Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
2.
Mini Rev Med Chem ; 21(7): 792-802, 2021.
Article in English | MEDLINE | ID: mdl-33238842

ABSTRACT

CDATA[The inherited mutations and underexpression of BRCA1 in sporadic breast cancers resulting in the loss or functional inactivation of BRCA1 may contribute to a high risk of breast cancer. Recent researchers have identified small molecules (BRCA1 mimetics) that fit into a BRCA1 binding pocket within Estrogen Receptor alpha (ERα), mimic the ability of BRCA1 to inhibit ERα activity, and overcome antiestrogen resistance. Studies indicate that most of the BRCA1 breast cancer cases are associated with p53 mutations. It indicates that there is a potential connection between BRCA1 and p53. Most p53 mutations are missense point mutations that occur in the DNA-binding domain. Structural studies have demonstrated that mutant p53 core domain misfolding, especially p53-R175H, is reversible. Mutant p53 reactivation with a new class of zinc metallochaperones (ZMC) restores WT p53 structure and functions by restoring Zn2+ to Zn2+ deficient mutant p53. Considering the role of WT BRCA1 and reactivation of p53 in tumor cells, our hypothesis is to target both tumor suppressor proteins by a novel biomolecule (ZMC). Since both proteins are present in the same cell and are functionally inactive, this state may be a novel efficacious therapeutic regime for breast cancer therapy. In addition, we propose to use Albumin Nanovector (ANV) formulation for target drug release.


Subject(s)
Albumins/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Molecular Chaperones/pharmacology , Nanoparticles/chemistry , Zinc/pharmacology , Antineoplastic Agents/chemistry , Cyclin D/antagonists & inhibitors , Cyclin D/metabolism , Drug Carriers/chemistry , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Female , Humans , Molecular Chaperones/chemistry , Signal Transduction/drug effects , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Zinc/chemistry
3.
Clin Transl Oncol ; 23(3): 612-619, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32710211

ABSTRACT

BACKGROUND: Glioblastoma (GB) remains an incurable and deadly brain malignancy that often proves resistant to upfront treatment with temozolomide. Nevertheless, temozolomide remains the most commonly prescribed FDA-approved chemotherapy for GB. The DNA repair protein methylguanine-DNA methyl transferase (MGMT) confers resistance to temozolomide. Unsurprisingly temozolomide-resistant tumors tend to possess elevated MGMT protein levels or lack inhibitory MGMT promotor methylation. In this study, cultured human temozolomide resistance GB (43RG) cells were introduced to the MGMT inhibitor O6-benzylguanine combined with temozolomide and either LY2835219 (CDK 4/6 inhibitor) or LY2157299 (TGF-ßRI inhibitor) seeking to overcome GB treatment resistance. METHODS: Treatment effects were assessed using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, western blot, cell viability, and cell cycle progression. RESULTS: Our in vitro study demonstrated that sequential treatment of O6-Benzylguanine with either LY2385219 or LY2157299-enhanced temozolomide enhanced sensitivity in MGMT+ 43RG cells. Importantly, normal human neurons and astrocytes remained impervious to the drug therapies under these conditions. Furthermore, LY2835219 has additional anti-proliferative effects on cell cycling, including induction of an RB-associated G (1) arrest via suppression of cyclin D-CDK4/6-Rb pathway. LY2157299 enhances anti-tumor effect by disrupting TGF-ß-dependent HIF-1α signaling and by activating both Smad and PI3K-AKT pathways towards transcription of S/G2 checkpoints. CONCLUSION: This study establishes the groundwork for the development of a combinatorial pharmacologic approach by using either LY2385219 or LY2157299 inhibitor plus O6-Benzylguanine to augment temozolomide response in temozolomide-resistant GB cells.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , DNA Modification Methylases/antagonists & inhibitors , DNA Repair Enzymes/antagonists & inhibitors , Glioblastoma/drug therapy , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Temozolomide/pharmacology , Tumor Suppressor Proteins/antagonists & inhibitors , Aminopyridines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Astrocytes/drug effects , Benzimidazoles/pharmacology , Brain Neoplasms/enzymology , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclin D/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , G1 Phase Cell Cycle Checkpoints , Glioblastoma/enzymology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Neurons/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Pyrazoles/pharmacology , Quinolines/pharmacology , Smad Proteins/drug effects
4.
Adv Cancer Res ; 148: 147-169, 2020.
Article in English | MEDLINE | ID: mdl-32723562

ABSTRACT

The mammalian cell cycle is driven by a complex of cyclins and their associated cyclin-dependent kinases (CDKs). Abnormal dysregulation of cyclin-CDK is a hallmark of cancer. D-type cyclins and their associated CDKs (CDK4 and CDK6) are key components of cell cycle machinery in driving G1 to S phase transition via phosphorylating and inactivating the retinoblastoma protein (RB). A body of evidence shows that the cyclin Ds-CDKs axis plays a critical role in cancer through various aspects, such as control of proliferation, senescence, migration, apoptosis, and angiogenesis. CDK4/6 dual-inhibitors show significant efficacy in pre-clinical or clinical cancer therapies either as single agents or in combination with hormone, chemotherapy, irradiation or immune treatments. Of note, as the associated partner of D-type cyclins, CDK6 shows multiple distinct functions from CDK4 in cancer. Depletion of the individual CDK may provide a therapeutic strategy for patients with cancer.


Subject(s)
Cyclin D/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Animals , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Humans , Neoplasms/enzymology , Phosphorylation , Protein Kinase Inhibitors/pharmacology
5.
Cell Cycle ; 18(21): 3030-3043, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31544588

ABSTRACT

This study was designed to purify molecules possess anti-cancer cell activity from the fruit body of Ganoderma leucocontextum. Bio-activity-guided purification and chromatographic separation of Ganoderma leucocontextum extract led to the enrichment of bioactive fractions and isolation of a single compound. The purified compound was identified as Ganoderiol F, which induced cancer cell death. In the in vivo experiments, we founded ethanol extract and ethyl acetate fraction inhibited tumor growth in the mice injected with 4T1 cells. We found that Ganoderiol F-mediated suppression of breast cancer cell viability occurred through cell cycle arrest. Ganoderiol F down-regulated expression of cyclin D, CDK4, CDK6, cyclin E and CDK2 and inhibited cell cycle progression arresting the cells in G1 phase. In addition, Ganoderiol F up-regulated pro-apoptotic Foxo3, down-regulated anti-apoptotic c-Myc, Bcl-2 and Bcl-w leading to apoptosis in human breast cancer cells MDA-MB-231. These results showed that c-Myc, cyclin D-CDK4/CDK6 and cyclin E-CDK2 are the central components of Ganoderiol F regulation of cell cycle progression. Hence Ganoderiol F may serve as a potential CDK4/CDK6 inhibitor for breast cancer therapy. Abbreviations: GLE: Ganoderma leucocontextum ethanol extract; GLEA: Ganoderma leucocontextum ethyl acetate fraction; GLPE: Ganoderma leucocontextum petroleum ether fraction; RP-HPLC: reversed-phase high-performance liquid chromatograph; DMEM: Dulbecco's modified Eagle's medium; FBS: fetal bovine serum; PAGE: polyacrylamide gel electrophoresis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Extracts/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Cyclin D/antagonists & inhibitors , Cyclin E/antagonists & inhibitors , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Female , Fruiting Bodies, Fungal/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Ganoderma/chemistry , Humans , Medicine, Chinese Traditional , Mice , Mice, Inbred BALB C
6.
Environ Toxicol ; 33(1): 41-51, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28941016

ABSTRACT

Vitis thunbergii var. taiwaniana (VTT) is a wild grape native to Taiwan, belonging to the Vitaceae family and Vitis genus, and widely used as folk herbal medicine. It is traditionally used for the treatment of diarrhea, hypertension, neuroprotection, jaundice, and arthritis. We used the wild-collected VTT and sterilized them to establish the plant tissue culture, and then took the leaves for DNA sequencing to determine its original base. We use methanol to extract VTT in four different solvents: 1-butanol, n-hexane, ethyl acetate, and water. These four preliminary extracts were used to treat human prostate cancer DU145 cells in vitro. We use the flow cytometry to check the cell survival situation. Finally, we found the ethyl acetate layer roughing product (referred VTEA) in human prostate cancer apoptotic effects of cell line DU-145. In the present studies, we use the crude extract of VTT to examine whether or not it can induce apoptosis of DU145 cells in vitro. Viability assays for extracts of VTT treatment showed that it had dose-dependent effect on human prostate cancer DU145 cells. We also found that the extract of VTT induces time-dependent mitochondrial and intrinsic-dependent apoptosis pathways. The in vitro cytotoxic effects were investigated by cell cycle analysis and the determination of apoptotic DNA fragmentation in DU145 cells. The cell cycle analysis showed that extracts of VTT induced a significant increase in the number of cells in G0 /G1 phase. The extract of VTT induced chromatin changes and apoptosis of DU145 cells also were confirmed by DAPI and PI staining that were measured by fluorescence microscopy and flow cytometry, respectively. Finally, the expression of relevant proteins was analyzed by Western blot analysis. These results promoted us to further evaluate apoptosis associated proteins and elucidate the possible signal pathway in DU-145 cells after treated with the extract of VTT.


Subject(s)
Apoptosis/drug effects , Cyclin D/metabolism , Cyclin E/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Plant Extracts/pharmacology , Signal Transduction/drug effects , Vitis/chemistry , Acetates/chemistry , Caspases/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cyclin D/antagonists & inhibitors , Cyclin E/antagonists & inhibitors , DNA Fragmentation/drug effects , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Methanol/chemistry , Microscopy, Confocal , Mitochondria/drug effects , Mitochondria/metabolism , Plant Extracts/analysis , Plant Extracts/chemistry , Prostatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Taiwan , Vitis/metabolism
7.
Biosci Biotechnol Biochem ; 80(6): 1095-106, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26924430

ABSTRACT

Angiogenesis, neovascularization from pre-existing vessels, is a key step in tumor growth and metastasis, and anti-angiogenic agents that can interfere with these essential steps of cancer development are a promising strategy for human cancer treatment. In this study, we characterized the anti-angiogenic effects of Coptis japonica Makino extract (CJME) and its mechanism of action. CJME significantly inhibited the proliferation, migration, and invasion of vascular endothelial growth factor (VEGF)-stimulated HUVECs. Furthermore, CJME suppressed VEGF-induced tube formation in vitro and VEGF-induced microvessel sprouting ex vivo. According to our study, CJME blocked VEGF-induced cell cycle transition in G1. CJME decreased expression of cell cycle-regulated proteins, including Cyclin D, Cyclin E, Cdk2, and Cdk4 in response to VEGF. Taken together, the results of our study indicate that CJME suppresses VEGF-induced angiogenic events such as proliferation, migration, and tube formation via cell cycle arrest in G1.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Coptis/chemistry , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation/drug effects , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/isolation & purification , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D/antagonists & inhibitors , Cyclin D/genetics , Cyclin D/metabolism , Cyclin E/antagonists & inhibitors , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Human Umbilical Vein Endothelial Cells , Humans , Male , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Plant Extracts/chemistry , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/pharmacology
8.
J Proteome Res ; 14(11): 4594-602, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26491887

ABSTRACT

Many studies have shown the Na(+)/K(+)-ATPase (NKA) might be a potential target for anticancer therapy. Cardiac glycosides (CGs), as a family of naturally compounds, inhibited the NKA activity. The present study investigates the antitumor effect of ouabain and elucidates the pharmacological mechanisms of CG activity in liver cancer HepG2 cell using SILAC coupled to LC-MS/MS method. Bioinformatics analysis of 330 proteins that were changed in cells under treatment with 0.5 µmol/L ouabain showed that the biological processes are associated with an acute inflammatory response, cell cycle, oxidation reduction, chromosome segregation, and DNA metabolism. We confirmed that ouabain induced chromosome segregation disorder and S-cell cycle block by decreasing the expression of AURKA, SMC2, Cyclin D, and p-CDK1 as well as increasing the expression of p53. We found that the overexpression or inhibition of AURKA significantly reduced or enhanced the ouabain-mediated the anticancer effects. Our findings suggest that AURKA is involved in the anticancer mechanisms of ouabain in HepG2 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Ouabain/pharmacology , S Phase/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Animals , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , CDC2 Protein Kinase , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Chromatography, Liquid , Chromosome Segregation/drug effects , Cyclin D/antagonists & inhibitors , Cyclin D/genetics , Cyclin D/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Female , Gene Regulatory Networks/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Nude , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , S Phase/genetics , Signal Transduction , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Tandem Mass Spectrometry , Tumor Suppressor Protein p53/agonists , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 21(13): 2905-10, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25941111

ABSTRACT

Cancer cells bypass normal controls over mitotic cell-cycle progression to achieve a deregulated state of proliferation. The retinoblastoma tumor suppressor protein (pRb) governs a key cell-cycle checkpoint that normally prevents G1-phase cells from entering S-phase in the absence of appropriate mitogenic signals. Cancer cells frequently overcome pRb-dependent growth suppression via constitutive phosphorylation and inactivation of pRb function by cyclin-dependent kinase (CDK) 4 or CDK6 partnered with D-type cyclins. Three selective CDK4/6 inhibitors, palbociclib (Ibrance; Pfizer), ribociclib (Novartis), and abemaciclib (Lilly), are in various stages of development in a variety of pRb-positive tumor types, including breast cancer, melanoma, liposarcoma, and non-small cell lung cancer. The emerging, positive clinical data obtained to date finally validate the two decades-old hypothesis that the cyclin D-CDK4/6 pathway is a rational target for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cyclin D/antagonists & inhibitors , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/therapeutic use , Signal Transduction
10.
Pigment Cell Melanoma Res ; 27(2): 275-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24289491

ABSTRACT

Expression profiling of microRNAs in melanoma lesional skin biopsies compared with normal donor skin biopsies, as well as melanoma cell lines compared with normal melanocytes, revealed that hsa-miR-206 was down-regulated in melanoma (-75.4-fold, P = 1.7 × 10(-4)). MiR-206 has been implicated in a large number of cancers, including breast, lung, colorectal, ovarian, and prostate cancers; however, its role in tumor development remains largely unknown, its biologic function is poorly characterized, and its targets affecting cancer cells are largely unknown. MiR-206 reduced growth and migration/invasion of multiple melanoma cell lines. Bioinformatics identified cell cycle genes CDK2, CDK4, Cyclin C, and Cyclin D1 as strong candidate targets. Western blots and 3'UTR reporter gene assays revealed that miR-206 inhibited translation of CDK4, Cyclin D1, and Cyclin C. Additionally, hsa-miR-206 transfection induced G1 arrest in multiple melanoma cell lines. These observations support hsa-miR-206 as a tumor suppressor in melanoma and identify Cyclin C, Cyclin D1, and CDK4 as miR-206 targets.


Subject(s)
Cyclin D/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , G1 Phase Cell Cycle Checkpoints/genetics , Melanoma/genetics , Melanoma/pathology , MicroRNAs/metabolism , Skin Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Base Sequence , Biopsy , Carcinogenesis/genetics , Carcinogenesis/pathology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Movement/genetics , Cell Proliferation , Computational Biology , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Melanoma/enzymology , MicroRNAs/genetics , Middle Aged , Models, Biological , Molecular Sequence Data , Neoplasm Invasiveness , Protein Biosynthesis , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Tissue Donors
11.
Cell Rep ; 5(4): 961-73, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24268780

ABSTRACT

The subgranular zone (SGZ) of the adult hippocampus contains a pool of quiescent neural progenitor cells (QNPs) that are capable of entering the cell cycle and producing newborn neurons. The mechanisms that control the timing and extent of adult neurogenesis are not well understood. Here, we show that QNPs of the adult SGZ express molecular-clock components and proliferate in a rhythmic fashion. The clock proteins PERIOD2 and BMAL1 are critical for proper control of neurogenesis. The absence of PERIOD2 abolishes the gating of cell-cycle entrance of QNPs, whereas genetic ablation of bmal1 results in constitutively high levels of proliferation and delayed cell-cycle exit. We use mathematical model simulations to show that these observations may arise from clock-driven expression of a cell-cycle inhibitor that targets the cyclin D/Cdk4-6 complex. Our findings may have broad implications for the circadian clock in timing cell-cycle events of other stem cell populations throughout the body.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Clocks/genetics , Hippocampus/growth & development , Neurogenesis/genetics , Period Circadian Proteins/genetics , ARNTL Transcription Factors/biosynthesis , Animals , Cell Cycle/genetics , Cell Proliferation , Cyclin D/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells , Neurons/cytology , Period Circadian Proteins/biosynthesis , Promoter Regions, Genetic
12.
Cancer Cell ; 22(4): 438-51, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23079655

ABSTRACT

D-cyclins represent components of cell cycle machinery. To test the efficacy of targeting D-cyclins in cancer treatment, we engineered mouse strains that allow acute and global ablation of individual D-cyclins in a living animal. Ubiquitous shutdown of cyclin D1 or inhibition of cyclin D-associated kinase activity in mice bearing ErbB2-driven mammary carcinomas triggered tumor cell senescence, without compromising the animals' health. Ablation of cyclin D3 in mice bearing Notch1-driven T cell acute lymphoblastic leukemias (T-ALL) triggered tumor cell apoptosis. Such selective killing of leukemic cells can also be achieved by inhibiting cyclin D associated kinase activity in mouse and human T-ALL models. Inhibition of cyclin D-kinase activity represents a highly-selective anticancer strategy that specifically targets cancer cells without significantly affecting normal tissues.


Subject(s)
Cyclin D/physiology , Neoplasms/etiology , Animals , Apoptosis , Cell Cycle Checkpoints , Cyclin D/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Female , Humans , Mammary Neoplasms, Experimental/etiology , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Receptor, ErbB-2/analysis
13.
Mol Vis ; 18: 758-64, 2012.
Article in English | MEDLINE | ID: mdl-22509106

ABSTRACT

PURPOSE: Nerve growth factor (NGF) plays an important role in promoting the healing of corneal wounds. However, the molecular mechanism by which NGF functions is unknown. We investigated the possible effects of NGF on phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) and mitogen activated protein kinase (MAPK)/extracellular signal-regulated kinase (Erk) pathways and cell growth in human corneal epithelial cells (HCECs). METHODS: We examined the effect of NGF on cell cycle and proliferation in HCECs by flow cytometry and cell proliferation assay, respectively. The levels of D-type cyclins in NGF-treated HCECs were determined by western blot. The tyrosine kinase A (TrkA), PI3K/Akt and MAPK/Erk pathways were then checked in cells stimulated with NGF for different time periods or cells undergoing a dose-dependent treatment. Furthermore, HCECs were treated with pathway inhibitors, LY294002 or PD98059, to confirm NGF-induced activations. RESULTS: We found that NGF had a positive effect on the growth of HCECs, and D-type cyclins, and it was correlated with the percentage of the G(1) to S progression. We also observed a time-dependent and dose-dependent effect of NGF on the PI3K/Akt and MAPK/Erk pathways. Furthermore, NGF affected cell cycle progression of HCECs by regulating cyclin D through Akt and Erk activation upon treatment with the pathway inhibitors, LY294002 for Akt or PD98059 for Erk pathways. CONCLUSIONS: NGF stimulation could promote cell proliferation and cell cycle progression of HCECs by activation of cyclin D via the PI3K/Akt and MAPK/Erk signaling pathways.


Subject(s)
Cell Cycle/drug effects , Cornea/cytology , Epithelial Cells/cytology , Nerve Growth Factor/pharmacology , Signal Transduction/drug effects , Cell Proliferation/drug effects , Chromones/pharmacology , Cornea/drug effects , Cornea/metabolism , Cyclin D/antagonists & inhibitors , Cyclin D/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Primary Cell Culture , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Time Factors
14.
Cell Cycle ; 11(3): 554-68, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22293404

ABSTRACT

Drosophila cyclinD (CycD) is the single fly ortholog of the mammalian cyclin D1 and promotes both cell cycle progression and cellular growth. However, little is known about how CycD promotes cell growth. We show here that CycD/Cdk4 hyperactivity leads to increased mitochondrial biogenesis (mitobiogenesis), mitochondrial mass, NRF-1 activity (Tfam transcript levels) and metabolic activity in Drosophila, whereas loss of CycD/Cdk4 activity has the opposite effects. Surprisingly, both CycD/Cdk4 addition and loss of function increase mitochondrial superoxide production and decrease lifespan, indicating that an imbalance in mitobiogenesis may lead to oxidative stress and aging. In addition, we provide multiple lines of evidence indicating that CycD/Cdk4 activity affects the hypoxic status of cells and sensitizes animals to hypoxia. Both mitochondrial and hypoxia-related effects can be detected at the global transcriptional level. We propose that mitobiogenesis and the hypoxic stress response have an antagonistic relationship, and that CycD/Cdk4 levels regulate mitobiogenesis contemporaneous to the cell cycle, such that only when cells are sufficiently oxygenated can they proliferate.


Subject(s)
Aging , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Drosophila Proteins/metabolism , Hypoxia , Mitochondria/metabolism , ATP Synthetase Complexes/metabolism , Animals , Cyclin D/antagonists & inhibitors , Cyclin D/genetics , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/genetics , DNA, Mitochondrial/metabolism , Drosophila/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Female , L-Lactate Dehydrogenase/metabolism , Male , Mitochondria/genetics , NF-E2-Related Factor 1/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Superoxides/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Chem Inf Model ; 52(1): 76-83, 2012 Jan 23.
Article in English | MEDLINE | ID: mdl-22172011

ABSTRACT

The cyclin dependent kinases (CDKs), each with their respective regulatory partner cyclin that are involved in the regulation of the cell cycle, apoptosis, and transcription, are potentially interesting targets for cancer therapy. The CDK6 complex with cyclin D (CDK6/cycD) drives cellular proliferation by phosphorylation of specific key target proteins. To understand the flavonoids that inhibit the CDK6/cycD functions, molecular dynamics simulations (MDSs) were performed on three inhibitors, fisetin (FST), apigenin (AGN), and chrysin (CHS), complexed with CDK6/cycD, including the two different binding orientations of CHS: FST-like (CHS_A) and deschloro-flavopiridol-like (CHS_B). For all three inhibitors, including both CHS orientations, the conserved interaction between the 4-keto group of the flavonoid and the backbone V101 nitrogen of CDK6 was strongly detected. The 3'- and 4'-OH groups on the flavonoid phenyl ring and the 3-OH group on the benzopyranone ring of inhibitor were found to significantly increase the binding and inhibitory efficiency. Besides the electrostatic interactions, especially through hydrogen bond formation, the van der Waals (vdW) interactions with the I19, V27, F98, H100, and L152 residues of CDK6 are also important factors in the binding efficiency of flavonoids against the CDK6/cycD complex. On the basis of the docking calculation and MM-PBSA method, the order of the predicted inhibitory affinities of these three inhibitors toward the CDK6/cycD was FST > AGN > CHS, which is in good agreement with the experimental data. In addition, CHS preferentially binds to the active CDK6 in a different orientation to FST and AGN but similar to its related analog, deschloro-flavopiridol. The obtained results are useful as the basic information for the further design of potent anticancer drugs specifically targeting the CDK6 enzyme.


Subject(s)
Apigenin/chemistry , Cyclin D/chemistry , Cyclin-Dependent Kinase 6/chemistry , Flavonoids/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Apigenin/pharmacology , Binding Sites , Crystallography, X-Ray , Cyclin D/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Flavonoids/pharmacology , Flavonols , Humans , Hydrogen Bonding , Molecular Conformation , Molecular Dynamics Simulation , Piperidines/chemistry , Piperidines/pharmacology , Protein Binding , Static Electricity , Thermodynamics
16.
Eur J Med Chem ; 46(12): 5825-32, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22000924

ABSTRACT

The design and synthesis of a small library of 4-aminopyrido[2,3-d]pyrimidine derivatives is reported. The potential activity of these compounds as CDK2/Cyclin A, CDK4/Cyclin D, EGFR and anti-tumor was evaluated by cytotoxicity studies in A431a, SNU638b, HCT116 and inhibition of CDK2-Cyclin A, CDK4/Cyclin D and EGFR enzyme activity in vitro. The anti-proliferative and CDK2-Cyclin A inhibitory activity of compounds 4c and 11a was significantly more active than roscovotine with IC(50) 0.3 and 0.09 µM respectively. Molecular modeling study, including fitting to a 3D-pharmacophore model, docking into cyclin dependant kinase2 (CDK2) active site and binding energy calculations were carried out and these studies suggested the same binding orientation inside the CDK2 binding pocket for these analogs compared to ATP.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Pyrimidines/chemistry , Pyrimidines/pharmacology , Antineoplastic Agents/chemical synthesis , Binding Sites , Cell Line, Tumor , Cyclin A/antagonists & inhibitors , Cyclin A/metabolism , Cyclin D/antagonists & inhibitors , Cyclin D/metabolism , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Neoplasms/drug therapy , Pyrimidines/chemical synthesis
17.
Anticancer Res ; 31(9): 2821-32, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21868525

ABSTRACT

Gallic acid (GA) induces apoptosis in different types of cancer cell lines. In this study, we investigate the apoptotic effects induced by GA in human promyelocytic leukemia HL-60 cells, and clarify the underlying mechanism. Our results showed that GA reduced the viability of HL-60 cells in a dose- and time-dependent manner. GA led to G(0)/G(1) phase arrest in HL-60 cells through promoting p21 and p27 and inhibiting the levels of cyclin D and cyclin E. GA caused DNA damage and fragmentation in HL-60 cells as assayed using DAPI staining and Comet assay. Flow cytometric analysis revealed that GA increased Ca(2+) levels and reduced the mitochondrial membrane potential (ΔΨ(m)) in HL-60 cells. Apoptotic protein expressions were determined by Western blotting. The results indicated that GA-mediated apoptosis of HL-60 cells mainly depended on mitochondrial pathway, by promoting the release of cytochrome c, apoptosis-inducing factor (AIF) and endonuclease G (Endo G) and by up-regulating the protein expression of Bcl-2-associated X protein (BAX), caspase-4, caspase-9 and caspase-3. In addition, GA also activated the death receptor-dependent pathway by enhancing the protein expressions of fatty acid synthase (FAS), FAS ligand (FASL), caspase-8 and BCL-2 interacting domain (BID). We determined the mRNA expression of the gene levels of these proteins by real-time PCR. The results showed that GA-mediated apoptosis of HL-60 cells mainly depended on up-regulation of the mRNA of caspase-8, caspase-9, caspase-3, AIF and Endo G. In conclusion, GA-induced apoptosis occurs through the death receptor and mitochondria-mediated pathways. The evaluation of GA as a potential therapeutic agent for treatment of leukemia seems warranted.


Subject(s)
Apoptosis/drug effects , Cyclin D/antagonists & inhibitors , Cyclin E/antagonists & inhibitors , G1 Phase/drug effects , Gallic Acid/pharmacology , Mitochondria/metabolism , Resting Phase, Cell Cycle/drug effects , Base Sequence , Comet Assay , DNA Primers , Flow Cytometry , HL-60 Cells , Humans , Polymerase Chain Reaction
18.
Cancer Biol Ther ; 11(7): 615-26, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21278493

ABSTRACT

The c-Myc protein, encoded by c-myc gene, in its wild-type form can induce tumors with a high frequency and can induce massive programmed cell death (PCD) in most transgenic mouse models, with greater efficiency than other oncogenes. Evidence also indicates that c-Myc can cause proliferative inhibition, i.e. mitoinhibition. The c-Myc-induced PCD and mitoinhibition, which may be attributable to its inhibition of cyclin D1 and induction of p53, may impose a pressure of compensatory proliferation, i.e. regeneration, onto the initiated cells (cancer progenitor cells) that occur sporadically and are resistant to the mitoinhibition. The initiated cells can thus proliferate robustly and progress to a malignancy. This hypothetical thinking, i.e. the concurrent PCD and mitoinhibition induced by c-Myc can promote carcinogenesis, predicts that an optimal balance is achieved between cell death and ensuing regeneration during oncogenic transformation by c-Myc, which can better promote carcinogenesis. In this perspective, we summarize accumulating evidence and challenge the current model that oncoprotein induces carcinogenesis by promoting cellular proliferation and/or inhibiting PCD. Inspired by c-myc oncogene, we surmise that many tumor-suppressive or growth-inhibitory genes may also be able to promote carcinogenesis in a similar way, i.e. by inducing PCD and/or mitoinhibition of normal cells to create a need for compensatory proliferation that drives a robust replication of initiating cells.


Subject(s)
Apoptosis/genetics , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Animals, Genetically Modified , Apoptosis Regulatory Proteins/metabolism , Cell Proliferation , Cyclin D/antagonists & inhibitors , Cyclin D/metabolism , Humans , Oncogene Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
19.
Mol Pharmacol ; 79(4): 672-80, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21220410

ABSTRACT

Amitriptyline is a classic tricyclic antidepressant (TCA) and has been used to treat the depression and anxiety of patients with cancer, but its relevance to cancer cell apoptosis is not known. In the present study, we demonstrated that amitriptyline inhibited cyclin D2 transactivation and displayed potential antimyeloma activity by inhibiting histone deacetylases (HDACs). Amitriptyline markedly decreased cyclin D2 promoter-driven luciferase activity, reduced cyclin D2 expression, and arrested cells at the G(0)/G(1) phase of the cell cycle. Amitriptyline-induced apoptosis was confirmed by Annexin V staining, and cleavage of caspase-3 and poly(ADP-ribose) polymerase-1. D-Cyclin expression is reported to be epigenetically regulated by histone acetylation. Thus, we examined the effects of amitriptyline on histone 3 (H3) acetylation and demonstrated that amitriptyline increased acetylation of H3 and expression of p27 and p21. Further studies indicated that amitriptyline interfered with HDAC function by down-regulation of HDAC3, -6, -7, and -8, but not HDAC2, and by interacting with HDAC7. Molecular docking analysis and molecular dynamics simulations revealed that amitriptyline bound to HDAC7 and formed strong van der Waals interactions with five residues of HDAC7, including Phe162, His192, Phe221, Leu293, and His326, thus inhibiting HDAC activity. Therefore, we found that amitriptyline inhibited cyclin D2 transactivation and HDAC activity and could be a promising treatment for multiple myeloma.


Subject(s)
Amitriptyline/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Apoptosis/drug effects , Cyclin D/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Multiple Myeloma/metabolism , Apoptosis/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cyclin D/metabolism , Histone Deacetylases/metabolism , Humans , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology
20.
Blood ; 117(6): 1986-97, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21135258

ABSTRACT

D-cyclins are universally dysregulated in multiple myeloma and frequently overexpressed in leukemia. To better understand the role and impact of dysregulated D-cyclins in hematologic malignancies, we conducted a high-throughput screen for inhibitors of cyclin D2 transactivation and identified 8-ethoxy-2-(4-fluorophenyl)-3-nitro-2H-chromene (S14161), which inhibited the expression of cyclins D1, D2, and D3 and arrested cells at the G(0)/G(1) phase. After D-cyclin suppression, S14161 induced apoptosis in myeloma and leukemia cell lines and primary patient samples preferentially over normal hematopoietic cells. In mouse models of leukemia, S14161 inhibited tumor growth without evidence of weight loss or gross organ toxicity. Mechanistically, S14161 inhibited the activity of phosphoinositide 3-kinase in intact cells and the activity of the phosphoinositide 3-kinases α, ß, δ, and γ in a cell-free enzymatic assay. In contrast, it did not inhibit the enzymatic activities of other related kinases, including the mammalian target of rapamycin, the DNA-dependent protein kinase catalytic subunit, and phosphoinositide-dependent kinase-1. Thus, we identified a novel chemical compound that inhibits D-cyclin transactivation via the phosphoinositide 3-kinase/protein kinase B signaling pathway. Given its potent antileukemia and antimyeloma activity and minimal toxicity, S14161 could be developed as a novel agent for blood cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Cyclin D/antagonists & inhibitors , Cyclin D/genetics , Leukemia/drug therapy , Leukemia/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Phosphoinositide-3 Kinase Inhibitors , Transcriptional Activation/drug effects , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Base Sequence , Benzopyrans/chemistry , Biological Transport, Active/drug effects , Cell Line, Tumor , Cell Membrane/metabolism , DNA Primers/genetics , Drug Evaluation, Preclinical , G1 Phase/drug effects , Humans , K562 Cells , Leukemia/genetics , Leukemia/pathology , Mice , Mice, SCID , Molecular Structure , Multiple Myeloma/genetics , Multiple Myeloma/pathology , NIH 3T3 Cells , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...