Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Braz. J. Pharm. Sci. (Online) ; 59: e22102, 2023. graf
Article in English | LILACS | ID: biblio-1439521

ABSTRACT

Abstract EphrinB2 plays a critical role in tumor growth. In this study, we studied the antitumor activity of imperatorin derivative IMP-1 in renal cell carcinoma (RCC) by regulating EphrinB2 pathway.. Results showed that IMP-1 inhibited the proliferation of 786-O cells in a dose- and time-dependent manner. More importantly, knockdown and transfection of EphrinB2 altered the inhibitory effect of IMP-1 on the activity of 786-O cells. IMP-1 arrested 786-O cell cycle at G0/G1 phase by decreasing the expression of cyclin D1 and cyclin E. Moreover, IMP-1 regulated Bcl-2 family proteins' expression, thus inducing apoptosis of 786-O cells. IMP-1 down-regulated the expression of EphrinB2, Syntenin1 and PICK1. Then, IMP-1 decreased the phosphorylation of Erk1/2 and AKT. In all, IMP-1 could regulate the EphrinB2 pathway in order to inhibit 786-O cell growth by arresting the cell cycle at G0/G1 phase and inducing cell apoptosis. Thus, IMP-1 may present as a potential strategy for RCC treatment.


Subject(s)
Carcinoma, Renal Cell/pathology , Neoplasms/classification , G1 Phase/genetics , Cyclin D1/adverse effects , Cyclin E/adverse effects
2.
Journal of Zhejiang University. Science. B ; (12): 467-475, 2019.
Article in English | WPRIM | ID: wpr-776716

ABSTRACT

The stem/progenitor cell has long been regarded as a central cell type in development, homeostasis, and regeneration, largely owing to its robust self-renewal and multilineage differentiation abilities. The balance between self-renewal and stem/progenitor cell differentiation requires the coordinated regulation of cell cycle progression and cell fate determination. Extensive studies have demonstrated that cell cycle states determine cell fates, because cells in different cell cycle states are characterized by distinct molecular features and functional outputs. Recent advances in high-resolution epigenome profiling, single-cell transcriptomics, and cell cycle reporter systems have provided novel insights into the cell cycle regulation of cell fate determination. Here, we review recent advances in cell cycle-dependent cell fate determination and functional heterogeneity, and the application of cell cycle manipulation for cell fate conversion. These findings will provide insight into our understanding of cell cycle regulation of cell fate determination in this field, and may facilitate its potential application in translational medicine.


Subject(s)
Animals , Humans , Cell Cycle , Cell Physiological Phenomena , Epigenomics , G1 Phase , G2 Phase , Translational Research, Biomedical
3.
Journal of Cancer Prevention ; : 10-17, 2018.
Article in English | WPRIM | ID: wpr-740094

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is one of the most important causes of death in men and thus new therapeutic approaches are needed. In this study, antiproliferative and anti-migration properties of a coumarin derivative esculetin were evaluated. METHODS: Human PCa cell lines PC3, DU145, and LNCaP were treated with various concentrations of esculetin for 24 to 72 hours, and cell viability was determined by the MTT test. Cell cycle and apoptosis were analyzed by using cell-based cytometer. Gene expression levels were assessed by reverse transcription and quantitative real-time PCR, cell migration was determined by the wound healing assay. The protein expression was measured by Western blotting. RESULTS: Esculetin inhibited cell proliferation in a dose- and time-dependent manner. Cell migration was inhibited by esculetin treatment. Administration of esculetin significantly reduced the cells survival, induced apoptosis and caused the G1 phase cell cycle arrest shown by image-based cytometer. The induced expression of cytochrome c, p53, p21 and p27, and down-regulated CDK2 and CDK4 may be the underlying molecular mechanisms of esculetin effect. Esculetin suppressed phosphorylation of Akt and enhanced protein expression of tumor-suppressor phosphatase and tensin homologue. CONCLUSIONS: Our findings showed that the coumarin derivative esculetin could be used in the management of PCa. However, further in vivo research is needed.


Subject(s)
Humans , Male , Apoptosis , Blotting, Western , Cause of Death , Cell Cycle Checkpoints , Cell Cycle , Cell Line , Cell Movement , Cell Proliferation , Cell Survival , Cytochromes c , G1 Phase , Gene Expression , Passive Cutaneous Anaphylaxis , Phosphorylation , Prostate , Prostatic Neoplasms , Real-Time Polymerase Chain Reaction , Reverse Transcription , Wound Healing
4.
Laboratory Animal Research ; : 264-269, 2018.
Article in English | WPRIM | ID: wpr-718841

ABSTRACT

Cell cycle dysfunction can cause severe diseases, including neurodegenerative disease and cancer. Mutations in cyclin-dependent kinase inhibitors controlling the G1 phase of the cell cycle are prevalent in various cancers. Mice lacking the tumor suppressors p16(Ink4a) (Cdkn2a, cyclin-dependent kinase inhibitor 2a), p19(Arf) (an alternative reading frame product of Cdkn2a,), and p27(Kip1) (Cdkn1b, cyclin-dependent kinase inhibitor 1b) result in malignant progression of epithelial cancers, sarcomas, and melanomas, respectively. Here, we generated knockout mouse models for each of these three cyclin-dependent kinase inhibitors using engineered nucleases. The p16(Ink4a) and p19(Arf) knockout mice were generated via transcription activator-like effector nucleases (TALENs), and p27(Kip1) knockout mice via clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease 9 (CRISPR/Cas9). These gene editing technologies were targeted to the first exon of each gene, to induce frameshifts producing premature termination codons. Unlike preexisting embryonic stem cell-based knockout mice, our mouse models are free from selectable markers or other external gene insertions, permitting more precise study of cell cycle-related diseases without confounding influences of foreign DNA.


Subject(s)
Animals , Mice , Cell Cycle , Codon, Nonsense , Cyclin-Dependent Kinase Inhibitor p16 , DNA , Exons , G1 Phase , Genome , Melanoma , Mice, Knockout , Mutagenesis, Insertional , Neurodegenerative Diseases , Phosphotransferases , Reading Frames , Sarcoma
5.
Biomolecules & Therapeutics ; : 322-327, 2018.
Article in English | WPRIM | ID: wpr-714733

ABSTRACT

A type of breast cancer with a defect in three molecular markers such as the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor is called triple-negative breast cancer (TNBC). Many patients with TNBC have a lower survival rate than patients with other types due to a poor prognosis. In this study, we confirmed the anti-cancer effect of a natural compound, Gomisin G, in TNBC cancer cells. Treatment with Gomisin G suppressed the viability of two TNBC cell lines, MDA-MB-231 and MDA-MB-468 but not non-TNBC cell lines such as MCF-7, T47D, and ZR75-1. To investigate the molecular mechanism of this activity, we examined the signal transduction pathways after treatment with Gomisin G in MDA-MB-231 cells. Gomisin G did not induce apoptosis but drastically inhibited AKT phosphorylation and reduced the amount of retinoblastoma tumor suppressor protein (Rb) and phosphorylated Rb. Gomisin G induced in a proteasome-dependent manner a decrease in Cyclin D1. Consequently, Gomisin G causes cell cycle arrest in the G1 phase. In contrast, there was no significant change in T47D cells except for a mild decrease in AKT phosphorylation. These results show that Gomisin G has an anti-cancer activity by suppressing proliferation rather than inducing apoptosis in TNBC cells. Our study suggests that Gomisin G could be used as a therapeutic agent in the treatment of TNBC patients.


Subject(s)
Humans , Apoptosis , Breast Neoplasms , Cell Cycle , Cell Cycle Checkpoints , Cell Line , Cell Proliferation , Cyclin D1 , Cyclins , Estrogens , G1 Phase , Phosphorylation , Prognosis , ErbB Receptors , Receptors, Progesterone , Retinoblastoma , Signal Transduction , Survival Rate , Triple Negative Breast Neoplasms
6.
Journal of Central South University(Medical Sciences) ; (12): 1009-1015, 2016.
Article in Chinese | WPRIM | ID: wpr-815140

ABSTRACT

To investigate effects of MARCH6 gene knockdown on MCF-7 cell proliferation and cell cycle.
 Methods: 293T cells were transfected with MARCH6 shRNA lentivirus. Fluorescence microscope was used to observe and verify the transfection efficiency. The initial effect of the MARCH6 gene knockdown in MCF-7 cells was observed via fluorescence microscope. Real-time PCR and Western blot were used to detect the expression of MARCH6. MTT and BrdU assay were used to examine cell proliferation, and staining flow cytometry was used to analyze cycle distribution of MCF-7 cells.
 Results: MARCH6 shRNA lentivirus was successfully transfected and about 80% of the cells expressed green fluorescent in comparison of the control. About 90% of the cells showed green fluorescence. The mRNA and protein in MCF-7 cells were transcription and expression of protein was significantly decreased after the transfection of MARCH6 shRNA lentivirus accompanied by a decrease in MCF-7 cell proliferation (P<0.01). Flow cytometry showed that the cell cycles were inhibited at the G1 phase and the proliferation index was significantly reduced.
 Conclusion: Knockdown of MARCH6 gene by RNA interference inhibits the proliferation of MCF-7 cells, suggesting that the expression of MARCH6 promotes proliferation of breast cancer cells through regulation of the cell cycle.


Subject(s)
Female , Humans , Adenocarcinoma , Genetics , Breast Neoplasms , Genetics , Cell Cycle , Cell Division , Cell Proliferation , Genetics , G1 Phase , Genetics , Gene Knockdown Techniques , Hyperplasia , Lentivirus , MCF-7 Cells , Physiology , Membrane Proteins , Physiology , RNA Interference , RNA, Messenger , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Transfection , Ubiquitin-Protein Ligases , Physiology
7.
Journal of Zhejiang University. Medical sciences ; (6): 364-370, 2016.
Article in Chinese | WPRIM | ID: wpr-239577

ABSTRACT

To investigate the effect of RAD18-siRNA on cell proliferation and chemotherapy sensitivity of esophageal squamous cell carcinoma (ESCC) ECA-109 cells.RAD18-siRNA was transfected into human ECA-109 cells by Lipofectamine 3000. Quantitative PCR and Western blot were performed to detect RAD18 and CyclinD1 expression; CCK-8 assay was used to determine cell proliferation and chemotherapy drug sensitivity; flow cytometry was used to determine cell cycle. Correlation between RAD18 and CyclinD1 mRNA expression was analyzed by Pearson's correlation.Compared with non-transfected cells, the expression of RAD18 in RAD18-siRNA group was significantly decreased (<0.05). The cell proliferation was inhibited (<0.05) and the cell number of G1 phase was increased, G2/M phase cells decreased (<0.05) in RAD18-siRNA group. After treatment with different concentrations of cisplatin or 5-FU, the survival rate of the two cell groups was reduced (all<0.05), and the IC50 of RAD18-siRNA group was significantly lower than that of non-transfected group (<0.05). The mRNA expression of RAD18 was positively correlated with CyclinD1 expression in ESCC tissues(=0.478,<0.01).Down-regulated expression of RAD18 can decrease the cell proliferation and increase chemo-sensitivity of ESCC cells, and CyclinD1 may participate in the process.


Subject(s)
Humans , Adjuvants, Pharmaceutic , Pharmacology , Carcinoma, Squamous Cell , Drug Therapy , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cisplatin , Pharmacology , Cyclin D1 , Genetics , DNA-Binding Proteins , Pharmacology , Down-Regulation , Genetics , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Methods , Drug Synergism , Esophageal Neoplasms , Drug Therapy , Fluorouracil , Pharmacology , G1 Phase , G2 Phase , Metaphase , RNA, Small Interfering , Pharmacology , Transfection , Ubiquitin-Protein Ligases , Pharmacology
8.
Natural Product Sciences ; : 293-298, 2016.
Article in English | WPRIM | ID: wpr-19613

ABSTRACT

Plant-derived triterpenoids commonly possesses biological properties such as anti-inflammatory, anti-microbial, anti-viral and anti-cancer. Luvunga scandens is one of the plant that produced triterpenoids. The aims of the study was to analyze cell cycle profile and to determine the expression of p53 unregulated modulator of apoptosis (PUMA), caspase-8 and caspase-9 genes at mRNA level in MCF-7 cell line treated with two triterpenoids, flindissol (1) and 3-oxotirucalla-7,24-dien-21-oic-acid (2) isolated from L. scandens. The compounds were tested for cell cycle analysis using flow cytometer and mRNA expression level using quantitative RT-PCR. The number of MCF-7 cells population which distributed in Sub G1 phase after treated with compound 1 and 2 were 7.7 and 9.3% respectively. The evaluation of the expression of genes showed that both compounds exhibited high level of expression of PUMA, caspase-8 and caspase-9 as normalized to β-actin via activation of those genes. In summary, the isolated compounds of L. scandens plant showed promising anticancer properties in MCF-7 cell lines.


Subject(s)
Apoptosis , Caspase 8 , Caspase 9 , Cell Cycle Checkpoints , Cell Cycle , Flow Cytometry , G1 Phase , Gene Expression , MCF-7 Cells , Plants , Puma , RNA, Messenger
9.
Annals of Dermatology ; : 600-606, 2016.
Article in English | WPRIM | ID: wpr-59028

ABSTRACT

BACKGROUND: Stress is a known cause of hair loss in many species. OBJECTIVE: In this study, we investigated the role of acute stress on hair growth using a rat model. METHODS: Rats were immobilized for 24 hours and blood samples, and skin biopsies were taken. The effect of stress-serum on the in vitro proliferation of rat and human dermal papilla cells (hDPCs), as well as serum cortisol and corticotropin-releasing hormone levels, were measured. Mast cell staining was performed on the biopsied tissue. In addition, Western blot and quantitative real time polymerase chain reaction were used to assess mast cell tryptase and cytokine expression, respectively in rat skin biopsies. RESULTS: Stress-serum treatment reduced significantly the number of viable hDPCs and arrested the cell cycle in the G1 phase, compared to serum from unrestrained rats (p<0.05, respectively). Moreover, restrained rats had significantly higher levels of cortisol in serum than unrestrained rats (p<0.01). Acute stress serum increased mast cell numbers and mast cell tryptase expression, as well as inducing interleukin (IL)-6 and IL-1β up-regulation. CONCLUSION: These results suggest that acute stress also has an inhibitory effect on hair growth via cortisol release in addition to substance P-mast cell pathway.


Subject(s)
Animals , Humans , Rats , Biopsy , Blotting, Western , Cell Cycle , Corticotropin-Releasing Hormone , G1 Phase , Hair , Hydrocortisone , In Vitro Techniques , Interleukins , Mast Cells , Models, Animal , Real-Time Polymerase Chain Reaction , Skin , Tryptases , Up-Regulation
10.
Dental press j. orthod. (Impr.) ; 20(1): 45-51, Jan-Feb/2015. tab, graf
Article in English | LILACS | ID: lil-741446

ABSTRACT

INTRODUCTION: The consensus about the relationship between TMD and orthodontic treatment has gone from a cause and effect association between TMD and orthodontic treatment to the idea that there is no reliable evidence supporting this statement. OBJECTIVE: To assess the beliefs, despite scientific evidence, of Brazilian orthodontists about the relationship between TMD and orthodontic treatment with regards to treatment, prevention and etiology of TMD. METHODS: A survey about the relationship between TMD and orthodontic treatment was prepared and sent to Brazilian orthodontists by e-mail and social networks. Answers were treated by means of descriptive statistics and strong associations between variables were assessed by qui-square test. RESULTS: The majority of orthodontists believe that orthodontic treatment not only is not the best treatment option for TMD, but also is not able to prevent TMD. Nevertheless, the majority of orthodontists believe that orthodontic treatment can cause TMD symptoms. CONCLUSION: This study suggests that orthodontists' beliefs about the relationship between orthodontic treatment and TMD are in accordance with scientific evidence only when referring to treatment and prevention of TMD. The majority of orthodontists believe that, despite scientific evidence, orthodontic treatment can cause TMD. .


INTRODUÇÃO: o consenso sobre a relação entre DTM e tratamento ortodôntico foi de uma associação de causa e efeito à ideia de que não há evidências confiáveis que suportem essa afirmação. OBJETIVO: avaliar as crenças, sem considerar as evidências, de ortodontistas brasileiros sobre a relação entre DTM e tratamento ortodôntico com relação ao tratamento, prevenção e etiologia da DTM. MÉTODOS: um questionário sobre a relação entre DTM e tratamento ortodôntico foi preparado e enviado a ortodontistas brasileiros por meio de e-mail e mídias sociais. As respostas foram analisadas por estatística descritiva, e fortes associações entre as variáveis foram verificadas pelo teste χ2. RESULTADOS: a maioria dos ortodontistas acredita que o tratamento ortodôntico não é o melhor tratamento para DTM. Além disso, acreditam que não é a melhor forma para sua prevenção. Também, a maioria dos ortodontistas acredita que o tratamento ortodôntico pode causar sintomas de DTM. CONCLUSÃO: este estudo sugere que as crenças dos ortodontistas sobre a relação entre tratamento ortodôntico e DTM estão de acordo com as evidências científicas apenas quando se trata do tratamento e da prevenção de DTM. A maioria dos ortodontistas acredita que, apesar das evidências científicas, o tratamento ortodôntico pode causar DTM. .


Subject(s)
Humans , Cell Cycle Proteins/metabolism , DNA Replication/genetics , Forkhead Transcription Factors/metabolism , G1 Phase/physiology , Gene Expression Regulation/genetics , Protein Serine-Threonine Kinases/metabolism , Replication Origin/genetics , Signal Transduction/genetics , Blotting, Western , Cell Fractionation , Cell Line , Cell Cycle Proteins/genetics , /metabolism , DNA Primers/genetics , Fluorescent Antibody Technique , Forkhead Transcription Factors/genetics , Immunoblotting , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/metabolism , Reverse Transcriptase Polymerase Chain Reaction , RNA Interference
11.
National Journal of Andrology ; (12): 792-796, 2015.
Article in Chinese | WPRIM | ID: wpr-276018

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the expression of long non-coding RNA-HOTAIR in prostate cancer cells and its effects on the growth and metastasis of the cells.</p><p><b>METHODS</b>Using quantitative reverse-transcription PCR (qRT-PCR), we determined the relative expression of HOTAIR in the normal human prostate epithelial cell line RWPE-I and prostate cancer cell lines PC-3 and DU145. We detected the effects of HOTAIR on the cell cycle and invasiveness of prostate cancer cells by RNA interference, flow cytometry, and Transwell mitration assay.</p><p><b>RESULTS</b>The expressions of HOTAIR in the PC3 and DU145 cells were increased 3.2 and 5.7 times, respectively, as compared with that in the normal RWPE-1 cells. After si-HOTAIR interference, the prostate cancer cells were arrested in the G2 phase and downregulated in the G1 phase. The invasive ability of the prostate cancer cells was evidently inhibited, with the inhibition rates of 32% and 44% of the PC3 cells and 43% and 34% of the DU145 cells for si-HOTAIR1 and si-HOTAIR2, respectively.</p><p><b>CONCLUSION</b>IncRNA HOTAIR is highly expressed in prostate cancer, which is associated with the growth and invasiveness of prostate cancer cells. HOTAIR is potentially a novel marker for the diagnosis and prognosis of prostate cancer.</p>


Subject(s)
Humans , Male , Cell Cycle , Cell Cycle Checkpoints , Cell Division , Cell Line, Tumor , Cell Proliferation , Down-Regulation , G1 Phase , G2 Phase , Neoplasm Invasiveness , Prognosis , Prostatic Neoplasms , Metabolism , Pathology , RNA Interference , RNA, Long Noncoding , Metabolism , RNA, Untranslated , Metabolism
12.
Nutrition Research and Practice ; : 111-116, 2015.
Article in English | WPRIM | ID: wpr-204524

ABSTRACT

BACKGROUND/OBJECTIVES: Inonotus obliquus (I. obliquus, Chaga mushroom) has long been used as a folk medicine to treat cancer. In the present study, we examined whether or not ethanol extract of I. obliquus (EEIO) inhibits cell cycle progression in HT-29 human colon cancer cells, in addition to its mechanism of action. MATERIALS/METHODS: To examine the effects of Inonotus obliquus on the cell cycle progression and the molecular mechanism in colon cancer cells, HT-29 human colon cancer cells were cultured in the presence of 2.5 - 10 microg/mL of EEIO, and analyzed the cell cycle arrest by flow cytometry and the cell cycle controlling protein expression by Western blotting. RESULTS: Treatment cells with 2.5 - 10 microg/mL of EEIO reduced viable HT-29 cell numbers and DNA synthesis, increased the percentage of cells in G1 phase, decreased protein expression of CDK2, CDK4, and cyclin D1, increased expression of p21, p27, and p53, and inhibited phosphorylation of Rb and E2F1 expression. Among I. obliquus fractions, fraction 2 (fractionated by dichloromethane from EEIO) showed the same effect as EEIO treatment on cell proliferation and cell cycle-related protein levels. CONCLUSIONS: These results demonstrate that fraction 2 is the major fraction that induces G1 arrest and inhibits cell proliferation, suggesting I. obliquus could be used as a natural anti-cancer ingredient in the food and/or pharmaceutical industry.


Subject(s)
Humans , Blotting, Western , Cell Cycle , Cell Cycle Checkpoints , Cell Proliferation , Colonic Neoplasms , Cyclin D1 , DNA , Drug Industry , Ethanol , Flow Cytometry , G1 Phase , G1 Phase Cell Cycle Checkpoints , HT29 Cells , Medicine, Traditional , Methylene Chloride , Phosphorylation
13.
Cancer Research and Treatment ; : 913-920, 2015.
Article in English | WPRIM | ID: wpr-90547

ABSTRACT

PURPOSE: Anomalies of Akt regulation, including overexpression in lung cancer, impart resistance to conventional chemotherapy and radiation, thereby implicating this kinase as a therapeutic intervention point. A novel scaffold of Akt inhibitors was developed through virtual screening of chemical databases available at Birla Institute of Technology and Science, Pilani, Hyderabad, based on docking studies using Maestro. A benzothienopyrimidine derivative (BIA-6) was identified as a potential lead molecule that inhibited Akt1 enzyme activity with an IC50 of 256 nM. MATERIALS AND METHODS: BIA-6 was tested for in vitro Akt1 inhibition using a fluorescence resonance energy transfer kit. Anti-proliferative activity was tested in NCI-H460, A549, NCI-H1975, and NCI-H2170 cell lines. The effect of the compound on p-Akt (S473) was estimated. RESULTS: BIA-6 allosterically caused a dose dependent reduction of growth of cell lines with a half maximal growth inhibition (GI50) range of 0.49 muM to 6.6 muM. Cell cycle analysis indicated that BIA-6 caused a G1 phase arrest at < 100 nM but led to apoptosis at higher doses. BIA-6 also exhibited synergism with standard chemotherapeutic agents. CONCLUSION: BIA-6 is a novel, allosteric Akt inhibitor with potent anti-cancer activity in lung cancer cell lines, that effectively blocks the phosphoinositide-3 kinase/Akt pathway with a high margin selectivity towards normal cells.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung , Cell Cycle , Cell Line , Databases, Chemical , Drug Synergism , Drug Therapy , Fluorescence Resonance Energy Transfer , G1 Phase , Inhibitory Concentration 50 , Lung Neoplasms , Lung , Mass Screening , Phosphotransferases
14.
Journal of Breast Cancer ; : 112-118, 2015.
Article in English | WPRIM | ID: wpr-30129

ABSTRACT

PURPOSE: Isocryptotanshinone (ICTS) is a natural bioactive product that is isolated from the roots of the widely used medical herb Salvia miltiorrhiza. However, few reports exist on the mechanisms underlying the therapeutic effects of ICTS. Here, we report that ICTS has anticancer activity and describe the mechanism underlying this effect. METHODS: The antiproliferative effect of ICTS was determined using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and clonogenic assays. The effect of ICTS on the cell cycle was measured using flow cytometry. Apoptosis was determined by Hoechst 33342 staining, DNA fragmentation assays, and Western blotting for apoptotic proteins. Finally, the effect of ICTS on mitogen-activated protein kinases (MAPKs) was determined by Western blotting. RESULTS: ICTS significantly inhibited proliferation of MCF-7 and MDA-MB-231 human breast cancer cells, HepG2 human liver cancer cells, and A549 human lung cancer cells in vitro. Among the tested cell lines, MCF-7 cells showed the highest sensitivity to ICTS. ICTS significantly inhibited colony formation by MCF-7 cells. Furthermore, exposure of MCF-7 cells to ICTS induced cell cycle arrest at the G1 phase and decreased mitochondrial membrane potential. Hoechst 33342 staining and Western blot analysis for apoptotic proteins suggested that ICTS induced apoptosis in MCF-7 cells. In addition, ICTS activated MAPK signaling in MCF-7 cells by inducing time- and concentration-dependent phosphorylation of JNK, ERK, and p38 MAPK. CONCLUSION: Our results suggest that ICTS inhibited MCF-7 cell proliferation by inducing apoptosis and activating MAPK signaling pathways.


Subject(s)
Humans , Apoptosis , Blotting, Western , Breast Neoplasms , Cell Cycle , Cell Cycle Checkpoints , Cell Line , DNA Fragmentation , Flow Cytometry , G1 Phase , Hep G2 Cells , Liver Neoplasms , Lung Neoplasms , MCF-7 Cells , Membrane Potential, Mitochondrial , Mitogen-Activated Protein Kinases , p38 Mitogen-Activated Protein Kinases , Phosphorylation , Salvia miltiorrhiza
15.
Indian J Exp Biol ; 2014 Apr; 52(4): 295-304
Article in English | IMSEAR | ID: sea-150359

ABSTRACT

Natural autophagy and autophagic cell death is being studied in the model system, D. discoideum, which has well known genetic and experimental advantages over the other known systems. There is no apoptotic machinery present in this organism which could interfere with the non-apoptotic cell death. The target of rapamycin (TOR) pathway is a major nutrient-sensing pathway which when inhibited by the drug rapamycin induces autophagy. Rapamycin was originally discovered as an anti-fungal agent but its use was abandoned when it was discovered to have potent immunosuppressive and anti-proliferative properties. It is a known drug used today for various cancer treatments and also for increasing longevity in many model organisms. It has a wide usage but its effects on other pathways or molecules are not known. This model system was used to study the action of rapamycin on autophagy induction. Using the GFP-Atg8, an autophagosome marker, it was shown that rapamycin treatment can induce autophagy by an accumulation of reactive oxygen species and intracellular free calcium. Rapamycin suppresses proliferation by induction of cell cycle arrest in the G1 phase. Taken together, the results suggest that the core machinery for autophagy is conserved in D. discoideum and it can serve as a good model system to delineate the action of rapamycin induced autophagy.


Subject(s)
Antioxidants/metabolism , Autophagy/drug effects , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Dictyostelium/drug effects , Dictyostelium/physiology , G1 Phase/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sirolimus/pharmacology
16.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 521-528, 2014.
Article in English | WPRIM | ID: wpr-351046

ABSTRACT

Ac-Phe-Lys-PABC-DOX (PDOX) is a smart doxorubicin (DOX) prodrug designed to decrease toxicities while maintaining the potent anticancer effects of DOX. This study was aimed at elucidating the effectiveness and toxicities of DOX and PDOX in patient-derived MCF-7 breast cancer cells in vitro. The MCF-7 cells were exposed to both PDOX and DOX, and cytotoxicities, cell cycle and P53/P21 signaling alterations were studied. Abundant cathepsin B was found in the MCF-7 cells, and treatment with PDOX and DOX triggered dose- and time-dependent cytotoxicity and resulted in a significant reduction in cell viability. The IC50 of PDOX and DOX was 3.91 and 0.94 μmol/L, respectively. Both PDOX and DOX caused an up-regulation of the P53/P21-related signal pathway, and PDOX significantly increased expression of P53 and caspase 3, and arrested the cell cycle at the G1/G2 phase. As compared with DOX, PDOX reduced toxicities, and it may have different action mechanisms on breast cancer cells.


Subject(s)
Female , Humans , Antibiotics, Antineoplastic , Pharmacology , Breast Neoplasms , Drug Therapy , Metabolism , Pathology , Caspase 3 , Metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21 , Doxorubicin , Pharmacology , Drug Screening Assays, Antitumor , Methods , G1 Phase , G2 Phase , Gene Expression Regulation, Neoplastic , Oligopeptides , Pharmacology , Signal Transduction , Tumor Suppressor Protein p53
17.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 535-541, 2014.
Article in English | WPRIM | ID: wpr-351044

ABSTRACT

The effects of over-expression of testis-specific expressed gene 1 (TSEG-1) on the viability and apoptosis of cultured spermatogonial GC-1spg cells were investigated, and the immortal spermatogonial cell line GC-1spg (CRL-2053™) was obtained as the cell model in order to explore the function of TSEG-1. We transfected the eukaryotic vector of TSEG-1, named as pEGFP-TSEG-1 into cultured spermatogonial GC-1spg cells. Over-expression of TSEG-1 inhibited the proliferation of GC-1spg cells, and arrested cell cycle slightly at G0/G1 phase. Transfection of TSEG-1 attenuated the transcript levels of Ki-67, PCNA and cyclin D1. In addition, over-expression of TSEG-1 induced early and late apoptosis, and reduced the mitochondrial membrane potential of GC-1spg cells. Moreover, transfection of TSEG-1 significantly enhanced the ratio of Bax/Bcl-2 and transcript levels of caspase 9, and decreased the expression of Fas and caspase 8 in GC-1spg cells. These results indicated over-expression of TSEG-1 suppresses the proliferation and induces the apoptosis of GC-1spg cells, which establishes a basis for further study on the function of TSEG-1.


Subject(s)
Animals , Male , Mice , Caspase 8 , Genetics , Cell Line , Cyclin D1 , Genetics , G1 Phase , Physiology , Histones , Genetics , Metabolism , Ki-67 Antigen , Genetics , Proliferating Cell Nuclear Antigen , Genetics , Resting Phase, Cell Cycle , Physiology , Spermatogonia , Cell Biology , Metabolism , bcl-2-Associated X Protein , Genetics
18.
Asian Pacific Journal of Tropical Medicine ; (12): 373-377, 2014.
Article in English | WPRIM | ID: wpr-819668

ABSTRACT

OBJECTIVE@#To investigate the effect of stromal interaction molecule 1(STIM1) knockdown on the proliferation and migration of endothelial progenitor cells (EPCs) after vascular injury and its mechanism.@*METHODS@#The rat bone marrow derived EPCs were divided into three groups: adenovirus negative control (group NSC), rat STIM1 adenovirus vector transfection group (group si/rSTIM1) and rat &human recombinant STIM1 adenovirus transfection group (group si/rSTIM1+hSTIM1). The STIM1 expressions in each group were detected by reverse transcription PCR after transfection; the cell proliferation was tested by [(3)H] thymidine incorporation assay ((3)H-TdR); Cell cycle was analyzed by flow cytometry; the cells' migration activity was detected by Boyden assay; Calcium ion concentration was detected by using laser confocal method.@*RESULTS@#48 h later after transfection, the expression level of STIM1 in si/rSTIM1 cells was significantly lower than that in NSC group (0.21 ± 0.12 vs 1.01 ± 0.01, P<0.05); EPCs that stayed in G1 phase in si/rSTIM1 group [(93.31 ± 0.24)%] were significantly more than that in NSC group [(78.03 ± 0.34)%, P<0.05]; EPCs' migration activity in si/rSTIM1 group (10.03±0.33) was significantly lower than that in NSC group: (32.11 ± 0.54, P<0.05); EPCs calcium ion concentration changes in EPCs in si/rSTIM1 group (38.03 ± 0.13) was significantly lower than that in NSC group (98.11 ± 0.34, P<0.05). While there was no significant difference between si/rSTIM1+hSTIM1 group and NSC group on the four indexes above.@*CONCLUSIONS@#Silence of STIM1 attenuates EPCs proliferation and migration after vascular injury, by mediating the calcium ion concentration in EPCs.


Subject(s)
Animals , Humans , Rats , Calcium , Metabolism , Cell Movement , Genetics , Cell Proliferation , Genetics , Endothelial Progenitor Cells , Cell Biology , Metabolism , Physiology , G1 Phase , Genetics , Gene Silencing , Membrane Glycoproteins , Genetics , Metabolism , Membrane Proteins , Genetics , Metabolism , Neoplasm Proteins , Genetics , Metabolism , Stromal Interaction Molecule 1 , Transfection , Vascular System Injuries , Metabolism
19.
Chinese Journal of Integrated Traditional and Western Medicine ; (12): 1369-1373, 2014.
Article in Chinese | WPRIM | ID: wpr-313020

ABSTRACT

<p><b>OBJECTIVE</b>To explore anti-cancer effect and mechanism of green tea extract (GTE) in three human oral squamous carcinoma cell lines (CAL-27, SCC-25 and KB).</p><p><b>METHODS</b>The cell lines were in vitro cultured and its growth inhibition was detected by MTT. After screening most sensitive cell line, effect of GTE on CAL-27 cell cycle was analyzed by flow cytometry. The protein expression of GTE on CAL-27 cell strain was determined by protein chip technique. The protein expression of CDK4, CDK6, and p-PDK1 was verified by using Western blot.</p><p><b>RESULTS</b>Compared with the control group, the inhibition rate on CAL-27 increased significantly after treated by 50, 100, 200, and 400 μg/mL GTE; the inhibition rate on KB increased after treated by 100, 200, and 400 μg/mL GTE; the inhibition rate on SCC-25 increased after treated by 25, 50, 100, 200, and 400 μg/mL GTE, all with statistical difference and in dose dependant manner (P < 0.01). Flow cytometric analysis showed that, when compared with the control group, 50 μg/mL GTE arrested CAL-27 cells in the G2/M phase (P < 0.05), and 100 μg/mL GTE arrested CAL-27 cells in the G2/M phase with concurrent decreased cells in the G0/G1 phase (P < 0.01). Totally 107 proteins were analyzed by protein chip technique. After treated by GTE, a total of 13 proteins significantly changed in CAL-27 cell line. Western blot showed that 25, 50, and 100 μg/mL GTE inhibited the expression of phopho-phosphoinositide-dependent protein kinase 1 (p-PDK1), cyclin-dependent kinase 4 (CDK4), and CDK6 of CAL-27 cell line with statistical difference (P < 0.05). The higher the drug concentration, the higher the inhibition rate (P < 0.05).</p><p><b>CONCLUSIONS</b>GTE could inhibit the proliferation of different human oral squamous carcinoma cell lines. CAL-27 is a sensitive cell line. GTE significantly affected EGFR and Notch signal network, and influenced changes of cell cycle related protein expression levels through the aforesaid channels, resulting in cell cycle arrest in S and G2/M phases.</p>


Subject(s)
Humans , Antineoplastic Agents, Phytogenic , Pharmacology , Therapeutic Uses , Antioxidants , Carcinoma, Squamous Cell , Cell Cycle , Cell Line, Tumor , Cyclin-Dependent Kinase 4 , Drugs, Chinese Herbal , Pharmacology , Therapeutic Uses , G1 Phase , Mouth Neoplasms , Tea
20.
Biol. Res ; 47: 1-12, 2014. ilus, graf, tab
Article in English | LILACS | ID: biblio-950760

ABSTRACT

BACKGROUND: Interactions between genes and their products give rise to complex circuits known as gene regulatory networks (GRN) that enable cells to process information and respond to external stimuli. Several important processes for life, depend of an accurate and context-specific regulation of gene expression, such as the cell cycle, which can be analyzed through its GRN, where deregulation can lead to cancer in animals or a directed regulation could be applied for biotechnological processes using yeast. An approach to study the robustness of GRN is through the neutral space. In this paper, we explore the neutral space of a Schizosaccharomyces pombe (fission yeast) cell cycle network through an evolution strategy to generate a neutral graph, composed of Boolean regulatory networks that share the same state sequences of the fission yeast cell cycle. RESULTS: Through simulations it was found that in the generated neutral graph, the functional networks that are not in the wildtype connected component have in general a Hamming distance more than 3 with the wildtype, and more than 10 between the other disconnected functional networks. Significant differences were found between the functional networks in the connected component of the wildtype network and the rest of the network, not only at a topological level, but also at the state space level, where significant differences in the distribution of the basin of attraction for the G1 fixed point was found for deterministic updating schemes. CONCLUSIONS: In general, functional networks in the wildtype network connected component, can mutate up to no more than 3 times, then they reach a point of no return where the networks leave the connected component of the wildtype. The proposed method to construct a neutral graph is general and can be used to explore the neutral space of other biologically interesting networks, and also formulate new biological hypotheses studying the functional networks in the wildtype network connected component.


Subject(s)
Schizosaccharomyces/physiology , Cell Cycle/physiology , Cyclin-Dependent Kinases/metabolism , Gene Regulatory Networks/physiology , Models, Biological , Schizosaccharomyces/genetics , Computer Graphics , Computer Simulation , G1 Phase/physiology , Neural Networks, Computer , Cell Cycle Proteins/metabolism , Computational Biology
SELECTION OF CITATIONS
SEARCH DETAIL