Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 18300, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110120

ABSTRACT

Gamma radiation is a commonly used adjuvant treatment for abdominally localized cancer. Since its therapeutic potential is limited due to gastrointestinal (GI) syndrome, elucidation of the regenerative response following radiation-induced gut injury is needed to develop a preventive treatment. Previously, we showed that Krüppel-like factor 4 (KLF4) activates certain quiescent intestinal stem cells (ISCs) marked by Bmi1-CreER to give rise to regenerating crypts following γ irradiation. In the current study, we showed that γ radiation-induced expression of p21Waf1/Cip1 in Bmi1-CreER cells is likely mitigated by MUSASHI-1 (MSI1) acting as a negative regulator of p21Waf1/Cip1 mRNA translation, which promotes exit of the Bmi1-CreER cells from a quiescent state. Additionally, Bmi1-specific Klf4 deletion resulted in decreased numbers of MSI1+ cells in regenerating crypts compared to those of control mice. We showed that KLF4 binds to the Msi1 promoter and activates its expression in vitro. Since MSI1 has been shown to be crucial for crypt regeneration, this finding elucidates a pro-proliferative role of KLF4 during the postirradiation regenerative response. Taken together, our data suggest that the interplay among p21Waf1/Cip1, MSI1 and KLF4 regulates Bmi1-CreER cell survival, exit from quiescence and regenerative potential upon γ radiation-induced injury.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gamma Rays/adverse effects , Intestinal Mucosa/radiation effects , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Radiation Injuries, Experimental/metabolism , Stem Cells/radiation effects , Animals , HEK293 Cells , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Kruppel-Like Factor 4 , Mice , Polymerase Chain Reaction , Stem Cells/metabolism
2.
Int J Med Mushrooms ; 19(8): 709-716, 2017.
Article in English | MEDLINE | ID: mdl-29199570

ABSTRACT

Numerous formulations derived from the shiitake medicinal mushroom, Lentinus edodes, demonstrate anticancer activities. We hypothesized that isolates from selenium (Se)-enriched mycelia of L. edodes would possess stronger cancer-preventive properties than current preparations. The aim of this study was to investigate whether the presence of Se-methyl-seleno-L-cysteine in mycelial extracts of L. edodes affects their cytotoxic activity (makes them stronger) or whether they are as effective as Se-containing polysaccharides. Extracts were prepared from Se-containing mycelia under various conditions and assayed for cytotoxic activity in cancer (PC3 and HeLa) and normal (HMEC-1) cell lines. The chemical composition of the extracts was examined; specifically, the amounts of potentially cytotoxic Se compounds (methylselenocysteine, selenomethionine, and Se-containing polysaccharides) were measured. The relationship between extract composition and biological activity was characterized. Mycelial cultures were cultivated in a 10-L bioreactor in medium enriched with sodium selenite. Mycelial extracts were prepared either at 100°C or at 4°C in acidic solution. Total Se content was determined using the atomic absorption spectrometry method, and methylselenocysteine and selenomethionine contents were measured using reverse-phase high-performance liquid chromatography. Protein, carbohydrate, and polyphenolic contents were determined with spectrophotometric methods, and Se-containing polysaccharides were measured with the use of precipitation. Anticancer activity of mycelial extracts was examined using the MTT cell viability assay. Extracts containing Se-methyl-seleno-L-cysteine or Se-polysaccharides prepared at 4°C and 100°C, respectively, display moderate, time-dependent, specific cytotoxic activity in HeLa and PC3 cell lines. The effect in HeLa cells is more pronounced in the extract prepared at 4°C than at 100°C. The effect is almost equal for the PC3 cell line. However, both extracts have no effect or only slightly stimulate normal (HMEC-1) cell viability. The selective cytotoxic activity of L. edodes extracts in cancer (PC3 and HeLa) cells is due to the presence of both Se-methyl-seleno-L-cysteine and selenated polysaccharides, perhaps in combination with other active ingredients.


Subject(s)
Antineoplastic Agents/isolation & purification , Selenocysteine/analogs & derivatives , Shiitake Mushrooms/chemistry , Antineoplastic Agents/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , HeLa Cells , Humans , Mycelium/chemistry , Polysaccharides/isolation & purification , Polysaccharides/pharmacology , Selenocysteine/isolation & purification , Selenocysteine/pharmacology
3.
Oncol Rep ; 33(5): 2143-50, 2015 May.
Article in English | MEDLINE | ID: mdl-25760094

ABSTRACT

Overexpression of the BH3-interacting domain death agonist (BID) protein sensitizes certain cancer cell lines to apoptosis induced by anticancer agents, particularly by those acting through death receptors (e.g. TRAIL). Previously, we showed that recombinant BID fused with TAT cell penetrating peptide (TAT-BID) allowed for controlled delivery of BID to different cancer cell lines and moderately sensitized some of them to TRAIL or slightly to camptothecin. In the present study, we showed that TAT-BID delivered to HeLa cells strongly sensitized them to doxorubicin, as identified by cell viability and apoptosis assays. Another cell line sensitized to doxorubicin was PC3, whereas A549 and LNCaP cells were sensitized moderately or not at all, respectively. Sensitization was more pronounced at 1 µM doxorubicin administered for 48 h than for lower doses and shorter treatments. TAT-BID and doxorubicin may thus be considered as a potential therapeutic combination for cervical carcinoma and advanced prostate cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/administration & dosage , Doxorubicin/pharmacology , Neoplasms/pathology , Blotting, Western , Cell Line, Tumor , Cell Survival , Drug Synergism , Fluorescent Antibody Technique , Gene Products, tat , Genetic Therapy/methods , Humans , Recombinant Proteins/administration & dosage
4.
BMC Cancer ; 14: 771, 2014 Oct 18.
Article in English | MEDLINE | ID: mdl-25326334

ABSTRACT

BACKGROUND: Low cellular level of BID is critical for viability of numerous cancer cells. Sensitization of cells to anticancer agents by BID overexpression from adenovirus or pcDNA vectors is a proposed strategy for cancer therapy; however it does not provide any stringent control of cellular level of BID. The aim of this work was to examine whether a fusion of BID with TAT cell penetrating peptide (TAT-BID) may be used for controlled sensitization of cancer cells to anticancer agents acting through death receptors (TRAIL) or DNA damage (camptothecin). Prostate cancer PC3 and LNCaP, non-small human lung cancer A549, and cervix carcinoma HeLa cells were used in the study. METHODS: Uptake of TAT-BID protein by cells was studied by quantitative Western blot analysis of cells extracts. Cells viability was monitored by MTT test. Apoptosis was detected by flow cytometry and cytochrome c release assay. RESULTS: TAT-BID was delivered to all cancer cells in amounts depending on time, dose and the cell line. Recombinant BID sensitized PC3 cells to TRAIL or, to lesser extent, to camptothecin. Out of remaining cells, TAT-BID sensitized A549, and only slightly HeLa cells to TRAIL. None of the latter cell lines were sensitized to camptothecin. In all cases the mutant not phosphorylable by CK2 (TAT-BIDT59AS76A) was similarly efficient in sensitization as the wild type TAT-BID. CONCLUSIONS: TAT-BID may be delivered to cancer cells in controlled manner and efficiently sensitizes PC3 and A549 cells to TRAIL. Therefore, it may be considered as a potential therapeutic agent that enhances the efficacy of TRAIL for the treatment of prostate and non-small human lung cancer.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , Peptide Fragments/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , tat Gene Products, Human Immunodeficiency Virus/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Humans , Mitochondria/drug effects , Mitochondria/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Time Factors
5.
BMC Vet Res ; 8: 170, 2012 Sep 23.
Article in English | MEDLINE | ID: mdl-22999388

ABSTRACT

BACKGROUND: Ghrelin is a natural ligand of the growth hormone secretagogue receptor (GHS-R). They are often co-expressed in multiple human tumors and related cancer cell lines what can indicate that the ghrelin/GHS-R axis may have an important role in tumor growth and progression. However, a role of ghrelin in canine tumors remains unknown. Thus, the aim of our study was two-fold: (1) to assess expression of ghrelin and its receptor in canine mammary cancer and (2) to examine the effect of ghrelin on carcinoma cells proliferation, apoptosis, migration and invasion. The expression of ghrelin and its receptor in canine mammary cancer tissues and cell lines (isolated from primary tumors and their metastases) was examined using Real-time qPCR and immunohistochemistry. For apoptosis analysis the Annexin V and propidium iodide dual staining was applied whereas cell proliferation was evaluated by MTT assay and BrdU incorporation test. The influence of ghrelin on cancer cells migration and invasion was assessed using Boyden chamber assays and wound healing assay. RESULTS: The highest expression of ghrelin was observed in metastatic cancers whereas the lowest expression of ghrelin receptor was detected in tumors of the 3rd grade of malignancy. Higher expression of ghrelin and its receptor was detected in cancer cell lines isolated from metastases than in cell lines isolated from primary tumors. In vitro experiments demonstrated that exposure to low doses of ghrelin stimulates cellular proliferation, inhibits apoptosis and promotes motility and invasion of canine mammary cancer cells. Growth hormone secretagogue receptor inhibitor ([D-Lys3]-GHRP6) as well as RNA interference enhances early apoptosis. CONCLUSION: The presence of ghrelin and GHS-R in all of the examined canine mammary tumors may indicate their biological role in cancer growth and development. Our experiments conducted in vitro confirmed that ghrelin promotes cancer development and metastasis.


Subject(s)
Apoptosis/physiology , Carcinoma/metabolism , Cell Movement/physiology , Dog Diseases/metabolism , Ghrelin/metabolism , Mammary Neoplasms, Animal/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Dogs , Female , Gene Expression Regulation, Neoplastic/physiology , Ghrelin/genetics , RNA Interference , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism
6.
Oncol Rep ; 27(1): 281-5, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21993828

ABSTRACT

Inhibitors of CK2 kinase inhibit cell proliferation and induce apoptosis in numerous cancer cell lines. Due to these properties, they are considered potentially useful in anticancer therapy. In this study, we show that the exact effect of the specific CK2 inhibitor TBB on PC-3 human prostate cancer cell viability depends on the time schedule of administration: it was not observed when the treatment was directly followed by the viability assay but it appeared when the treatment and the assay were separated by a 24-h incubation without the inhibitor. Such a pattern was maintained when the TBB treatment was combined with either camptothecin or TRAIL. The time schedule-dependence of cell viability was not reflected by a similar dependence of induction of apoptosis. Despite this, the schedule in which a treatment with the CK2 inhibitor precedes that with an anticancer drug seems to be a good choice for a potential therapy against androgen-refractory prostate cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell Survival/drug effects , Prostatic Neoplasms/metabolism , Triazoles/administration & dosage , Apoptosis/drug effects , Camptothecin/administration & dosage , Cell Line, Tumor , Checkpoint Kinase 2 , Enzyme Inhibitors/administration & dosage , Humans , Male , Protein Serine-Threonine Kinases/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...