Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 243
Filter
1.
Endocr J ; 67(2): 177-184, 2020 Feb 28.
Article in English | MEDLINE | ID: mdl-31666445

ABSTRACT

Cushing's disease is primarily caused by autonomic hypersecretion of adrenocorticotropic hormone (ACTH) from a pituitary adenoma. In Cushing's disease, mutations in the ubiquitin-specific protease 8 (USP8) have been detected. These mutations are associated with hyperactivation of USP8 that prevent epidermal growth factor receptor (EGFR) degradation. This leads to increased EGFR stability and results in the maintenance of EGFR signaling in Cushing's disease. USP8 inhibitors can suppress the growth of various tumors. In this study, the effects of a potent USP8 inhibitor, DUBs-IN-2, on ACTH production and cell proliferation were examined in mouse corticotroph tumor (AtT-20) cells. Proopiomelanocortin (Pomc) mRNA levels and ACTH levels were decreased in AtT-20 cells by DUBs-IN-2. Further, cell proliferation was inhibited, and apoptosis was induced by DUBs-IN-2. Transcript levels of pituitary tumor-transforming gene 1 (Pttg1), a pituitary tumor growth marker, were increased; and transcript levels of stress response growth arrest and DNA damage-inducible 45 (Gadd45ß) and Cdk5 and ABL enzyme substrate 1 (Cables1) mRNA levels were increased in response to the drug. Gadd45ß or Cables1 knockdown partially inhibited the DUBs-IN-2-induced decrease in cell proliferation, but not Pomc mRNA levels. Both GADD45ß and CABLES1 may be responsible, at least in part, for the USP8-induced suppression of corticotroph tumor cell proliferation. USP-8 may be a new treatment target in Cushing's disease.


Subject(s)
ACTH-Secreting Pituitary Adenoma/metabolism , Adenoma/metabolism , Adrenocorticotropic Hormone/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Indenes/pharmacology , Pyrazines/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Adrenocorticotropic Hormone/metabolism , Animals , Antigens, Differentiation/drug effects , Antigens, Differentiation/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 5/drug effects , Cyclin-Dependent Kinase 5/genetics , Cyclins/drug effects , Cyclins/genetics , Endopeptidases , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Gene Knockdown Techniques , Mice , Pituitary ACTH Hypersecretion/metabolism , Pro-Opiomelanocortin/drug effects , Pro-Opiomelanocortin/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Securin/drug effects , Securin/genetics
2.
Biochem Biophys Res Commun ; 513(4): 1041-1047, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31010673

ABSTRACT

Cardiovascular disease is a leading cause of death worldwide. Mammalian cardiomyocytes (CMs) proliferate during embryonic development, whereas they largely lose their regenerative capacity after birth. Defined factors expressed in cardiac progenitors or embryonic CMs may activate the cell cycle and induce CM proliferation in postnatal and adult hearts. Here, we report that the overexpression of Tbx6, enriched in the cardiac mesoderm (progenitor cells), induces CM proliferation in postnatal and adult mouse hearts. By screening 24 factors enriched in cardiac progenitors or embryonic CMs, we found that only Tbx6 could induce CM proliferation in primary cultured postnatal rat CMs. Intriguingly, it did not induce the proliferation of cardiac fibroblasts. We next generated a recombinant adeno-associated virus serotype 9 vector encoding Tbx6 (AAV9-Tbx6) for transduction into mouse CMs in vivo. The subcutaneous injection of AAV9-Tbx6 into neonatal mice induced CM proliferation in postnatal and adult mouse hearts. Mechanistically, Tbx6 overexpression upregulated multiple cell cycle activators including Aurkb, Mki67, Ccna1, and Ccnb2 and suppressed the tumor suppressor Rb1. Thus, Tbx6 promotes CM proliferation in postnatal and adult mouse hearts by modifying the expression of cell cycle regulators.


Subject(s)
Cell Proliferation/drug effects , Myocardium/cytology , Myocytes, Cardiac/cytology , T-Box Domain Proteins/physiology , Adenoviridae/genetics , Animals , Animals, Newborn , Cell Cycle Proteins/drug effects , Cells, Cultured , Cyclins/drug effects , Genetic Vectors/administration & dosage , Heart , Mice , Rats , Regeneration , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/pharmacology
3.
Plant Physiol Biochem ; 113: 20-31, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28157579

ABSTRACT

Glucose and sucrose play a dual role: as carbon and energy sources and as signaling molecules. In order to address the impact that sugars may have on maize seeds during germination, embryo axes were incubated with or without either of the two sugars. Expression of key cell cycle markers and protein abundance, cell patterning and de novo DNA synthesis in root meristem zones were analyzed. Embryo axes without added sugars in imbibition medium were unable to grow after 7 days; in sucrose, embryo axes developed seminal and primary roots with numerous root hairs, whereas in glucose axes showed a twisted morphology, no root hair formation but callus-like structures on adventitious and primary seminal roots. More and smaller cells were observed with glucose treatment in root apical meristems. de novo DNA synthesis was stimulated more by glucose than by sucrose. At 24 h of imbibition, expression of ZmCycD2;2a and ZmCycD4;2 was increased by sucrose and reduced by glucose. CDKA1;1 and CDKA2;1 expression was stimulated equally by both sugars. Protein abundance patterns were modified by sugars: ZmCycD2 showed peaks on glucose at 12 and 36 h of imbibition whereas sucrose promoted ZmCycD3 protein accumulation. In presence of glucose ZmCycD3, ZmCycD4 and ZmCycD6 protein abundance was reduced after 24 h. Finally, both sugars stimulated ZmCDKA protein accumulation but at different times. Overall, even though glucose appears to act as a stronger mitogen stimulator, sucrose stimulated the expression of more cell cycle markers during germination. This work provides evidence of a differential response of cell cycle markers to sucrose and glucose during maize germination that may affect the developmental program during plantlet establishment.


Subject(s)
Germination/drug effects , Glucose/pharmacology , Sucrose/pharmacology , Zea mays/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/biosynthesis , Cyclins/drug effects , DNA, Plant/biosynthesis , Glucose/metabolism , Glucose/physiology , Plant Development/drug effects , Plant Proteins/biosynthesis , Plant Roots/cytology , Plant Roots/drug effects , Seeds/cytology , Seeds/drug effects , Sucrose/metabolism , Zea mays/cytology , Zea mays/embryology
4.
J Appl Toxicol ; 35(8): 952-69, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25534134

ABSTRACT

The effects of thallium [Tl(I) and Tl(III)] on the PC12 cell cycle were evaluated without (EGF(-)) or with (EGF(+)) media supplementation with epidermal growth factor (EGF). The following markers of cell-cycle phases were analyzed: cyclin D1 (G1 ); E2F-1, cyclin E and cytosolic p21 (G1 →S transition); nuclear PCNA and cyclin A (S); and cyclin B1 (G2). The amount of cells in each phase and the activation of the signaling cascade triggered by EGF were also analyzed. Tl(I) and Tl(III) (5-100 µM) caused dissimilar effects on PC12 cell proliferation. In EGF(-) cells, Tl(I) increased the expression of G1 →S transition markers and nuclear PCNA, without affecting cyclin A or cyclin B1. In addition to those, cyclin B1 was also increased in EGF(+) cells. In EGF(-) cells, Tl(III) increased the expression of cyclin D1, all the G1→S and S phase markers and cyclin B1. In EGF(+) cells, Tl(III) increased cyclin D1 expression and decreased all the markers of G1 →S transition and the S phase. Even when these cations did not induce the activation of EGF receptor (EGFR) in EGF(-) cells, they promoted the phosphorylation of ERK1/2 and Akt. In the presence of EGF, the cations anticipated EGFR phosphorylation without affecting the kinetics of EGF-dependent ERK1/2 and Akt phosphorylation. Altogether, results indicate that Tl(I) promoted cell proliferation in both EGF(-) and EGF(+) cells. In contrast, Tl(III) promoted the proliferation of EGF(-) cells but delayed it in EGF(+) cells, which may be related to the toxic effects of this cation in PC12 cells.


Subject(s)
Cell Cycle/drug effects , Cyclins/drug effects , Epidermal Growth Factor/drug effects , Titanium/toxicity , Animals , Cations , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , ErbB Receptors/drug effects , ErbB Receptors/genetics , Humans , MAP Kinase Signaling System/drug effects , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/genetics , Oxidation-Reduction , PC12 Cells , Phosphorylation , Rats , Signal Transduction/drug effects
5.
Mol Nutr Food Res ; 55(7): 989-98, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21618425

ABSTRACT

SCOPE: Dysfunction of histone acetyltransferases (HATs) or histone deacetylases (HDACs) involved in histones acetylation has been associated with cancer. Inhibitors of these enzymes are becoming potential targets for new therapies. METHODS AND RESULTS: This study reports by Western-Blot analysis, that peptide lunasin is mainly an in vitro inhibitor of histone H4 acetylation by P300/cAMP-response element-binding protein (CBP)-associated factor (PCAF), with IC50 values dependent on the lysine position sensitive to be acetylated (0.83 µM (H4-Lys 8), 1.27 µM (H4-Lys 12) and 0.40 µM (H4-Lys 5, 8, 12, 16)). Lunasin is also capable of inhibiting H3 acetylation (IC50 of 5.91 µM (H3-Lys 9) and 7.81 µM (H3-Lys 9, 14)). Studies on structure-activity relationship establish that lunasin's sequence are essential for inhibiting H4 acetylation whereas poly-D sequence is the main active sequence responsible for H3 acetylation inhibition. Lunasin also inhibits H3 and H4 acetylation and cell proliferation (IC50 of 181 µM) in breast cancer MDA-MB-231 cells. Moreover, this peptide decreases expression of cyclins and cyclin dependent kinases-4 and -6, implicated in cell cycle pathways. CONCLUSION: Results from this study demonstrates lunasin's role as modulator of histone acetylation and protein expression that might contribute on its chemopreventive properties against breast cancer.


Subject(s)
Histone Acetyltransferases/antagonists & inhibitors , Histones/metabolism , Soybean Proteins/chemistry , Soybean Proteins/pharmacology , Acetylation/drug effects , Amino Acid Sequence , Biomarkers/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase 4/drug effects , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/drug effects , Cyclin-Dependent Kinase 6/metabolism , Cyclins/drug effects , Cyclins/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histones/drug effects , Humans , Inhibitory Concentration 50 , Lysine/metabolism , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Structure-Activity Relationship , p300-CBP Transcription Factors/antagonists & inhibitors
6.
Arch Toxicol ; 85(12): 1541-50, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21512803

ABSTRACT

Okadaic acid is one of the toxins responsible for the human intoxication known as diarrhetic shellfish poisoning, which appears after the consumption of contaminated shellfish. The main diarrhetic shellfish poisoning toxins are okadaic acid, dinophysistoxin-1, -2, and -3. In vivo, after intraperitoneal injection, dinophysistoxin-2 is approximately 40% less toxic than okadaic acid in mice. The cytotoxic and genotoxic effect of okadaic acid varies very significantly in different cell lines, so similar responses could be expected for dinophysistoxin-2. In order to determine whether this was the case, we studied the effect of okadaic acid and dinophysistoxin-2 in two hepatic cell lines (HepG2 and Clone 9). The cytotoxicity of these toxins, as well as their effects on the cell cycle and its regulation on both cell lines, were determined. Okadaic acid and dinophysistoxin-2 resulted to be equipotent in clone 9 cultures, while okadaic acid was more potent than dinophysistoxin-2 in HepG2 cell cultures. Both toxins had opposite effects on the cell cycle; they arrested the cell cycle of clone 9 cells in G2/M inducing aberrant mitosis while arresting the cell cycle of HepG2 in G0/G1. When the effect of the toxins on p53 subcellular distribution was studied, p53 was detected in the nuclei of both cell types. The effect of the toxins on the gene expression of cyclins and cyclin-dependent kinases was different for both cell lines. The toxins induced an increase in gene expression of cyclins A, B, and D in clone 9 cells while they induced a decrease in cyclins A and B in HepG2 cells. They also induced a decrease in cyclin-dependent kinase 1 in HepG2 cells.


Subject(s)
Cell Cycle/drug effects , Liver/drug effects , Okadaic Acid/toxicity , Pyrans/toxicity , Animals , CDC2 Protein Kinase/drug effects , CDC2 Protein Kinase/metabolism , Cell Cycle Checkpoints/drug effects , Cyclins/drug effects , Cyclins/genetics , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Injections, Intraperitoneal , Liver/cytology , Liver/pathology , Mitosis/drug effects , Rats , Tumor Suppressor Protein p53/metabolism
7.
Am J Pathol ; 178(5): 2109-20, 2011 May.
Article in English | MEDLINE | ID: mdl-21514426

ABSTRACT

Substantial evidence implicates the ubiquitin-conjugating enzyme E2C (UBE2C) gene, in several human cancers, including colorectal carcinoma (CRC). We therefore investigated the prognostic value of UBE2C alterations in CRC and UBE2C signaling in CRC cell lines. UBE2C protein expression and UBE2C gene copy number were evaluated on clinical samples by immunohistochemistry and fluorescence in situ hybridization in a TMA format. The effect of the proteasome inhibitor bortezomib and small-interfering RNA knockdown was assessed by apoptotic assays and immunoblotting. UBE2C dysregulation was associated with proliferative marker Ki-67, accumulation of cyclin A and B1, and a poor overall survival. UBE2C expression was an independent prognostic marker in early-stage (I and II) CRC. UBE2C depletion resulted in suppression of cellular growth and accumulation of cyclin A and B1. In vitro, bortezomib treatment of CRC cells caused inhibition of cell viability via down-regulation of UBE2C. UBE2C knockdown by bortezomib or transfection with specific small-interfering RNA against UBE2C also caused cells to be arrested at the G2/M level, leading to accumulation of cyclin A and cyclin B1. In vivo, a significant reduction in tumor volume and weight was noted in mice treated with a combination of subtoxic doses of oxaliplatin and bortezomib compared with treatment with oxaliplatin or bortezomib alone. Altogether, our results suggest that UBE2C and the ubiquitin-proteasome pathway may be potential targets for therapeutic intervention in CRC.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Cell Cycle/drug effects , Colorectal Neoplasms/metabolism , Pyrazines/pharmacology , Ubiquitin-Conjugating Enzymes/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Biomarkers, Tumor/analysis , Bortezomib , Cell Cycle/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclins/drug effects , Cyclins/metabolism , Gene Dosage , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mice , Mice, Nude , Prognosis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Ubiquitin-Conjugating Enzymes/drug effects , Ubiquitin-Conjugating Enzymes/genetics , Xenograft Model Antitumor Assays
8.
Toxicol Appl Pharmacol ; 250(2): 213-20, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21070798

ABSTRACT

Aniline exposure is associated with toxicity to the spleen leading to splenomegaly, hyperplasia, fibrosis and a variety of sarcomas of the spleen on chronic exposure. In earlier studies, we have shown that aniline exposure leads to iron overload, oxidative stress and activation of redox-sensitive transcription factors, which could regulate various genes leading to a tumorigenic response in the spleen. However, molecular mechanisms leading to aniline-induced cellular proliferation in the spleen remain largely unknown. This study was, therefore, undertaken on the regulation of G1 phase cell cycle proteins (cyclins), expression of cyclin-dependent kinases (CDKs), phosphorylation of retinoblastoma protein (pRB) and cell proliferation in the spleen, in an experimental condition preceding a tumorigenic response. Male SD rats were treated with aniline (0.5 mmol/kg/day via drinking water) for 30 days (controls received drinking water only), and splenocyte proliferation, protein expression of G1 phase cyclins, CDKs and pRB were measured. Aniline treatment resulted in significant increases in splenocyte proliferation, based on cell counts, cell proliferation markers including proliferating cell nuclear antigen (PCNA), nuclear Ki67 protein (Ki67) and minichromosome maintenance (MCM), MTT assay and flow cytometric analysis. Western blot analysis of splenocyte proteins from aniline-treated rats showed significantly increased expression of cyclins D1, D2, D3 and E, as compared to the controls. Similarly, real-time PCR analysis showed significantly increased mRNA expression for cyclins D1, D2, D3 and E in the spleens of aniline-treated rats. The overexpression of these cyclins was associated with increases in the expression of CDK4, CDK6, CDK2 as well as phosphorylation of pRB protein. Our data suggest that increased expression of cyclins, CDKs and phosphorylation of pRB protein could be critical in cell proliferation, and may contribute to aniline-induced tumorigenic response in the spleen.


Subject(s)
Aniline Compounds/toxicity , Carcinogens/toxicity , Cyclin-Dependent Kinases/drug effects , Cyclins/drug effects , Gene Expression Regulation/drug effects , Animals , Blotting, Western , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Disease Models, Animal , Flow Cytometry , Male , Phosphorylation/drug effects , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Spleen/cytology , Spleen/drug effects , Spleen/metabolism
9.
Cancer Epidemiol Biomarkers Prev ; 18(6): 1720-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19505905

ABSTRACT

Frequent exposure to nickel compounds has been considered as one of the potential causes of human lung cancer. However, the molecular mechanism of nickel-induced lung carcinogenesis remains obscure. In the current study, slight S-phase increase, significant G(2)/M cell cycle arrest, and proliferation blockage were observed in human bronchial epithelial cells (Beas-2B) upon nickel exposure. Moreover, the induction of cyclin D1 and cyclin E by nickel was shown for the first time in human pulmonary cells, which may be involved in nickel-triggered G(1)/S transition and cell transformation. In addition, we verified that hypoxia-inducible factor-1alpha, an important transcription factor of nickel response, was not required for the cyclin D1 or cyclin E induction. The role of p53 in nickel-induced G(2)/M arrest was excluded, respecting that its protein level, ser(15) phosphorylation, and transcriptional activity were not changed in nickel response. Further study revealed that cyclin A was not activated in nickel response, and cyclin B1, which not only promotes G(2)/M transition but also prevents M-phase exit of cells if not degraded in time, was up-regulated by nickel through a manner independent of hypoxia-inducible factor. More importantly, our results verified that overexpressed cyclin B1, veiling the effect of cyclin D1 or cyclin E, mediated nickel-caused M-phase blockage and cell growth inhibition, which may render pulmonary cells more sensitive to DNA damage and facilitates cancer initiation. These results will not only deepen our understanding of the molecular mechanism involved in nickel carcinogenecity, but also lead to the further study on chemoprevention of nickel-associated human cancer.


Subject(s)
Carcinogens/toxicity , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclins/drug effects , Nickel/toxicity , Respiratory Mucosa/drug effects , Blotting, Western , Cell Line , Cyclins/biosynthesis , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Suppressor Protein p53/metabolism
10.
Cancer Chemother Pharmacol ; 64(3): 529-38, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19184021

ABSTRACT

AIMS: We evaluated both in vitro and in vivo antitumoral properties of an isolated compound from Wilbrandia ebracteata, dihydrocucurbitacin-B (DHCB), using B16F10 cells (murine melanoma). MATERIALS AND METHODS: We made use of MTT and (3)H-Thymidine assays to investigate the cell viability and cell proliferation, flow cytometry analysis to monitor cell cycle and apoptosis, western blot analysis to evaluate the expression of cell cycle proteins, imunofluorescence analysis and in vivo tumor growth and metastasis. RESULTS: Dihydrocucurbitacin-B significantly reduced cell proliferation without important effects on cells viability. DHCB lead cells to accumulate in G2/M phases accompanied by the appearance of polyploid cells, confirmed by fluorescence assays that demonstrated a remarkable alteration in the cell cytoskeleton and formation of binuclear cells. Annexin-V-FITC incorporation demonstrated that DHCB did not induce apoptosis. About 10 microg/mL DHCB was found to decrease cyclin-A, and especially in cyclin-B1. The in vivo experiments showed that DHCB treatment (once a day up to 12 days; p.o.) was able to reduce the tumor growth and lung metastasis up to 83.5 and 50.3%, respectively. CONCLUSIONS: Dihydrocucurbitacin-B reduces cell proliferation due to a decrease in the expression of cyclins, mainly cyclin-B1 and disruption of the actin cytoskeleton, arresting B16F10 cells in G2/M phase. Taken together, the in vitro and in vivo experiments suggest that DHCB was effective against cancer, however, it remains to be proved if DHCB will be a good candidate for drug development.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cucurbitaceae/chemistry , Melanoma, Experimental/drug therapy , Triterpenes/pharmacology , Actins/drug effects , Actins/metabolism , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclins/drug effects , Cyclins/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Triterpenes/isolation & purification
11.
PLoS Pathog ; 5(1): e1000263, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19148268

ABSTRACT

Cyclin T1 is a regulatory subunit of a general RNA polymerase II elongation factor known as P-TEFb. Cyclin T1 is also required for Tat transactivation of HIV-1 LTR-directed gene expression. Translation of Cyclin T1 mRNA has been shown to be repressed in human monocytes, and this repression is relieved when cells differentiate to macrophages. We identified miR-198 as a microRNA (miRNA) that is strongly down-regulated when monocytes are induced to differentiate. Ectopic expression of miR-198 in tissue culture cells reduced Cyclin T1 protein expression, and plasmid reporter assays verified miR-198 target sequences in the 3' untranslated region (3'UTR) of Cyclin T1 mRNA. Cyclin T1 protein levels increased when an inhibitor of miR-198 was transfected into primary monocytes, and overexpression of miR-198 in primary monocytes repressed the normal up-regulation of Cyclin T1 during differentiation. Expression of an HIV-1 proviral plasmid and HIV-1 replication were repressed in a monocytic cell line upon overexpression of miR-198. Our data indicate that miR-198 functions to restrict HIV-1 replication in monocytes, and its mechanism of action appears to involve repression of Cyclin T1 expression.


Subject(s)
Cyclins/drug effects , HIV-1/genetics , MicroRNAs/physiology , Monocytes/virology , 3' Untranslated Regions/drug effects , Cell Differentiation , Cell Line , Cyclin T , Down-Regulation , HIV Infections/genetics , HIV Long Terminal Repeat , Humans , Macrophages/virology , Monocytes/drug effects , Monocytes/metabolism , RNA, Messenger/metabolism , Virus Replication/genetics
12.
Neurosci Lett ; 450(3): 347-50, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19103257

ABSTRACT

The extent to which neurons proceed into the cell cycle and the mechanisms whereby cell cycle re-entry leads to apoptosis vary in response to agonists. We previously showed upregulation of early G1 regulators in thrombin-treated neurons yet neurons did not proceed to S phase but to apoptosis. The objective of this study is to explore mechanisms which might prevent S phase entry and promote apoptosis in thrombin-treated neurons. Cultured rat brain neurons are exposed to thrombin (200nM) for 30min to 4.5h and the expression of cyclin C, cyclin dependent kinases (cdk1, cdk2, cdk3, cdk8) and the cell cycle inhibitor p27 assessed. Our data show a simultaneous decrease of both cyclin C and cdk3 proteins soon after thrombin treatment. The decrease in cyclin C also correlates with decreases in cdk1 and cdk2, at both mRNA and protein levels. There is no change in expression of cdk8 or the cell cycle inhibitor p27 in response to thrombin treatment. These results suggest that decreases in G1-S regulators cyclin C and cdks 3, cdk2 and cdk1 in response to thrombin could make conditions unfavorable for S phase entry and favor neuronal apoptosis.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Neurons/enzymology , Thrombin/pharmacology , Animals , Apoptosis/drug effects , CDC2 Protein Kinase/drug effects , CDC2 Protein Kinase/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/drug effects , Cells, Cultured , Cyclin C , Cyclin-Dependent Kinase 2/drug effects , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 3 , Cyclin-Dependent Kinase 8 , Cyclin-Dependent Kinase Inhibitor p27/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinases/drug effects , Cyclin-Dependent Kinases/metabolism , Cyclins/drug effects , Cyclins/metabolism , G1 Phase/drug effects , G1 Phase/physiology , Neurons/drug effects , Rats , S Phase/drug effects , S Phase/physiology , Thrombin/metabolism
13.
Toxicol In Vitro ; 22(6): 1605-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18652887

ABSTRACT

Pyrogallol (PG) is a polyphenol compound and has been known to be an O(2)(.-) generator. We evaluated the effects of PG on the growth of human pulmonary adenocarcinoma Calu-6 cells in relation to the cell cycle. DNA flow cytometric analysis indicated that PG induced a G2 phase arrest of the cell cycle in Calu-6 cells at 72 h. PG down-regulated the expression of CDKI (p27), CDK2, CDK4 and CDK6 as well as cyclin D1, and increased cyclin A and cyclin B1 proteins. In addition, O(2)(.-) levels were significantly increased in PG-treated cells. Treatment with catalase rescued Calu-6 cells from PG-induced apoptosis, and also prevented the growth inhibition as well as a G2 phase arrest by PG, which were accompanied with the down-regulation of O(2)(.-) levels. In conclusion, PG inhibited the growth of Calu-6 cells by inducing the cell cycle arrest, accompanied with an increase in O(2)(.-) levels.


Subject(s)
Antioxidants/pharmacology , Cell Cycle/drug effects , Lung Neoplasms/metabolism , Pyrogallol/pharmacology , Adenocarcinoma/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases/drug effects , Cyclin-Dependent Kinases/metabolism , Cyclins/drug effects , Cyclins/metabolism , Down-Regulation/drug effects , Flow Cytometry , G2 Phase/drug effects , Humans , Superoxides/metabolism
14.
Arthritis Rheum ; 58(6): 1657-63, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18512786

ABSTRACT

OBJECTIVE: To investigate the effect of ciprofloxacin on the proliferation and cell cycle progression of tendon cells, and to explore the potential molecular mechanism of ciprofloxacin-associated tendinopathy by analyzing the expression of cell cycle-related cyclin and cyclin-dependent kinase (CDK). METHODS: Rat Achilles tendon cells were treated with ciprofloxacin and then assessed by MTT assay, flow cytometric analysis, and fluorescence confocal microscopy. Levels of messenger RNA (mRNA) for CDK-1 and cyclin B were determined by reverse transcriptase-polymerase chain reaction. Protein expression of CDK-1, cyclin B, checkpoint kinase 1 (CHK-1), and polo-like kinase 1 (PLK-1) was determined by Western blot analysis. RESULTS: Ciprofloxacin inhibited tendon cell proliferation and caused cell cycle arrest at the G2/M phase. Confocal microscopy revealed that chromosomes in ciprofloxacin-treated cells neither properly aligned along the equatorial planes nor segregated successfully during metaphase. Mitotic arrest, misaligned chromosomes, and poor bipolar spindle formation were observed in ciprofloxacin-treated cells. CDK-1 and cyclin B protein and mRNA were both down-regulated. CHK-1 protein expression was also suppressed, but PLK-1 protein expression was up-regulated by ciprofloxacin. CONCLUSION: Our findings suggest a possible mechanism of ciprofloxacin-associated tendinopathy. Down-regulation of CHK-1 and up-regulation of PLK-1 may account for mitotic arrest observed in ciprofloxacin-treated cells.


Subject(s)
Achilles Tendon/cytology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Ciprofloxacin/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Achilles Tendon/growth & development , Animals , Cells, Cultured , Cyclin-Dependent Kinases/drug effects , Cyclins/drug effects , Rats , Rats, Sprague-Dawley , Tendinopathy/chemically induced , Tendinopathy/pathology
15.
Int J Cancer ; 123(1): 41-50, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18435416

ABSTRACT

Prostate cancer (PCa) is the leading cause of cancer-related deaths in men; urgent measures are warranted to lower this deadly malignancy. Silymarin is a known cancer chemopreventive agent, but the relative anticancer efficacy of its constituents is still unknown. Here, we compared the efficacy of 7 pure flavonolignan compounds isolated from silymarin, namely silybin A, silybin B, isosilybin A, isosilybin B, silydianin, isosilydianin, silychristin and isosilychristin, in advanced human PCa PC3 cells. Silybin A, silybin B, isosilybin A, isosilybin B, silibinin and silymarin strongly inhibited the colony formation by PC3 cells (p < 0.001), while silydianin, silychristin and isosilychristin had marginal effect (p < 0.05). Using cell growth and death assays, we identified isosilybin B as the most effective isomer. FACS analysis for cell cycle also showed that silybin A, silybin B, isosilybin A, isosilybin B, silibinin and silymarin treatment resulted in strong cell cycle arrest in PC3 cells after 72 hr of treatment, while the effect of silydianin, silychristin and isosilychristin was marginal (if any). Western blot analysis also showed the differential effect of these compounds on the levels of cell cycle regulators-cyclins (D, E, A and B), CDKs (Cdk2, 4 and Cdc2), CDKIs (p21 and p27) and other cell cycle regulators (Skp2, Cdc25A, B, C and Chk2). This study provided further evidence for differential anticancer potential among each silymarin constituent, which would have potential implications in devising better formulations of silymarin against prostate and other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/prevention & control , Protective Agents/pharmacology , Silymarin/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor Proteins/drug effects , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cyclin-Dependent Kinases/drug effects , Cyclin-Dependent Kinases/metabolism , Cyclins/drug effects , Cyclins/metabolism , Drug Screening Assays, Antitumor/methods , Flavonolignans/pharmacology , Flow Cytometry , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isomerism , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Silybin , Silymarin/analogs & derivatives , Silymarin/chemistry , Time Factors , cdc25 Phosphatases/drug effects , cdc25 Phosphatases/metabolism
16.
Toxicol In Vitro ; 22(1): 87-95, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17920232

ABSTRACT

Sulfur is commonly used in Asia as an herbal medicine to treat inflammation and cancer, and potent chemopreventive effects have been demonstrated in various in vivo and in vitro models for sulfur-containing compounds found in naturally occurring products. Here, we report the growth inhibitory and apoptosis-related effects of a newly developed highly purified sulfur (HPS) on immortalized human oral keratinocytes (IHOKs) and on oral cancer cells representing two stages of oral cancer (HN4, HN12) based on a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, Western blotting, cell cycle analysis, and nuclear staining. The purity of the sulfur preparation was verified by high-performance liquid chromatography. HPS inhibited the proliferation of immortalized and malignant oral keratinocytes in a dose- and time-dependent manner. FITC-annexin V staining, DNA fragmentation testing, and Hoechst 33258 staining revealed that HPS inhibited cell growth via apoptosis. HPS increased the sub-G1 cell cycle fraction, with decreased expression of cyclins D1, D2, and E and their activating partners cdk2, cdk4, and cdk6, and a concomitant induction of p53 and p21/WAF1. Furthermore, HPS treatment increased the cytosolic level of cytochrome c and resulted in caspase-3 activation; this effect was correlated with Bax up-regulation and Bcl-2 down-regulation. Thus, these data suggest that HPS is a potential candidate for anti-cancer therapy in oral cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Mouth Neoplasms/drug therapy , Sulfur Compounds/pharmacology , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclins/drug effects , Cyclins/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Neoplasm Staging , Sulfur Compounds/administration & dosage , Time Factors
17.
Environ Health Perspect ; 115(10): 1460-6, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17938736

ABSTRACT

BACKGROUND: Formaldehyde (FA) is classified as a human carcinogen and has been linked to increased leukemia rates in some epidemiologic studies. Inhalation of FA induces sensory irritation at relatively low concentrations. However, little is known concerning the cellular alterations observed after FA exposure in humans. OBJECTIVES: Our aim was to profile global gene expression in Hs 680.Tr human tracheal fibroblasts exposed to FA and to develop biomarkers for the evaluation of FA exposure in humans. METHODS AND RESULTS: We used gene expression analysis, and identified 54 genes designated as FA responsive. On the basis of these data, we conducted an exploratory analysis of the expression of these genes in human subjects exposed to high or low levels of FA. We monitored FA exposure by measuring the urinary concentration of thiazolidine-4-carboxylate (TZCA), a stable and quantitative cysteinyl adduct of FA. Nine genes were selected for real-time PCR analysis; of these, BHLHB2, CCNL1, SE20-4, C8FW, PLK2, and SGK showed elevated expression in subjects with high concentrations of TZCA. CONCLUSION: The identification of gene marker candidates in vitro using microarray analysis and their validation using human samples obtained from exposed subjects is a good tool for discovering genes of potential mechanistic interest and biomarkers of exposure. Thus, these genes are differentially expressed in response to FA and are potential effect biomarkers of FA exposure.


Subject(s)
Fibroblasts/drug effects , Formaldehyde/adverse effects , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers , Cell Line , Complement C8/drug effects , Complement C8/genetics , Cyclins/drug effects , Cyclins/genetics , DNA-Binding Proteins , Environmental Exposure , Formaldehyde/metabolism , Homeodomain Proteins/drug effects , Homeodomain Proteins/genetics , Humans , Immediate-Early Proteins/drug effects , Immediate-Early Proteins/genetics , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Thiazolidines/urine
18.
J Pharm Pharmacol ; 59(8): 1087-93, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17725850

ABSTRACT

Oleanolic acid (3beta-hydroxy-olean-12-en-28-oic acid; OA) has a wide variety of bioactivities and is used for medicinal purposes in many Asian countries. Various derivatives of OA have been synthesized in attempts to improve the potency. Here we describe the anti-tumour activity of a novel OA derivative, N-[(3beta)-3-(acetyloxy)-28-oxoolean-12-en-28-yl]-glycine methyl ester (AOA-GMe). AOAGMe was a more potent inhibitor of the growth of B16 melanoma cells than its parent compound OA, both in-vitro and in-vivo. AOA-GMe also exhibited dose-dependent inhibition of human K562 leukaemia cells, but had almost no toxicity in normal human peripheral blood mononuclear cells. AOA-GMe induced cell cycle arrest in G0/G1 and blocked G1-S transition, which correlated well with marked decreases in levels of cyclin D, cyclin-dependent kinase CDK4 and phosphorylated retinoblastoma protein, and increases in the cyclin-dependent kinase inhibitor p15. OA did not show such activities. These results suggest that AOA-GMe may induce growth arrest in tumour cells through regulation of proteins involved in the cell cycle.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Glycine/analogs & derivatives , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemical synthesis , Blotting, Western , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D , Cyclin-Dependent Kinase 4/drug effects , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p15/drug effects , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclins/drug effects , Cyclins/metabolism , Dose-Response Relationship, Drug , Female , Flow Cytometry , Glycine/administration & dosage , Glycine/adverse effects , Glycine/chemical synthesis , Glycine/pharmacology , Humans , Leukocytes, Mononuclear , Medicine, East Asian Traditional , Mice , Mice, Inbred C57BL , Oleanolic Acid/administration & dosage , Oleanolic Acid/adverse effects , Oleanolic Acid/chemical synthesis , Phosphorylation , Retinoblastoma Protein/drug effects , Retinoblastoma Protein/metabolism
19.
Cancer Res ; 67(12): 5976-82, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575168

ABSTRACT

Prostate cancer chemoprevention is an alternative and potential strategy to control this malignancy. Herein, we evaluated the chemopreventive efficacy of grape seed extract (GSE) against prostate cancer in transgenic adenocarcinoma of the mouse prostate (TRAMP) mice where animals were fed with GSE by oral gavage at 200 mg/kg body weight dose during 4 to 28 weeks of age. Our results showed a significant reduction (46%, P < 0.01) in the weight of genitourinary tract organs in the GSE-fed mice. The GSE-fed group of mice had a higher incidence of prostatic intraepithelial neoplasia but showed strong reduction in the incidence of adenocarcinoma compared with mice in control group. Prostate tissue from the GSE group showed approximately 50% (P < 0.001) decrease in proliferating cell nuclear antigen (PCNA)-positive cells and 64% (P < 0.01) reduction in total PCNA protein level compared with the control group; however, GSE increased apoptotic cells by 8-fold. Furthermore, GSE strongly decreased the protein levels of cyclin B1, cyclin A, and cyclin E by 84% (P < 0.05), 96% (P < 0.05), and 89% (P < 0.001), respectively. The protein expression of cyclin-dependent kinases 2 and 6 and Cdc2 was also decreased by more than 90% (P < 0.05) in the prostate from the GSE-fed group. Together, for the first time, we identified that oral GSE inhibits prostate cancer growth and progression in TRAMP mice, which could be mediated via a strong suppression of cell cycle progression and cell proliferation and an increase in apoptosis.


Subject(s)
Adenocarcinoma/prevention & control , Antineoplastic Agents/therapeutic use , Phytotherapy , Prostatic Neoplasms/prevention & control , Seeds/chemistry , Vitis/chemistry , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Administration, Oral , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/drug effects , Cyclin-Dependent Kinases/metabolism , Cyclins/drug effects , Cyclins/metabolism , Disease Progression , Immunoblotting , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Transgenic , Plant Extracts/therapeutic use , Proliferating Cell Nuclear Antigen/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
20.
J Pharmacol Exp Ther ; 322(3): 1261-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17562851

ABSTRACT

We have studied the effects of dihydrocucurbitacin B, a triterpene isolated from Cayaponia tayuya roots, on different models of delayed type hypersensitivity (DTH) in mice, as well as on T-lymphocyte proliferation and the mediators involved. In experiments with mice, dihydrocucurbitacin B inhibited the inflammatory reactions induced by oxazolone, dinitrofluorobenzene, and sheep red blood cells, reducing both the edema and cell infiltration. Moreover, the analysis of inflamed tissues showed that dihydrocucurbitacin B reduced the presence of the most relevant cytokines implicated in these processes, including interleukin-1 beta, interleukin-4, and tumor necrosis factor-alpha. Dihydrocucurbitacin B was also found to inhibit the proliferation of phytohemagglutinin-stimulated human T lymphocytes (IC(50) = 1.48 microM), halting the cell cycle in the G(0) phase. In addition, the triterpene reduced the production of interleukin-2, interleukin-4, interleukin-10, and interferon-gamma in human T lymphocytes, and it hampered the induction of the principal cyclins involved in the cell cycle, including A(1), B(1), D(2), and E(1). Finally, dihydrocucurbitacin B was found to exert a selective inhibition on the nuclear factor of activated T cells (NFAT) in human lymphocytes without affecting the calcium influx. Taken together, these results suggest that dihydrocucurbitacin B curbs DTH reactions by inhibiting NFAT, which in turn suppresses the proliferation of the most relevant cells involved in DTH reactions, namely the T cells.


Subject(s)
Hypersensitivity, Delayed/drug therapy , Lymphocyte Activation/drug effects , Triterpenes/pharmacology , Animals , Cyclins/drug effects , Cytokines/biosynthesis , Cytokines/drug effects , Disease Models, Animal , Inflammation/prevention & control , Mice , NFATC Transcription Factors/antagonists & inhibitors , Resting Phase, Cell Cycle
SELECTION OF CITATIONS
SEARCH DETAIL
...