Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Bioorg Chem ; 120: 105634, 2022 03.
Article in English | MEDLINE | ID: mdl-35114524

ABSTRACT

Novel (Z)-3-((4,6-diphenylpyrimidin-2-ylamino)methylene)-2,3-dihydrochromen-4-one derivatives were designed and synthesized to find chemotherapeutic agents. Derivative 9 was selected based on its clonogenicity against cancer cells and synthetic yield for further biological experiments. It showed decreases in aurora kinase A, B, and C phosphorylation from western blot analysis. Derivative 9 upregulated the expression of G1 cell cycle inhibitory proteins including p21 and p27, and G1 progressive cyclin D1, and downregulated G1-to-S progressive cyclins, resulting in cell cycle arrest at the G1/S boundary. It stimulated the cleavage of caspase-9, -3, -7, and poly (ADP-ribose) polymerase, resulting in triggering apoptosis through a caspase-dependent pathway. In addition, derivative 9 inhibited in vivo tumor growth in a syngeneic tumor implantation mouse model. The findings of this study suggest that derivative 9 can be considered as a lead compound for chemotherapeutic agents.


Subject(s)
Antineoplastic Agents , Caspases , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Caspases/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Mice , Poly(ADP-ribose) Polymerases/metabolism
2.
J Nat Prod ; 83(1): 118-126, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31894983

ABSTRACT

Ohmyungsamycin A (1), a novel cyclic peptide discovered from a marine Streptomyces sp., was previously reported with antibacterial and anticancer activities. However, the antitumor activities and the underlying molecular mechanisms of 1 remain to be elucidated. Compound 1 inhibited the proliferation and tumor growth of HCT116 human colorectal cancer cells based on both in vitro cell cultures and an in vivo animal model. A cDNA microarray analysis revealed that 1 downregulated genes involved in cell cycle checkpoint control. Compound 1 also induced G0/G1 cell cycle arrest that was mediated by the regulation of S-phase kinase-associated protein 2 (Skp2)-p27 axis and minichromosome maintenance protein 4 (MCM4). Furthermore, a longer exposure of 1 exhibited an accumulation of a sub-G1 phase cell population, which is characteristic of apoptotic cells. The induction of apoptosis by 1 was also associated with the modulation of caspase family proteins. Compound 1 effectively suppressed tumor growth in a xenograft mouse model subcutaneously implanted with HCT116 cells. In addition, analysis of tumors revealed that 1 upregulated the expression of the CDK inhibitor p27 but downregulated the expression of Skp2 and MCM4. These findings demonstrate the involvement of 1 in cell cycle regulation and the induction of apoptosis in human colorectal cancer cells.


Subject(s)
Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Minichromosome Maintenance Complex Component 4/metabolism , Peptides, Cyclic/pharmacology , S-Phase Kinase-Associated Proteins/metabolism , Animals , Apoptosis , Cell Cycle , Colorectal Neoplasms , Cyclin-Dependent Kinase Inhibitor p27/genetics , Humans , Mice , Minichromosome Maintenance Complex Component 4/genetics , Molecular Structure , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/genetics , Up-Regulation
3.
Braz J Med Biol Res ; 52(5): e8412, 2019.
Article in English | MEDLINE | ID: mdl-31038581

ABSTRACT

Multiple myeloma (MM) is a malignant neoplasm of plasma, and exhibits several harmful effects including osteolytic injuries, hypercalcemia, and immune dysfunction. Many patients with MM succumb to the underlying malignancy. An S-phase kinase-related protein 2 (Skp2) inhibitor, designated SKPin C1, has been developed and confirmed to have an inhibitory effect on metastatic melanoma cells. This study aimed to determine the effect of SKPin C1 on MM. Normal B lymphocytes, THP-1 cells, and MM U266 and RPMI 8226 cells were exposed to various dosages of SKPin C1 for 48 h. Cell proliferation was determined by MTT, EdU staining, and cell cycle assays. Western blot assays were performed to assess intracellular protein levels of Skp2, p27, and cleaved caspase-3. The amount of ubiquitin attached to p27 was determined using an immunoprecipitation assay. The viability of U266 and RPMI 8226 cells was significantly inhibited by 10 µM SKPin C1 and the inhibitory effect was enhanced with increasing doses of SKPin C1. In contrast, 50 µM SKPin C1 only marginally decreased viability of normal B lymphocytes in 12 h. Skp2 and p27 expression in U266 and RPMI 8226 cells was higher and lower, respectively, than that in the normal B lymphocytes. Treatment with SKPin C1 or Skp2 knockdown increased p27 protein levels in U266 and RPMI 8226 cells by preventing p27 from being ubiquitinated, which slowed the cell cycle, inhibited cell proliferation, and triggered apoptosis. Therefore, this study suggested SKPin C1 as a potent inhibitor against aberrant proliferation and immortalization of MM.


Subject(s)
Apoptosis , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Multiple Myeloma/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Cell Cycle , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Humans , Multiple Myeloma/physiopathology , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , Ubiquitinated Proteins/metabolism , Ubiquitination/physiology
4.
Braz. j. med. biol. res ; 52(5): e8412, 2019. graf
Article in English | LILACS | ID: biblio-1001528

ABSTRACT

Multiple myeloma (MM) is a malignant neoplasm of plasma, and exhibits several harmful effects including osteolytic injuries, hypercalcemia, and immune dysfunction. Many patients with MM succumb to the underlying malignancy. An S-phase kinase-related protein 2 (Skp2) inhibitor, designated SKPin C1, has been developed and confirmed to have an inhibitory effect on metastatic melanoma cells. This study aimed to determine the effect of SKPin C1 on MM. Normal B lymphocytes, THP-1 cells, and MM U266 and RPMI 8226 cells were exposed to various dosages of SKPin C1 for 48 h. Cell proliferation was determined by MTT, EdU staining, and cell cycle assays. Western blot assays were performed to assess intracellular protein levels of Skp2, p27, and cleaved caspase-3. The amount of ubiquitin attached to p27 was determined using an immunoprecipitation assay. The viability of U266 and RPMI 8226 cells was significantly inhibited by 10 μM SKPin C1 and the inhibitory effect was enhanced with increasing doses of SKPin C1. In contrast, 50 μM SKPin C1 only marginally decreased viability of normal B lymphocytes in 12 h. Skp2 and p27 expression in U266 and RPMI 8226 cells was higher and lower, respectively, than that in the normal B lymphocytes. Treatment with SKPin C1 or Skp2 knockdown increased p27 protein levels in U266 and RPMI 8226 cells by preventing p27 from being ubiquitinated, which slowed the cell cycle, inhibited cell proliferation, and triggered apoptosis. Therefore, this study suggested SKPin C1 as a potent inhibitor against aberrant proliferation and immortalization of MM.


Subject(s)
Humans , Apoptosis , S-Phase Kinase-Associated Proteins/metabolism , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Multiple Myeloma/metabolism , Cell Cycle , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Ubiquitination/physiology , Ubiquitinated Proteins/metabolism , Multiple Myeloma/physiopathology
5.
Breast Cancer ; 24(5): 658-666, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28070831

ABSTRACT

BACKGROUND: Prolyl oligopeptidase (POP, EC 3.4.1.26) is a serine peptidase that hydrolyzes post-proline peptide bonds in peptides that are <30 amino acids in length. We previously reported that POP inhibition suppressed the growth of NB-1 human neuroblastomas cells and KATO III human gastric cancer cells. POP activity is commonly elevated in many cancers, which includes breast cancer. However, the effect of POP inhibition as a candidate breast cancer therapy is unknown. METHODS: The effects of POP inhibition and knockdown on the proliferation of cultured human estrogen receptor-positive (ER+) MCF7 and T47D, and ER-negative (ER-) MDA-MB-231 breast cancer cell lines and the MCF12A non-tumorigenic epithelial cell line were tested by analyzing their influence on cell proliferation (WST-1 assay), cell viability (trypan blue exclusion assay), and cell cycle arrest (cell cycle analysis, cell cycle regulator proteins expression). RESULTS: POP-specific inhibitors 3-({4-[2-(E)-styrylphenoxy]butanoyl}-L-4-hydroxyprolyl)-thiazolidine (SUAM-14746) and benzyloxycarbonyl-thiopropyl-thioprolinal and RNAi-mediated POP knockdown inhibited the proliferation of MCF7 cells without inducing cell death. SUAM-14746-induced growth inhibition was also observed in T47D and MDA-MB-231 cells, but not in MCF12A cells. This growth inhibition was associated with G1 phase arrest; reduced cyclin D1 and D3, cyclin-dependent kinase 4 (CDK4), E2F1, and retinoblastoma protein (pRb) expression; and increased cyclin-dependent kinase inhibitor 1B (p27kip1) expression. Moreover, the SUAM-14746-mediated cell cycle arrest of MCF7 cells was associated with increased pRb2/p130 protein expression and an increase in the number of cells in the quiescent G0 state, as defined by low RNA levels. CONCLUSIONS: SUAM-14746 inhibited breast cancer cell growth in a cytostatic manner without inducing lethality, and POP-specific inhibitors may be an effective treatment against ER+ and ER- breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Proline/analogs & derivatives , Serine Endopeptidases/metabolism , Thiazolidines/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin D3/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , E2F1 Transcription Factor/metabolism , Female , Gene Knockdown Techniques , Humans , Proline/pharmacology , Proline/therapeutic use , Prolyl Oligopeptidases , Protease Inhibitors/therapeutic use , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism , Retinoblastoma Binding Proteins/metabolism , Serine Endopeptidases/genetics , Thiazolidines/therapeutic use , Ubiquitin-Protein Ligases/metabolism
6.
J Microbiol Biotechnol ; 25(3): 413-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25674801

ABSTRACT

Recently, we isolated HY253, a novel decahydrofluorene analog with a molecular structure of 7,8a-divinyl-2,4a,4b,5,6,7,8,8a,9,9a-decahydro-1H-fluorene-2,4a,4b,9a-tetraol from the roots of Aralia continentalis, which is known as Dokwhal, a traditional medicinal herb. Moreover, we previously reported its cytotoxic activity on cancer cell proliferation in human lung cancer A549 and cervical cancer HeLa cells. The current study aimed to evaluate its detailed molecular mechanisms in cell cycle arrest and apoptotic induction in human hepatocellular carcinoma HepG2 cells. Flow cytometric analysis of HepG2 cells treated with 60 micrometer HY253 revealed appreciable cell cycle arrest at the G1 phase via inhibition of Rb phosphorylation and down-regulation of cyclin D1. Furthermore, using western blots, we found that up-regulation of cyclin-dependent kinase inhibitors, such as p21(CIP1) and p27(KIP1), was associated with this G1 phase arrest. Moreover, TUNEL assay and immunoblottings revealed apoptotic induction in HepG2 cells treated with 100 micrometer HY253 for 24 h, which is associated with cytochrome c release from mitochondria, via down-regulation of anti-apoptotic Bcl-2 protein, which in turn resulted in activation of caspase-9 and -3, and proteolytic cleavage of poly(ADP-ribose) polymerase (PARP). Accordingly, we suggest that HY253 may be a potent chemotherapeutic hit compound for treating human liver cancer cells via up-regulation and activation of the p53 gene.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Fluorenes/pharmacology , Liver Neoplasms/drug therapy , Aralia/chemistry , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cytochromes c/metabolism , Flow Cytometry , G1 Phase/drug effects , Hep G2 Cells , Humans , Mitochondria/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Suppressor Protein p53/genetics
7.
J Neurooncol ; 118(1): 19-28, 2014 May.
Article in English | MEDLINE | ID: mdl-24595467

ABSTRACT

MicroRNAs are a family of small noncoding RNAs regulating gene expression by sequence-selective mRNA targeting, leading to a translational repression or mRNA degradation. The oncomiR miR-221 is highly expressed in human gliomas, as confirmed in this study in samples of low and high grade gliomas, as well in the cell lines U251, U373 and T98G. In order to alter the biological functions of miR-221, a peptide nucleic acid targeting miR-221 (R8-PNA-a221) was produced, bearing a oligoarginine peptide (R8) to facilitate uptake by glioma cells. The effects of R8-PNA-a221 were analyzed in U251, U373 and T98G glioma cells and found to strongly inhibit miR-221. In addition, the effects of R8-PNA-a221 on p27(Kip1) (a target of miR-221) were analyzed in U251 and T98G cells by RT-qPCR and by Western blotting. No change of p27(Kip1) mRNA content occurs in U251 cells in the presence of PNA-a221 (lacking the R8 peptide), whereas significant increase of p27(Kip1) mRNA was observed with the R8-PNA-a221. These data were confirmed by Western blot assay. A clear increment of p27(Kip1) protein expression in the samples treated with R8-PNA-a221 was detected. In addition, R8-PNA-a221 was found able to increase TIMP3 expression (another target of miR-221) in T98G cells. These results suggest that PNAs against oncomiRNA miR-221 might be proposed for experimental treatment of human gliomas.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/metabolism , MicroRNAs/metabolism , Peptide Nucleic Acids/pharmacology , Adult , Analysis of Variance , Annexin A5/metabolism , Apoptosis/drug effects , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry , Glioma/genetics , Humans , Male , MicroRNAs/genetics , Models, Molecular , Time Factors
8.
Mol Pharm ; 9(5): 1222-30, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22486588

ABSTRACT

Arginine-rich cell-penetrating peptides (CPPs), including oligoarginine peptides, have been widely used as a tool for intracellular delivery of various molecules with low membrane permeability. We previously reported the enhanced cytosolic entry of arginine-rich CPPs by the attachment of a short peptide segment, the penetration accelerating sequence (Pas). In this study, the importance of hydrophobic sequences, especially phenylalanine residues, in the Pas segment was demonstrated for this enhanced translocation through cell membranes. The advantage of using Pas for intracellular delivery was particularly marked for delivering cargoes with a relatively small molecular weight, such as bioactive peptides. In addition, the results of this study indicate the important roles that the total hydrophobicity of the PasR8 conjugates play in cytosolic translocation and the eventual bioactivity thus attained.


Subject(s)
Oligopeptides/chemistry , Peptide Fragments/chemistry , Arginine/chemistry , Cell Line, Tumor , Cell Survival , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions
9.
J Biol Chem ; 284(40): 27315-26, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19654316

ABSTRACT

Pericellular proteolysis by membrane-type 1 matrix metalloproteinase (MT1-MMP) plays a pivotal role in tumor cell invasion. Localization of MT1-MMP at the invasion front of cells, e.g. on lamellipodia and invadopodia, has to be regulated in coordination with reorganization of the actin cytoskeleton. However, little is known about how such invasion-related actin structures are regulated at the sites where MT1-MMP localizes. During analysis of MT1-MMP-associated proteins, we identified a heretofore uncharacterized protein. This protein, which we call p27RF-Rho, enhances activation of RhoA by releasing it from inhibition by p27(kip1) and thereby regulates actin structures. p27(kip1) is a well known cell cycle regulator in the nucleus. In contrast, cytoplasmic p27(kip1) has been demonstrated to bind GDP-RhoA and inhibit GDP-GTP exchange mediated by guanine nucleotide exchange factors. p27RF-Rho binds p27(kip1) and prevents p27(kip1) from binding to RhoA, thereby freeing the latter for activation. Knockdown of p27RF-Rho expression renders cells resistant to RhoA activation stimuli, whereas overexpression of p27RF-Rho sensitizes cells to such stimulation. p27RF-Rho exhibits a punctate distribution in invasive human tumor cell lines. Stimulation of the cells with lysophosphatidic acid induces activation of RhoA and induces the formation of punctate actin structures within foci of p27RF-Rho localization. Some of the punctate actin structures co-localize with MT1-MMP and cortactin. Down-regulation of p27RF-Rho prevents both redistribution of actin into the punctate structures and tumor cell invasion. Thus, p27RF-Rho is a new potential target for cancer therapy development.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Collagen/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Laminin/metabolism , Matrix Metalloproteinase 14/metabolism , Proteoglycans/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Conserved Sequence , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Drug Combinations , Enzyme Activation , Gene Expression Regulation , Guanosine Triphosphate/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Matrix Metalloproteinase 2/metabolism , Substrate Specificity , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors
10.
J Neurosci ; 29(3): 775-88, 2009 Jan 21.
Article in English | MEDLINE | ID: mdl-19158303

ABSTRACT

Although survival-promoting effects of insulin-like growth factor-1 (IGF-1) during neurogenesis are well characterized, mitogenic effects remain less well substantiated. Here, we characterize cell cycle regulators and signaling pathways underlying IGF-1 effects on embryonic cortical precursor proliferation in vitro and in vivo. In vitro, IGF-1 stimulated cell cycle progression and increased cell number without promoting cell survival. IGF-1 induced rapid increases in cyclin D1 and D3 protein levels at 4 h and cyclin E at 8 h. Moreover, p27(KIP1) and p57(KIP2) expression were reduced, suggesting downregulation of negative regulators contributes to mitogenesis. Furthermore, the phosphatidylinositol 3-kinase (PI3K)/Akt pathway specifically underlies IGF-1 activity, because blocking this pathway, but not MEK (mitogen-activated protein kinase kinase)/ERK (extracellular signal-regulated kinase), prevented mitogenesis. To determine whether mechanisms defined in culture relate to corticogenesis in vivo, we performed transuterine intracerebroventricular injections. Whereas blockade of endogenous factor with anti-IGF-1 antibody decreased DNA synthesis, IGF-1 injection stimulated DNA synthesis and increased the number of S-phase cells in the ventricular zone. IGF-1 treatment increased phospho-Akt fourfold at 30 min, cyclins D1 and E by 6 h, and decreased p27(KIP1) and p57(KIP2) expression. Moreover, blockade of the PI3K/Akt pathway in vivo decreased DNA synthesis and cyclin E, increased p27(KIP1) and p57(KIP2) expression, and prevented IGF-1-induced cyclin E mRNA upregulation. Finally, IGF-1 injection in embryos increased postnatal day 10 brain DNA content by 28%, suggesting a role for IGF-1 in brain growth control. These results demonstrate a mitogenic role for IGF-1 that tightly controls both positive and negative cell cycle regulators, and indicate that the PI3K/Akt pathway mediates IGF-1 mitogenic signaling during corticogenesis.


Subject(s)
Cell Cycle/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cyclins/metabolism , Insulin-Like Growth Factor I/pharmacology , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Bromodeoxyuridine/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Humans , In Vitro Techniques , Injections, Intraventricular/methods , Mice , Mice, Inbred C57BL , Neurons/drug effects , Pregnancy , Rats , Signal Transduction/drug effects , Thymidine/metabolism , Time Factors
11.
Mol Cancer Ther ; 7(5): 1164-75, 2008 May.
Article in English | MEDLINE | ID: mdl-18483304

ABSTRACT

The tumor suppressor gene CDKN1B encodes for a 27-kDa cyclin-dependent kinase inhibitory protein, p27Kip1, which together with its well-established role in the inhibition of cell proliferation, displays additional activities in the control of gene transcription and cell motility. p27Kip1 thus represents a good candidate for a gene therapy approach, especially in those cancers refractory to the conventional therapies, like human glioblastoma. Here, we show that overexpression of p27Kip1 in glioblastoma cell lines induced cell cycle arrest and inhibition of cell motility through extracellular matrix substrates. The use of adenoviral vectors in the treatment of glioblastoma in vivo showed that p27Kip1 was able to block not only cancer cell growth but also local invasion and tumor-induced neoangiogenesis. The latter effect was due to the ability of p27 to impair both endothelial cell growth and motility, thus preventing proper vessel formation in the tumor. The block of neoangiogenesis depended on cytoplasmic p27Kip1 antimigratory activity and was linked to its ability to bind to and inhibit the microtubule-destabilizing protein stathmin. Our work provides the first evidence that a successful p27Kip1-based gene therapy is linked to tumor microenvironment modification, thus opening new perspectives to the use of gene therapy approaches for the treatment of refractory cancers.


Subject(s)
Brain Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Glioblastoma/pathology , Neovascularization, Pathologic/metabolism , Animals , Cell Movement , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Doxycycline/pharmacology , Endothelial Cells/metabolism , Female , Genetic Vectors/metabolism , Glioblastoma/blood supply , Glioblastoma/drug therapy , Humans , Mice , Mice, Nude , Stathmin/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 24(5): 434-7, 2008 May.
Article in Chinese | MEDLINE | ID: mdl-18466694

ABSTRACT

AIM: To explore CIAPIN1 gene expression in lung carcinoma tissues and its regulatory in the growth of NCI-H446. METHODS: Fifty-one formalin-fixed paraffin-embedded specimens of primary lung cancer and their non-cancerous counterparts were detected by SABC immunohistochemistry method. Adenoviral vector construction was recombined and the gene was transduced into NCI-H446 cells. The expression of CIAPIN1 protein was identified by Western blot. Trypan blue staining was used to count the alive cells and to draw a cellular growth curve. The changes of cell cycle and apoptosis were analyzed by flow cytometry. RESULTS: The positive rate of CIAPIN1 expression in cancer tissues (39.2%)was much lower than that in non-cancerous counterparts (100%, P<0.05). In Ad-CIAPIN1 group, the growth of transfected NCI-H446 cells in vitro was significantly inhibited. In addition the Ad-CIAPIN1-induced cell apoptosis and a predominant arrest in the G1/S phase (P<0.01) were observed. CONCLUSION: The down-regulation of CIAPIN1 expression in tumor is associated with the development of lung carcinoma. Transduction of NCI-H446 CIAPIN1-negative cell, with Ad-CIAPIN1 can inhibit cell growth, suggesting that CIAPIN1 can be a new tumor-related suppressor gene.


Subject(s)
Cell Cycle/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Lung Neoplasms/chemistry , Small Cell Lung Carcinoma/chemistry , Antineoplastic Agents, Phytogenic , Carcinoma, Bronchogenic , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/pharmacology , RNA, Small Interfering/pharmacology , Small Cell Lung Carcinoma/metabolism , Tumor Suppressor Proteins/pharmacology
14.
Virology ; 370(2): 264-72, 2008 Jan 20.
Article in English | MEDLINE | ID: mdl-17931677

ABSTRACT

The phosphatidylinositol-3-kinase (PI3K) and AKT (protein kinase B) signaling pathways play an important role in regulating cell cycle progression and cell survival. In previous studies, we demonstrated that AKT is activated in HTLV-1-transformed cells and that Tax activation of AKT is linked to p53 inhibition and cell survival. In the present study, we extend these observations to identify regulatory pathways affected by AKT in HTLV-1-transformed cells. We demonstrate that inhibition of AKT reduces the level of phosphorylated Bad, an important member of the pro-apoptotic family of proteins. Consistent with the decrease of phosphorylated Bad, cytochrome c is released from the mitochondria and caspase-9 is activated. Pretreatment of the cells with caspase-9 specific inhibitor z-LEHD-FMK or pan caspase inhibitor Ac-DEVD-CHO prevented LY294002-induced apoptosis. Of interest, p53 siRNA prevents LY294002-induced apoptosis in HTLV-1-transformed cells, suggesting that p53 reactivation is linked to apoptosis. In conclusion, the AKT pathway is involved in targeting multiple proteins which regulate caspase- and p53-dependent apoptosis in HTLV-1-transformed cells. Since AKT inhibitors simultaneously inhibit NF-kappaB and activate p53, these drugs should be promising candidates for HTLV-1-associated cancer therapy.


Subject(s)
Apoptosis/physiology , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 1/pathogenicity , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Apoptosis/drug effects , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Transformed , Chromones/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Cytochromes c/metabolism , Human T-lymphotropic virus 1/genetics , Humans , Models, Biological , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Thiophenes/pharmacology , Tumor Suppressor Protein p53/metabolism , bcl-Associated Death Protein/metabolism
15.
Obesity (Silver Spring) ; 14(12): 2136-44, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17189539

ABSTRACT

OBJECTIVE: Subcellular localization has been shown to play an important role in determining activity and accumulation of p27 protein during cell cycle progression. The purpose of this study was to examine p27 localization and ubiquitylation in relation to E3 ligase expression during adipocyte hyperplasia. RESEARCH METHODS AND PROCEDURES: This study used the murine 3T3-L1 preadipocyte model to examine p27 regulation during synchronous cell cycle progression. Cell lysates were isolated over time after hormonal stimulation, fractionated to cytosolic and nuclear compartments, and immunoblotted for relative protein determinations. RESULTS: Data presented in this study show that p27 was present in the cytosol and nucleus in density-arrested preadipocytes and that abundance in both compartments decreased in a phase-specific manner as preadipocytes synchronously re-entered the cell cycle during early phases of adipocyte differentiation. Blocking CRM1-mediated nuclear export did not prevent degradation, nor did it cause nuclear accumulation of p27, suggesting that distinct mechanisms mediating cytosolic and nuclear p27 degradation were involved. Treating preadipocytes with a potent and specific proteasome inhibitor during hormonal stimulation prevented Skp2 accumulation and p27(187) phosphorylation, which are essential events for SCF(Skp2) E3 ligase activity and nuclear p27 ubiquitylation during S/G(2) phase progression. Proteasome blockade also resulted in the first evidence of cytosolic p27 ubiquitylation during late G(1) phase as preadipocytes undergo the transition from quiescence to proliferation. DISCUSSION: These data are consistent with the postulate that p27 is ubiquitylated and targeted for degradation by the 26S proteasome in a phase-specific manner by distinct ubiquitin E3 ligases localized to the cytosol and nucleus during adipocyte hyperplasia.


Subject(s)
Adipose Tissue/metabolism , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Cell Differentiation , Cell Fractionation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , G1 Phase , G2 Phase , Mice , Protein Kinase Inhibitors/pharmacology , S Phase , Ubiquitin/metabolism
16.
Dig Dis Sci ; 51(3): 580-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16614970

ABSTRACT

The purpose of this study was to establish if estrogen-induced hepatocyte proliferation in vitro involves the cell cycle regulators cyclin D1, p21(Cip1), and p27(Kip1). Male rat hepatocytes were cultured in presence of 17-beta-estradiol (E2) +/- ICI-182780, a pure estrogen antagonist, and [3H]-thymidine, as required. DNA synthesis as well as p21(Cip1), p27(Kip1), and cyclin D1mRNA and protein levels were evaluated at different times (12, 24, 36, and 48 hours) of incubation. E2-increased DNA synthesis was correlated with cyclin D1 and p21(Cip1) (mRNA and protein) variations that were reversed by the addition of ICI-182780. p27(Kip1) protein levels progressively increased regardless of the presence of E2 or ICI-182780. Our data confirm that estrogens' stimulatory effect is related to their ability to increase cyclin D1 levels. The increase of p21(Cip1) is probably related to the reentry of hepatocytes in the quiescent state. p27(Kip1) protein is not able to arrest hepatocyte proliferation.


Subject(s)
Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Estrogens/pharmacology , Hepatocytes/drug effects , Animals , Cells, Cultured , Cyclin D1/drug effects , Cyclin-Dependent Kinase Inhibitor p21/analysis , Hepatocytes/metabolism , Immunoblotting , Male , Models, Animal , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Sensitivity and Specificity
17.
Zhonghua Wai Ke Za Zhi ; 44(19): 1349-52, 2006 Oct 01.
Article in Chinese | MEDLINE | ID: mdl-17217824

ABSTRACT

OBJECTIVE: To investigate the effects of mutant exogenous P27(kip1) gene on chemosensitivity of human cholangiocarcinoma cell line. METHODS: The recombinant vector was constructed and the mutant P27(kip1) gene was transfected into human cholangiocarcinoma cell line (QBC(939)). RT-PCR and Western blot were used to determine the expression of target genes. The effects of 5-fluorouracil (5-FU), mitomycin C (MMC) and cyclophosphamide (CTX) on the transfected cells were detected by assaying the apoptotic rate and growth inhibition by methabenzthiazuron (MTT) assay and flow cytometry (FCM). RESULTS: The mutant exogenous P27(kip1) gene was expressed effectively in the cells, and the expression enhanced the apoptosis and growth inhibition of QBC(939) inducted by 5-FU, MMC and CTX. The ratio of growth inhibiting increased significantly from 41.89% (5-FU), 45.59% (MMC), 38.91% (CTX) to 56.15% (5-FU), 55.65% (MMC), 51.69% (CTX), and apoptosis index from 13.76% +/- 3.03% (5-FU), 11.76% +/- 3.99% (MMC), 10.46% +/- 2.10% (CTX) to 41.39% +/- 4.32% (5-FU), 35.94% +/- 2.71% (MMC), 34.46% +/- 2.32% (CTX) (P < 0.05). CONCLUSIONS: The exogenous P27(kip1) gene transfer can remarkably increase the drug sensibility of the cholangiocarcinoma cells. The strategy targeted to control the cell cycle may be more effective in cancer treatment by combination of P27(kip1) gene therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic , Cholangiocarcinoma/pathology , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Adenoviridae/genetics , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Drug Synergism , Genetic Vectors , Humans , Transfection
18.
Cancer Cell ; 8(6): 455-66, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16338659

ABSTRACT

Primary mouse embryonic fibroblasts lacking expression of all three retinoblastoma protein family members (TKO MEFs) have lost the G1 restriction point. However, in the absence of mitogens these cells become highly sensitive to apoptosis. Here, we show that TKO MEFs that survive serum depletion pass G1 but completely arrest in G2. p21CIP1 and p27KIP1 inhibit Cyclin A-Cdk2 activity and sequester Cyclin B1-Cdk1 in inactive complexes in the nucleus. This response is alleviated by mitogen restimulation or inactivation of p53. Thus, our results disclose a cell cycle arrest mechanism in G2 that restricts the proliferative capacity of mitogen-deprived cells that have lost the G1 restriction point. The involvement of p53 provides a rationale for the synergism between loss of Rb and p53 in tumorigenesis.


Subject(s)
Cell Cycle/drug effects , Cell Cycle/physiology , Mitogens/physiology , Retinoblastoma Protein/physiology , Retinoblastoma-Like Protein p107/physiology , Retinoblastoma-Like Protein p130/physiology , Animals , Apoptosis/physiology , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/drug effects , CDC2 Protein Kinase/metabolism , Cell Cycle/genetics , Cyclin A/antagonists & inhibitors , Cyclin A/metabolism , Cyclin B/drug effects , Cyclin B/metabolism , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , DNA Damage , Fibroblasts/cytology , Fibroblasts/metabolism , G1 Phase/drug effects , G1 Phase/physiology , G2 Phase/drug effects , G2 Phase/physiology , Mice , Mice, Knockout , Mitogens/pharmacology , Neurons/drug effects , Neurons/physiology , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p130/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...