Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681807

ABSTRACT

Epithelioid sarcoma (ES) is a rare disease representing <1% of soft tissue sarcomas. Current therapies are based on anthracycline alone or in combination with ifosfamide or other cytotoxic drugs. ES is still characterized by a poor prognosis with high rates of recurrence. Indeed, for years, ES survival rates have remained stagnant, suggesting that conventional treatments should be revised and improved. New therapeutic approaches are focused to target the key regulators of signaling pathways, the causative markers of tumor pathophysiology. To this end, we selected, among the drugs to which an ES cell line is highly sensitive, those that target signaling pathways known to be dysregulated in ES. In particular, we found a key role for GSK-3ß, which results in up-regulation in tumor versus normal tissue samples and associated to poor prognosis in sarcoma patients. Following this evidence, we evaluated CHIR99021, a GSK-3 inhibitor, as a potential drug for use in ES therapy. Our data highlight that, in ES cells, CHIR99021 induces cell cycle arrest, mitotic catastrophe (MC) and autophagic response, resulting in reduced cell proliferation. Our results support the potential efficacy of CHIR99021 in ES treatment and encourage further preclinical and clinical studies.


Subject(s)
Autophagy/drug effects , Mitosis/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Sarcoma/pathology , Soft Tissue Neoplasms/pathology , Adult , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/physiology , Humans , Mitosis Modulators/pharmacology , Sarcoma/mortality , Soft Tissue Neoplasms/mortality , Survival Analysis
2.
Br J Cancer ; 121(2): 139-149, 2019 07.
Article in English | MEDLINE | ID: mdl-31235865

ABSTRACT

BACKGROUND: Crenolanib is a tyrosine kinase inhibitor targeting PDGFR-α, PDGFR-ß and Fms related tyrosine kinase-3 (FLT3) that is currently evaluated in several clinical trials. Although platelet-derived growth factor receptor (PDGFR) signalling pathway is believed to play an important role in angiogenesis and maintenance of functional vasculature, we here demonstrate a direct angiostatic activity of crenolanib independently of PDGFR signalling. METHODS: The activity of crenolanib on cell viability, migration, sprouting, apoptosis and mitosis was assessed in endothelial cells, tumour cells and fibroblasts. Alterations in cell morphology were determined by immunofluorescence experiments. Flow-cytometry analysis and mRNA expression profiles were used to investigate cell differentiation. In vivo efficacy was investigated in human ovarian carcinoma implanted on the chicken chorioallantoic membrane (CAM). RESULTS: Crenolanib was found to inhibit endothelial cell viability, migration and sprout length, and induced apoptosis independently of PDGFR expression. Treated cells  showed altered actin arrangement and nuclear aberrations. Mitosis was affected at several levels including mitosis entry and centrosome clustering. Crenolanib suppressed human ovarian carcinoma tumour growth and angiogenesis in the CAM model. CONCLUSIONS: The PDGFR/FLT3 inhibitor crenolanib targets angiogenesis and inhibits tumour growth in vivo unrelated to PDGFR expression. Based on our findings, we suggest a broad mechanism of action of crenolanib.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Mitosis Modulators/pharmacology , Piperidines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Chickens , Female , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Receptors, Platelet-Derived Growth Factor/analysis , Receptors, Platelet-Derived Growth Factor/physiology
3.
Int J Mol Sci ; 19(2)2018 Jan 26.
Article in English | MEDLINE | ID: mdl-29373494

ABSTRACT

Plakins are a family of seven cytoskeletal cross-linker proteins (microtubule-actin crosslinking factor 1 (MACF), bullous pemphigoid antigen (BPAG1) desmoplakin, envoplakin, periplakin, plectin, epiplakin) that network the three major filaments that comprise the cytoskeleton. Plakins have been found to be involved in disorders and diseases of the skin, heart, nervous system, and cancer that are attributed to autoimmune responses and genetic alterations of these macromolecules. Despite their role and involvement across a spectrum of several diseases, there are no current drugs or pharmacological agents that specifically target the members of this protein family. On the contrary, microtubules have traditionally been targeted by microtubule inhibiting agents, used for the treatment of diseases such as cancer, in spite of the deleterious toxicities associated with their clinical utility. The Research Collaboratory for Structural Bioinformatics (RCSB) was used here to identify therapeutic drugs targeting the plakin proteins, particularly the spectraplakins MACF1 and BPAG1, which contain microtubule-binding domains. RCSB analysis revealed that plakin proteins had 329 ligands, of which more than 50% were MACF1 and BPAG1 ligands and 10 were documented, clinically or experimentally, to have several therapeutic applications as anticancer, anti-inflammatory, and antibiotic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Microfilament Proteins/metabolism , Plakins/metabolism , Animals , Antineoplastic Agents/chemistry , Binding Sites , Humans , Microfilament Proteins/chemistry , Mitosis Modulators/chemistry , Mitosis Modulators/pharmacology , Plakins/chemistry , Protein Binding
5.
J Neurosci Methods ; 268: 106-16, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27154027

ABSTRACT

BACKGROUND: Sensorineural deafness is mainly caused by damage to hair cells and degeneration of the spiral ganglion neurons (SGN). Cochlear implants can functionally replace lost hair cells and stimulate the SGN electrically. The benefit from cochlear implantation depends on the number and excitability of these neurons. To identify potential therapies for SGN protection, in vitro tests are carried out on spiral ganglion cells (SGC). NEW METHOD: A glial cell-reduced and neuron-enhanced culture of neonatal rat SGC under mitotic inhibition (cytarabine (AraC)) for up to seven days is presented. Serum containing and neurotrophin-enriched cultures with and without AraC-addition were analyzed after 4 and 7 days. RESULTS: The total number of cells was significantly reduced, while the proportion of neurons was greatly increased by AraC-treatment. Cell type-specific labeling demonstrated that nearly all fibroblasts and most of the glial cells were removed. Neither the neuronal survival, nor the neurite outgrowth or soma diameter were negatively affected. Additionally neurites remain partly free of surrounding non-neuronal cells. COMPARISON WITH EXISTING METHOD: Recent culture conditions allow only for short-term cultivation of neonatal SGC and lack information on the influence of non-neuronal cells on SGN and of direct contact of neurites with test-materials. CONCLUSIONS: AraC-addition reduces the number of non-neuronal cells and increases the ratio of SGN in culture, without negative impact on neuronal viability. This treatment allows longer-term cultivation of SGC and provides deeper insight into SGN-glial cell interaction and the attachment of neurites on test-material surfaces.


Subject(s)
Cell Culture Techniques , Neuroglia , Neurons , Spiral Ganglion , Animals , Animals, Newborn , Cell Count , Cell Size/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytarabine/pharmacology , Female , Immunohistochemistry , Male , Mitosis Modulators/pharmacology , Neuroglia/cytology , Neuroglia/drug effects , Neuroglia/physiology , Neuronal Outgrowth/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Rats, Sprague-Dawley , Spiral Ganglion/cytology , Spiral Ganglion/drug effects , Spiral Ganglion/physiology , Time Factors
6.
Braz J Med Biol Res ; 48(5): 382-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25760027

ABSTRACT

Lung cancer often exhibits molecular changes, such as the overexpression of the ErbB1 gene that encodes epidermal growth factor receptor (EGFR). ErbB1 amplification and mutation are associated with tumor aggressiveness and low response to therapy. The aim of the present study was to design a schedule to synchronize the cell cycle of A549 cell line (a non-small cell lung cancer) and to analyze the possible association between the micronuclei (MNs) and the extrusion of ErbB1 gene extra-copies. After double blocking, by the process of fetal bovine serum deprivation and vincristine treatment, MNs formation was monitored with 5-bromo-2-deoxyuridine (BrdU) incorporation, which is an S-phase marker. Statistical analyses allowed us to infer that MNs may arise both in mitosis as well as in interphase. The MNs were able to replicate their DNA and this process seemed to be non-synchronous with the main cell nuclei. The presence of ErbB1 gene in the MNs was evaluated by fluorescent in situ hybridization (FISH). ErbB1 sequences were detected in the MNs, but a relation between the MNs formation and extrusion of amplified ErbB1 could not be established. The present study sought to elucidate the meaning of MNs formation and its association with the elimination of oncogenes or other amplified sequences from the tumor cells.


Subject(s)
Antimetabolites/metabolism , Bromodeoxyuridine/metabolism , Cell Cycle/genetics , Gene Silencing/physiology , Genes, erbB-1/genetics , Micronuclei, Chromosome-Defective , Animals , Cattle , Cell Cycle/drug effects , Cell Line, Tumor , DNA Replication , G1 Phase , Gene Amplification/physiology , Humans , In Situ Hybridization, Fluorescence , Micronuclei, Chromosome-Defective/chemically induced , Microscopy, Confocal , Mitosis Modulators/pharmacology , Mitotic Index/statistics & numerical data , S Phase , Vincristine/pharmacology
7.
Dev Neurobiol ; 74(12): 1243-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24909558

ABSTRACT

Dickkopf-3 (Dkk-3) and Dkkl-1 (Soggy) are secreted proteins of poorly understood function that are highly expressed in subsets of neurons in the brain. To explore their potential roles during neuronal development, we examined their expression in Ntera-2 (NT2) human embryonal carcinoma cells, which differentiate into neurons upon treatment with retinoic acid (RA). RA treatment increased the mRNA and protein levels of Dkk-3 but not of Dkkl-1. Ectopic expression of both Dkk-3 and Dkkl-1 induced apoptosis in NT2 cells. Gene silencing of Dkk-3 did not affect NT2 cell growth or differentiation but altered their response to RA in suspension cultures. RA treatment of NT2 cells cultured in suspension resulted in morphological changes that led to cell attachment and flattening out of cell aggregates. Although there were no significant differences in the expression levels of cell adhesion molecules in control and Dkk-3-silenced cells, this morphological response was not observed in Dkk-3-silenced cells. These findings suggest that Dkk-3 plays a role in the regulation of cell interactions during RA-induced neuronal differentiation.


Subject(s)
Embryonal Carcinoma Stem Cells/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Mitosis Modulators/pharmacology , Neurogenesis/physiology , Tretinoin/pharmacology , Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Blotting, Western , Caspase 3/metabolism , Caspase 7/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Chemokines , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/drug effects , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Transfection
8.
J Phys Chem B ; 118(10): 2605-14, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24328194

ABSTRACT

G-quadruplex-binding and telomerase-inhibiting capacities of G-quadruplex ligands were examined under a cell nuclei-mimicking condition including excess double-stranded DNA (λ DNA) and molecular crowding cosolute (PEG 200). Under the cell nuclei-mimicking condition, a cationic porphyrin (TMPyP4) did not bind to the G-quadruplex despite the high affinity (Ka = 3.6 × 10(6) M(-1)) under a diluted condition without λ DNA and PEG 200. Correspondingly, TMPyP4 inhibited telomerase activity under the diluted condition (IC50 = 1.6 µM) but not under the cell nuclei-mimicking condition. In contrast, the Ka and IC50 values of an anionic copper phthalocyanine (Cu-APC) under the diluted (2.8 × 10(4) M(-1) and 0.86 µM) and the cell nuclei-mimicking (2.8 × 10(4) M(-1) and 2.1 µM) conditions were similar. In accordance with these results, 10 µM TMPyP4 did not affect the proliferation of HeLa cells, while Cu-APC efficiently inhibited the proliferation (IC50 = 1.4 µM). These results show that the cell nuclei-mimicking condition is effective to predict capacities of G-quadruplex ligands in the cell. In addition, the antiproliferative effect of Cu-APC on normal cells was smaller than that on HeLa cells, indicating that the cell nuclei-mimicking condition is also useful to predict side effects of ligands.


Subject(s)
Cell Nucleus/metabolism , G-Quadruplexes , Telomerase/metabolism , Cell Proliferation/drug effects , DNA/metabolism , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Indoles/pharmacology , Ligands , Mitosis Modulators/pharmacology , Models, Biological , Organometallic Compounds/pharmacology , Polyethylene Glycols/metabolism , Porphyrins/metabolism , Static Electricity , Telomerase/antagonists & inhibitors , Telomere/metabolism
9.
Arch Pharm Res ; 36(11): 1377-84, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23918651

ABSTRACT

Adipocytes are the key player in adipose tissue inflammation and subsequent systemic insulin resistance and its development involves complex process of proliferation and differentiation of preadipocytes. Fistein, a polyphenol flavonoid, is known to exert anti-inflammatory, anti-carcinogenic and anti-diabetic effects. In this study, we aimed to investigate the effect of fisetin on adipocyte proliferation and differentiation in 3T3-L1 preadipocyte cell line and its mechanism of action. We found that fisetin inhibits adipocyte differentiation in a concentration dependent manner, which were evidenced by Oil Red O staining and the protein expression of mature adipocyte marker genes fatty acid synthase and peroxisome proliferator-activated receptor γ. Moreover, the proliferation of preadipocytes was also markedly suppressed by treatment of fisetin for 24 and 48 h in the differentiation medium. We also found that fisetin inhibition of adipocyte differentiation was largely due to the effect on mitotic clonal expansion. Fisetin suppression of preadipocyte proliferation at early stage of differentiation was accompanied by the changes of expression of a series of cell cycle regulatory proteins. Altogether, our results suggest that the inhibition of adipocyte differentiation by fisetin may be at least in part mediated by cell cycle arrest during adipogenesis.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Adipogenesis/drug effects , Cell Differentiation/drug effects , Flavonoids/pharmacology , Mitosis Modulators/pharmacology , Mitosis/drug effects , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Fatty Acid Synthase, Type I/metabolism , Flavonols , Mice , PPAR gamma/metabolism
10.
J Pathol ; 226(3): 482-94, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21953249

ABSTRACT

Chromosomal instability (CIN) has been implicated in multidrug resistance and the silencing of the ceramide transporter, CERT, promotes sensitization to diverse cytotoxics. An improved understanding of mechanisms governing multidrug sensitization might provide insight into pathways contributing to the death of CIN cancer cells. Using an integrative functional genomics approach, we find that CERT-specific multidrug sensitization is associated with enhanced autophagosome-lysosome flux, resulting from the expression of LAMP2 following CERT silencing in colorectal and HER2(+) breast cancer cell lines. Live cell microscopy analysis revealed that CERT depletion induces LAMP2-dependent death of polyploid cells following exit from mitosis in the presence of paclitaxel. We find that CERT is relatively over-expressed in HER2(+) breast cancer and CERT protein expression acts as an independent prognostic variable and predictor of outcome in adjuvant chemotherapy-treated patients with primary breast cancer. These data suggest that the induction of LAMP2-dependent autophagic flux through CERT targeting may provide a rational approach to enhance multidrug sensitization and potentiate the death of polyploid cells following paclitaxel exposure to limit the acquisition of CIN and intra-tumour heterogeneity.


Subject(s)
Autophagy/physiology , Breast Neoplasms/drug therapy , Chromosomal Instability/physiology , Protein Serine-Threonine Kinases/deficiency , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Breast Neoplasms/genetics , Ceramides/metabolism , Ceramides/pharmacology , Cisplatin/pharmacology , Drug Resistance, Multiple/genetics , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Female , Gene Expression , Gene Silencing/physiology , Humans , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/physiology , Middle Aged , Mitosis Modulators/pharmacology , Polyploidy , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Receptor, ErbB-2 , Tumor Cells, Cultured
11.
Methods Mol Biol ; 761: 65-74, 2011.
Article in English | MEDLINE | ID: mdl-21755441

ABSTRACT

Understanding cell growth and cell division involves the study of regulatory events that occur in a cell cycle phase-dependent manner. Studies analyzing cell cycle regulatory mechanisms and cell cycle progression invariably require synchronization of cell populations at specific cell cycle stages. Several methods have been established to synchronize cells, including serum deprivation, contact inhibition, centrifugal elutriation, and drug-dependent synchronization. Despite potential adverse cellular consequences of synchronizing cells by pharmacological agents, drug-dependent methods can be advantageous when studying later cell cycle events to ensure specific enrichment at selected mitotic stages. This chapter describes protocols used in our laboratory for isolating mitotic mammalian cells in a large-scale manner. In particular, we discuss the technical aspects of adherent or suspension cell isolation, the methods necessary to enrich cells at different mitotic stages and the optimized culture conditions.


Subject(s)
Cell Culture Techniques/methods , Mitosis/drug effects , Mitosis/genetics , Cell Cycle/physiology , Cell Line, Tumor , HeLa Cells , Humans , Mitosis Modulators/pharmacology , Staining and Labeling
12.
Methods Mol Biol ; 761: 1-23, 2011.
Article in English | MEDLINE | ID: mdl-21755438

ABSTRACT

Widespread interest in cell synchronization is maintained by the studies of control mechanisms involved in cell cycle regulation. During the synchronization distinct subpopulations of cells are obtained representing different stages of the cell cycle. These subpopulations are then used to study regulatory mechanisms of the cycle at the level of macromolecular biosynthesis (DNA synthesis, gene expression, protein synthesis), protein phosphorylation, development of new drugs, etc. Although several synchronization methods have been described, it is of general interest that scientists get a compilation and an updated view of these synchronization techniques. This introductory chapter summarizes: (1) the basic concepts and principal criteria of cell cycle synchronizations, (2) the most frequently used synchronization methods, such as physical fractionation (flow cytometry, dielectrophoresis, cytofluorometric purification), chemical blockade, (3) synchronization of embryonic cells, (4) synchronization at low temperature, (5) comparison of cell synchrony techniques, (6) synchronization of unicellular organisms, and (7) the effect of synchronization on transfection.


Subject(s)
Cell Cycle/genetics , Animals , Cell Cycle/drug effects , Cells/drug effects , Cells/metabolism , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Flow Cytometry , Humans , Mitosis Modulators/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Temperature , Transfection
13.
Methods Mol Biol ; 761: 125-36, 2011.
Article in English | MEDLINE | ID: mdl-21755445

ABSTRACT

Synchronized cells have been invaluable in many kinds of cell cycle and cell proliferation studies. Butyrate induces cell cycle arrest and apoptosis in Madin Darby Bovine Kidney (MDBK) cells. We explore the possibility of using butyrate-blocked cells to obtain synchronized cells and we characterize the properties of butyrate-induced cell cycle arrest. The site of growth inhibition and cell cycle arrest was analyzed using 5-bromo-2'-deoxyuridine (BrdU) incorporation and flow cytometry analyses. Exposure of MDBK cells to 10 mM butyrate caused growth inhibition and cell cycle arrest in a reversible manner. Butyrate affected the cell cycle at a specific point both immediately after mitosis and at a very early stage of the G1 phase. After release from butyrate arrest, MDBK cells underwent synchronous cycles of DNA synthesis and transited through the S phase. It takes at least 8 h for butyrate-induced G1-synchronized cells to begin the progression into the S phase. One cycle of cell division for MDBK cells is about 20 h. By combining BrdU incorporation and DNA content analysis, not only can the overlapping of different cell populations be eliminated, but the frequency and nature of individual cells that have synthesized DNA can also be determined.


Subject(s)
Butyrates/pharmacology , Cell Cycle/drug effects , Mitosis Modulators/pharmacology , Animals , Aphidicolin/pharmacology , Cattle , Cell Line , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Flow Cytometry , Mitosis/drug effects , Mitosis/genetics , Nucleic Acid Synthesis Inhibitors , Ploidies , Staining and Labeling
14.
Methods Mol Biol ; 761: 151-61, 2011.
Article in English | MEDLINE | ID: mdl-21755447

ABSTRACT

HeLa is one of the oldest and most commonly used cell lines in biomedical research. Owing to the ease of which they can be effectively synchronized by various methods, HeLa cells have been used extensively for studies of the cell cycle. Here we describe several protocols for synchronization of HeLa cells from different phases of the cell cycle. Synchronization in G(1) phase can be achieved with the HMG-CoA reductase inhibitor lovastatin, S phase with a double thymidine block procedure, and G(2) phase with the CDK inhibitor RO3306. Cells can also be enriched in mitosis by treating with nocodazole and mechanical shake-off. Release of the cells from these blocks enables researchers to follow gene expression and other events through the cell cycle. We also describe several protocols, including flow cytometry, BrdU labeling, immunoblotting, and time-lapse microscopy, for validating the synchrony of the cells and monitoring the progression of the cell cycle after release.


Subject(s)
Cell Cycle/drug effects , Cyclins/metabolism , Flow Cytometry , G1 Phase/drug effects , G2 Phase/drug effects , HeLa Cells , Humans , Lovastatin/pharmacology , Mitosis/drug effects , Mitosis Modulators/pharmacology , Nocodazole/pharmacology , S Phase/drug effects , Thymidine/pharmacology , Time-Lapse Imaging
15.
Methods Mol Biol ; 761: 137-49, 2011.
Article in English | MEDLINE | ID: mdl-21755446

ABSTRACT

Uracil-DNA glycosylases (UDG/UNG) are enzymes that remove uracil from DNA and initiate base-excision repair. These enzymes play a key role in maintaining genomic integrity by reducing the mutagenic events caused by G:C to A:T transition mutations. The recent finding that a family of RNA editing enzymes (AID/APOBECs) can deaminate cytosine in DNA has raised the interest in these base-excision repair enzymes. The methodology presented here focuses on determining the regulation of the nuclear isoform of uracil-DNA glycosylase (nUDG), a 36,000 Da protein. In synchronized HeLa cells, nUDG protein levels decrease to barely detectable levels during the S phase of the cell cycle. Immunoblot analysis of immunoprecipitated or affinity-isolated nUDG reveals ubiquitin-conjugated nUDG when proteolysis is inhibited by agents that block proteasomal-dependent protein degradation.


Subject(s)
Cell Cycle/physiology , Cell Nucleus/enzymology , Uracil-DNA Glycosidase/metabolism , Cell Cycle/drug effects , HEK293 Cells , HeLa Cells , Humans , Mitosis Modulators/pharmacology , Ubiquitination/physiology
16.
Methods Mol Biol ; 761: 173-200, 2011.
Article in English | MEDLINE | ID: mdl-21755449

ABSTRACT

The budding yeast Saccharomyces cerevisiae and fission yeast Schizosaccharomyces pombe are amongst the simplest and most powerful model systems for studying the genetics of cell cycle control. Because yeast grows very rapidly in simple and economical media, large numbers of cells can easily be obtained for genetic, molecular, and biochemical studies of the cell cycle. The use of synchronized cultures greatly aids in the ease and interpretation of cell cycle studies. In principle, there are two general methods for obtaining synchronized yeast populations. Block and release methods can be used to induce cell cycle synchrony. Alternatively, centrifugal elutriation can be used to select synchronous populations. Because each method has innate advantages and disadvantages, the use of multiple approaches helps in generalizing results. An overview of the most commonly used methods to generate synchronized yeast cultures is presented along with working Notes, a section that includes practical comments, experimental considerations and observations, and hints regarding the pros and cons innate to each approach.


Subject(s)
Cell Cycle/physiology , Yeasts/growth & development , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Separation , Centrifugation , Flow Cytometry , Mitosis Modulators/pharmacology , Staining and Labeling , Yeasts/drug effects , Yeasts/genetics , Yeasts/metabolism
17.
Methods Mol Biol ; 761: 211-25, 2011.
Article in English | MEDLINE | ID: mdl-21755451

ABSTRACT

When removed from the follicles, during the 44 h process of in vitro maturation (IVM) fully grown porcine oocytes resume meiosis spontaneously from the late diplotene stage of the first meiotic prophase and proceed to the metaphase-II (MII) stage at which they remain arrested until fertilization. However, the resumption may start at various times causing heterogeneity in the nuclear stage and also in cytoplasmic characteristics (i.e., the activity of certain protein kinases) within a population. Those oocytes that reach the MII stage earlier than others undergo an ageing process which is detrimental for further embryo development. The synchronization of nuclear progression is possible by a transient inhibition of meiotic resumption during the first 20-22 h of IVM either by (1) the elevation of intracellular levels of cyclic adenosine monophosphate (cAMP) or (2) suppressing the activity of the metaphase promoting factor (MPF). A protocol for each approach is described.


Subject(s)
Oocytes/growth & development , Animals , Meiosis/drug effects , Mitosis Modulators/pharmacology , Oocyte Retrieval , Oocytes/cytology , Oocytes/drug effects , Swine
18.
Methods Mol Biol ; 761: 201-10, 2011.
Article in English | MEDLINE | ID: mdl-21755450

ABSTRACT

Protozoans are single-cell eukaryotes and many of the best studied protozoans are parasitic to humans (e.g., Plasmodium falciparum causing malaria and Trypanosoma brucei causing sleeping sickness). These organisms are distantly related to humans but with retained eukaryotic type of cellular processes, making them good model systems for studies of the evolution of basic processes like the cell cycle. Giardia intestinalis causes 250 million cases of diarrhea yearly and is one of the earliest diverging protozoans. It has recently been possible to synchronize its cell cycle using compounds that inhibit different steps of the cell cycle and the detailed protocol is described here.


Subject(s)
Cell Cycle/physiology , Giardia lamblia/growth & development , Cell Cycle/drug effects , Flow Cytometry , Giardia lamblia/drug effects , Mitosis Modulators/pharmacology , Staining and Labeling
19.
Methods Mol Biol ; 761: 239-47, 2011.
Article in English | MEDLINE | ID: mdl-21755453

ABSTRACT

Somatic cell nuclear transfer (SCNT) is a technically and biologically challenging procedure during which a differentiated committed nucleus undergoes rapid reprogramming into the totipotent state in a few hours. SCNT can be utilized to generate patient- and disease-specific embryonic stem cell (ESC) lines, which carry great promise in improving our understanding of major disease conditions and hope for better therapies. In this section, we will describe how mouse SCNT is performed and survey the importance of donor cell cycle synchronization and the methods to perform it.


Subject(s)
Cell Cycle/genetics , Nuclear Transfer Techniques , Animals , Cell Cycle/drug effects , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cultural Deprivation , Culture Media, Serum-Free , Female , Humans , Mice , Mitosis Modulators/pharmacology
20.
J Reprod Dev ; 57(1): 49-56, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20834195

ABSTRACT

Mitogen-activated protein kinase (MAPK) and maturation/M phase promoting factor (MPF) play crucial roles in oocyte meiotic maturation in mammals. However, the underlying molecular mechanisms have not been addressed. In this study, the effects of the MEK/MAPK pathway inhibitor U0126 and the MPF inhibitor roscovitine on meiotic maturation and maternal gene expression in pig cumulus-oocyte complexes (COCs) and denuded oocytes (DOs) were investigated. Both inhibitors can reversibly block the resumption of meiosis in pig oocytes. COCs or DOs initially cultured in drug-free medium for 24 h and then transferred to medium containing U0126 showed normal progress to the Metaphase II stage (MII); (90.38 vs. 92.16% control group). In contrast, roscovitine treatment from 24 to 44 h significantly inhibited maturation of COCs and DOs. To explore the underlying molecular mechanisms, expression patterns and polyadenylation states of five representative maternal transcripts, cyclin B1, Cdc2, C-mos, GDF9 and BMP15, were examined by real-time PCR and poly(A)-test PCR (PAT assay). U0126 treatment resulted in aberrant expression of Cdc2 and GDF9, while roscovitine significantly maintained all five maternal transcripts at very high levels in treated COCs compared with the control group. The polyadenylation of these mRNAs increased as well. Furthermore, the experiments were repeated in DOs, and the results also indicated that both Cdc2 and GDF9 showed significantly higher expression in both mRNA and polyadenylation levels in the drug treatment groups. Together, these results provide the first demonstration in a mammalian system that MAPK and MPF play important roles in regulation of maternal gene expression during oocyte maturation.


Subject(s)
Gene Expression Regulation, Developmental , MAP Kinase Signaling System , Maturation-Promoting Factor/metabolism , Mitosis , Oocytes/metabolism , Oogenesis , Animals , Bone Morphogenetic Protein 15/genetics , Bone Morphogenetic Protein 15/metabolism , Butadienes/pharmacology , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cumulus Cells/physiology , Cyclin B1/genetics , Cyclin B1/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Growth Differentiation Factor 9/genetics , Growth Differentiation Factor 9/metabolism , MAP Kinase Signaling System/drug effects , Maturation-Promoting Factor/antagonists & inhibitors , Mitosis/drug effects , Mitosis Modulators/pharmacology , Nitriles/pharmacology , Oocytes/drug effects , Oogenesis/drug effects , Polyadenylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-mos/genetics , Proto-Oncogene Proteins c-mos/metabolism , Purines/pharmacology , RNA, Messenger/metabolism , Roscovitine , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...