or DeltaTCF4 genes to cirrhotic rats to investigate the role of beta-catenin in the development of liver cirrhosis development. beta-catenin expression was associated with liver cirrhosis development in cirrhotic human and rat liver. beta-catenin adenovirus was capable of accelerating cirrhosis progress but this progression was unaffected by administration of DeltaTCF4 adenovirus. beta-catenin was mainly located in the intercellular regions between liver cells and was highly concentrated in the hepatic sinusoid wall, where alpha-smooth muscle actin (SMA) was also mainly distributed. The binding of beta-catenin to alpha-SMA was also increased in cirrhotic liver. Portal vein blood pressure was significantly increased in the group administered beta-catenin adenovirus, but not in that receiving DeltaTCF4 adenovirus. These results suggest that high concentrations of beta-catenin at the hepatic intercellular membrane and the hepatic sinusoid wall contribute to hepatic hyperpiesia in liver cirrhosis patients. beta-catenin functions as a structural molecule, but not as a signaling molecule, during liver cirrhosis development.


Subject(s)
Hypertension, Portal/metabolism , Liver Cirrhosis/metabolism , beta Catenin/metabolism , Adenoviridae/genetics , Animals , Carbon Tetrachloride/toxicity , Disease Models, Animal , Disease Progression , Female , Humans , Hypertension, Portal/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Male , Middle Aged , Rats , Rats, Sprague-Dawley , beta Catenin/genetics
20.
Cell Stress Chaperones ; 14(4): 343-53, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18989758

ABSTRACT

The cell cycle is controlled by regulators functioning at the right time and at the right place. We have found that calmodulin (CaM) has specific distribution patterns during different cell-cycle stages. Here, we identify cell-cycle-specific binding proteins of CaM and examine their function during cell-cycle progression. We first applied immunoprecipitation methods to isolate CaM-binding proteins from cell lysates obtained at different cell-cycle phases and then identified these proteins using mass spectrometry methods. A total of 41 proteins were identified including zinc finger proteins, ribosomal proteins, and heat shock proteins operating in a Ca(2+)-dependent or independent manner. Fifteen proteins were shown to interact with CaM in a cell-phase-specific manner. The association of the selected proteins and CaM were confirmed with in vitro immunoprecipitation and immunostaining methods. One of the identified proteins, heat shock protein 70 (Hsp70), was further studied with respect to its cell-cycle-related function. In vivo fluorescence resonance energy transfer (FRET) analysis showed that the interaction of CaM and Hsp70 was found in the nucleus during the S phase. Overexpression of Hsp70 is shown to arrest cells at S phase and, thus, induce cell apoptosis. When we disrupted the CaM-Hsp70 association with HSP70 truncation without the CaM-binding domain, we found that S-phase arrest and apoptosis could be rescued. The results suggest that the spatial and temporal association of CaM and Hsp70 can regulate cell-cycle progression and cell apoptosis.


Subject(s)
Apoptosis , Calmodulin/metabolism , HSP70 Heat-Shock Proteins/metabolism , Calmodulin/analysis , Cell Line , Fluorescence Resonance Energy Transfer , HSP70 Heat-Shock Proteins/analysis , Humans , Immunoprecipitation , Protein Binding , Protein Structure, Tertiary , S Phase
SELECTION OF CITATIONS
SEARCH DETAIL