Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
J Clin Invest ; 130(4): 2097-2110, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31961825

ABSTRACT

Despite the effective clinical use of steroids for the treatment of Diamond Blackfan anemia (DBA), the mechanisms through which glucocorticoids regulate human erythropoiesis remain poorly understood. We report that the sensitivity of erythroid differentiation to dexamethasone is dependent on the developmental origin of human CD34+ progenitor cells, specifically increasing the expansion of CD34+ progenitors from peripheral blood (PB) but not cord blood (CB). Dexamethasone treatment of erythroid-differentiated PB, but not CB, CD34+ progenitors resulted in the expansion of a newly defined CD34+CD36+CD71hiCD105med immature colony-forming unit-erythroid (CFU-E) population. Furthermore, proteomics analyses revealed the induction of distinct proteins in dexamethasone-treated PB and CB erythroid progenitors. Dexamethasone treatment of PB progenitors resulted in the specific upregulation of p57Kip2, a Cip/Kip cyclin-dependent kinase inhibitor, and we identified this induction as critical; shRNA-mediated downregulation of p57Kip2, but not the related p27Kip1, significantly attenuated the impact of dexamethasone on erythroid differentiation and inhibited the expansion of the immature CFU-E subset. Notably, in the context of DBA, we found that steroid resistance was associated with dysregulated p57Kip2 expression. Altogether, these data identify a unique glucocorticoid-responsive human erythroid progenitor and provide new insights into glucocorticoid-based therapeutic strategies for the treatment of patients with DBA.


Subject(s)
Anemia, Diamond-Blackfan/metabolism , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Dexamethasone/pharmacology , Drug Resistance/drug effects , Erythroid Precursor Cells/metabolism , Up-Regulation/drug effects , Adult , Anemia, Diamond-Blackfan/drug therapy , Anemia, Diamond-Blackfan/pathology , Antigens, CD/metabolism , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Erythroid Precursor Cells/pathology , Female , Humans , Male
2.
J BUON ; 24(1): 301-309, 2019.
Article in English | MEDLINE | ID: mdl-30941985

ABSTRACT

PURPOSE: There is a need for identifying molecular prognostic biomarkers to better predict clinical outcomes in patients with renal cell carcinoma (RCC). This study investigated the pattern of cyclin D1 and p57 expression in RCC patients and evaluated their relation with clinicopathological characteristics and overall survival (OS). METHODS: Immunohistochemistry was applied to paraffin-embedded tissue sections of 74 RCC patients. Two cut-off groups were defined by the fraction of positive cells as follows: ≤10% and >10% positive cells for cyclin D1, and ≤5% and >5% positive cells for p57. RESULTS: Cyclin D1 expression in >10% of positive cells was observed mostly in the clear cell RCC, while p57 expression in ≤5% of positive cells was found in 86% of chromophobe RCC specimens. The higher expression of cyclin D1 and lower expression of p57 were more frequent in grade I-II tumors. OS was associated with unfavorable clinicopathological characteristics. However, cyclin D1/p57 expression did not influence the survival rates. CONCLUSION: Although cyclin D1 and p57 expression did not affect survival rates in RCC patients, proper validation and establishment of the qualitative cut-off point are needed for these tumors.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cyclin D1/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Kidney Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/biosynthesis , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Middle Aged , Paraffin Embedding , Retrospective Studies , Survival Rate
3.
Biomed Pharmacother ; 110: 646-655, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30544064

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common digestive system malignant tumors. Due to the resistance to radiotherapy, the prognosis in patients with HCC is poor. Based on previous studies and online tools prediction, we hypothesized that miR-92b, which was reported to promote HCC cell proliferation, might bind to p57kip2, a well-known tumor suppressor, to modulate the radioresistance of HCC to ionizing radiation (IR) -based radiotherapy. In the present study, a higher miR-92b expression in HCC tissues and cell lines was observed; a high miR-92b expression was correlated with poorer prognosis in patients with HCC. The overexpression of miR-92b enhanced the radioresistance of HCC to IR treatment by promoting cancer cell proliferation, attenuating cell apoptosis and remove IR-induced cell cycle at G2/M phase. Through directly binding to the 3'-UTR of p57kip2, miR-92b negatively regulated the protein levels of p57kip2; miR-92b inhibition enhanced the cell effect of IR on HCC cells, which could be attenuated by the p57kip2 knockdown, in other words, miR-92b modulated the radioresistance of HCC to IR-based radiotherapy through p57kip2. Taken together, miR-92b inhibits p57kip2 expression in HCC tissues and cell lines, thus enhancing the radioresistance of HCC to IR-based radiotherapy; targeting miR-92b to rescue p57kip2 expression in HCC might help sensitive HCC cells to IR-based radiotherapy.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/radiotherapy , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Liver Neoplasms/metabolism , Liver Neoplasms/radiotherapy , MicroRNAs/biosynthesis , Adult , Female , HEK293 Cells , Hep G2 Cells , Humans , Male , Middle Aged , Radiation, Ionizing
4.
J Clin Invest ; 129(1): 209-214, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30352048

ABSTRACT

The loss of insulin-secreting ß cells is characteristic among type I and type II diabetes. Stimulating proliferation to expand sources of ß cells for transplantation remains a challenge because adult ß cells do not proliferate readily. The cell cycle inhibitor p57 has been shown to control cell division in human ß cells. Expression of p57 is regulated by the DNA methylation status of the imprinting control region 2 (ICR2), which is commonly hypomethylated in Beckwith-Wiedemann syndrome patients who exhibit massive ß cell proliferation. We hypothesized that targeted demethylation of the ICR2 using a transcription activator-like effector protein fused to the catalytic domain of TET1 (ICR2-TET1) would repress p57 expression and promote cell proliferation. We report here that overexpression of ICR2-TET1 in human fibroblasts reduces p57 expression levels and increases proliferation. Furthermore, human islets overexpressing ICR2-TET1 exhibit repression of p57 with concomitant upregulation of Ki-67 while maintaining glucose-sensing functionality. When transplanted into diabetic, immunodeficient mice, the epigenetically edited islets show increased ß cell replication compared with control islets. These findings demonstrate that epigenetic editing is a promising tool for inducing ß cell proliferation, which may one day alleviate the scarcity of transplantable ß cells for the treatment of diabetes.


Subject(s)
Beckwith-Wiedemann Syndrome/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , DNA Demethylation , Genetic Loci , Insulin-Secreting Cells/metabolism , Up-Regulation , Beckwith-Wiedemann Syndrome/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Insulin-Secreting Cells/pathology , Ki-67 Antigen/biosynthesis
5.
Int J Cardiol ; 274: 319-325, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30077532

ABSTRACT

BACKGROUND: A significant proportion of patients develop left ventricular (LV) remodeling leading to heart failure after acute myocardial infarction (AMI). Being able to identify these patients would represent a step forward towards personalized medicine. The present study aimed to determine the ability of cyclin dependent kinase inhibitor 1C (CDKN1C) to risk stratify AMI patients, in a sex-specific manner. METHODS: CDKN1C expression was measured in blood samples obtained at admission in a test cohort of 447 AMI patients and a validation cohort of 294 patients. The study end-point was LV function assessed by the ejection fraction (EF) at follow-up. RESULTS: In the test cohort, CDKN1C was lower in patients with a reduced EF (<40%) compared to patients with preserved EF (≥50%). This observation was specific to women. CDKN1C was a significant univariate predictor of LV function in women only. In multivariable analysis including demographic and clinical parameters, CDKN1C predicted LV function in women (odds ratio [95% confidence interval] 0.44 [0.23-0.82]) but not in men (0.90 [0.70-1.16]). Addition of CDKN1C to a multivariable clinical model reduced the Akaike information criterion, attesting for an incremental predictive value, in women (p = 0.006) but not in men (p = 0.41). Bootstrap internal validation confirmed the added value of CDKN1C in women. The female-specific predictive value of CDKN1C was validated in the independent cohort. CONCLUSION: CDKN1C is a novel female-specific biomarker of LV function after AMI.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/genetics , Gene Expression Regulation , Myocardial Infarction/genetics , RNA/genetics , Stroke Volume/physiology , Ventricular Function, Left/physiology , Ventricular Remodeling/physiology , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Female , Follow-Up Studies , Humans , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/physiopathology , Polymerase Chain Reaction , Retrospective Studies , Sex Factors
6.
Mech Ageing Dev ; 177: 109-117, 2019 01.
Article in English | MEDLINE | ID: mdl-29928906

ABSTRACT

Replicative capacity of normal human cells decreases as telomeric sequence is lost at each division. It is believed that when a subset of chromosomes reach a critically short length, an ATM-initiated and p53-mediated transcriptional response inhibits cell growth, promoting cell senescence. In addition to loss of telomeric sequence, senescence can be induced by other stresses including ionizing radiation, oxidative damage, chemical crosslinkers like the chemotherapeutic agent cisplatin, as well as overactivation of oncogenes and tumor suppressors. Our group found that the expression of an isoform of the INhibitor of Growth 1 gene called ING1a increases approximately 10-fold as fibroblasts approach senescence and that forced expression rapidly induces a senescent phenotype in primary diploid fibroblasts, epithelial and endothelial cells that resembles replicative senescence by most physical and biochemical measures. ING1a induces these changes through strongly inhibiting endocytosis to block mitogen signaling by inducing the expression of intersectin 2, a key scaffolding protein of the endosomal pathway. This, in turn increases the expression of Rb and of p57Kip2 and p16INK4a that serve to maintain Rb is an active, growth inhibitory state. The ING1a model is currently being used to better understand the mechanism(s) responsible for activating Rb to enforce the senescent state.


Subject(s)
Cellular Senescence/physiology , Gene Expression Regulation/physiology , Inhibitor of Growth Protein 1/biosynthesis , Models, Biological , Adaptor Proteins, Vesicular Transport/biosynthesis , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/genetics , Endocytosis/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/pathology , Humans , Inhibitor of Growth Protein 1/genetics , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
7.
Twin Res Hum Genet ; 20(5): 389-394, 2017 10.
Article in English | MEDLINE | ID: mdl-28803575

ABSTRACT

CDKN1C and KCNQ1OT1 are imprinted genes that might be potential regulators of placental development. This study investigated placental expressions of CDKN1C and KCNQ1OT1 in monozygotic twins with and without selective intrauterine growth restriction (sIUGR). Seventeen sIUGR and fifteen normal monozygotic(MZ) twin pairs were examined. Placental mRNA expressions of CDKN1C and KCNQ1OT1 were detected by real-time fluorescent quantitative PCR. CDKN1C protein expression was detected by immunohistochemical assay and Western-blotting. In the sIUGR group, smaller fetuses had a smaller share of the placenta, and CDKN1C protein expression was significantly increased while KCNQ1OT1 mRNA expression was significantly decreased. The CDKN1C/KCNQ1OT1 mRNA ratio was lower in the larger fetus than in the smaller fetus (p < .05). In the control group, CDKN1C protein expression showed no difference between larger and smaller fetuses, while KCNQ1OT1 mRNA expression was significantly lower in the larger fetus, and the CDKN1C/KCNQ1OT1 mRNA ratio was higher in the larger fetus than in the smaller fetus (p < .05). Our findings showed that pathogenesis of sIUGR may be related to the co-effect of the up-regulated protein expression of CDKN1C and down-regulated mRNA expression of KCNQ1OT1 in the placenta.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Fetal Growth Retardation/metabolism , Gene Expression Regulation, Developmental , Placenta/metabolism , Twins, Monozygotic , Adult , Cyclin-Dependent Kinase Inhibitor p57/genetics , Female , Fetal Growth Retardation/genetics , Humans , Infant, Newborn , Male , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , Pregnancy
8.
Hum Mol Genet ; 25(24): 5407-5417, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27798108

ABSTRACT

Silver Russell Syndrome (SRS) syndrome is an imprinting disorder involving low birth weight with complex genetics and diagnostics. Some rare SRS patients carry maternally inherited microduplications spanning the imprinted genes CDKN1C, PHLDA2, SLC22A18 and KCNQ1, suggesting that overexpression of one of more of these genes contributes to the SRS phenotype. While this molecular alteration is very rare, feeding difficulties are a very common feature of this condition. Given that SRS children also have very low body mass index, understanding the underpinning biology of the eating disorder is important, as well as potential co-occurring behavioural alterations. Here, we report that a mouse model of this microduplication exhibits a number of behavioural deficits. The mice had a blunted perception of the palatability of a given foodstuff. This perception may underpin the fussiness with food. We additionally report hypoactivity, unrelated to anxiety or motoric function, and a deficit in the appropriate integration of incoming sensory information. Importantly, using a second genetic model, we were able to attribute all altered behaviours to elevated expression of a single gene, Cdkn1c. This is the first report linking elevated Cdkn1c to altered behaviour in mice. Importantly, the findings from our study may have relevance for SRS and highlight a potentially underreported aspect of this disorder.


Subject(s)
Behavior, Animal/physiology , Cyclin-Dependent Kinase Inhibitor p57/genetics , Hyperkinesis/genetics , Silver-Russell Syndrome/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , DNA Methylation/genetics , Disease Models, Animal , Gene Duplication , Genomic Imprinting , Humans , Hyperkinesis/physiopathology , Infant, Low Birth Weight , Mice , Mice, Transgenic , Silver-Russell Syndrome/physiopathology
9.
Mol Reprod Dev ; 83(5): 405-12, 2016 05.
Article in English | MEDLINE | ID: mdl-26988311

ABSTRACT

p57(kip2) (encoded by the Cdkn1c gene) is a member of the cip/kip family of cyclin-dependent kinase inhibitors that mediates cell cycle arrest in G1, allowing cells to differentiate. In the placenta, p57(kip2) is involved in endoreduplication, formation of trophoblast giant cells, trophoblast invasion, and expansion of placental cell layers. Here, we quantitatively and qualitatively define the cell- and region-specific expression of mouse placental p57(kip2) using laser-capture microdissection, in situ hybridization, and immunohistochemistry. Cdkn1c RNA was quantified by real-time quantitative PCR. Co-expression of Pl1 was used to identify trophoblast giant cells while Tbpba was used to identify spongiotrophoblast cells. Timed sacrifices were also carried out at embryonic days E7.5, E8.5, E9.5, and E12.5 to profile the expression in embryos and their placentas. At E8.5, intense expression of Cdkn1c was seen in invasive TGCs and the ectoplacental cone. Cdkn1c expression was more diffuse and more abundant in the labyrinth that in the junctional zone at both E9.5 and E12.5. Immunohistochemistry revealed robust p57(kip2) staining in trophoblast giant cells and in the ectoplacental cone at E8.5. p57(kip2) protein was seen in giant cells and throughout the labyrinth, although its abundance was reduced in the junctional zone at E9.5, and became more diffuse by E12.5. The early and intense expression in trophoblast giant cells is consistent with a role for p57(kip2) in the invasive phenotype of these cells. Mol. Reprod. Dev. 83: 405-412, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental/physiology , Trophoblasts/metabolism , Animals , Embryo, Mammalian/cytology , Female , Mice , Pregnancy , RNA, Messenger/biosynthesis , Trophoblasts/cytology
10.
Int J Hematol ; 102(5): 558-68, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26362858

ABSTRACT

The aim of the present study was to investigate the effects of rapamycin and its underlying mechanisms on acute lymphoblastic leukemia (ALL) cells. We found that the p14, p15, and p57 genes were not expressed in ALL cell lines (Molt-4 and Nalm-6) and adult ALL patients, whereas mTOR, 4E-BP1, and p70S6K were highly expressed. In Molt-4 and Nalm-6 cells exposed to rapamycin, cell viability decreased and the cell cycle was arrested at the G1/S phase. Rapamycin restored p14, p15, and p57 gene expression through demethylation of the promoters of these genes. As expected, rapamycin also increased p14 and p15 protein expression in both Molt-4 and Nalm-6 cells, as well as p57 protein expression in Nalm-6 cells. Rapamycin additionally decreased mTOR and p70S6K mRNA levels, as well as p70S6K and p-p70S6K protein levels. However, depletion of mTOR by siRNA did not alter the expression and promoter methylation states of p14, p15, and p57. These results indicate that the inhibitory effect of rapamycin may be due mainly to increased p14, p15, and p57 expression via promoter demethylation and decreased mTOR and p70S6K expression in ALL cell lines. These results suggest a potential role for rapamycin in the treatment of adult ALL.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Gene Expression Regulation, Leukemic/drug effects , Oncogene Proteins/biosynthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Adult , Cell Line, Tumor , Female , Genes, Tumor Suppressor , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
11.
PLoS Pathog ; 11(7): e1005031, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26153983

ABSTRACT

We show that two host-encoded primary RNAs (pri-miRs) and the corresponding microRNA (miR) clusters--widely reported to have cell transformation-associated activity--are regulated by EBNA3A and EBNA3C. Utilising a variety of EBV-transformed lymphoblastoid cell lines (LCLs) carrying knockout-, revertant- or conditional-EBV recombinants, it was possible to demonstrate unambiguously that EBNA3A and EBNA3C are both required for transactivation of the oncogenic miR-221/miR-222 cluster that is expressed at high levels in multiple human tumours--including lymphoma/leukemia. ChIP, ChIP-seq, and chromosome conformation capture analyses indicate that this activation results from direct targeting of both EBV proteins to chromatin at the miR-221/miR-222 genomic locus and activation via a long-range interaction between enhancer elements and the transcription start site of a long non-coding pri-miR located 28 kb upstream of the miR sequences. Reduced levels of miR-221/miR-222 produced by inactivation or deletion of EBNA3A or EBNA3C resulted in increased expression of the cyclin-dependent kinase inhibitor p57KIP2, a well-established target of miR-221/miR-222. MiR blocking experiments confirmed that miR-221/miR-222 target p57KIP2 expression in LCLs. In contrast, EBNA3A and EBNA3C are necessary to silence the tumour suppressor cluster miR-143/miR-145, but here ChIP-seq suggests that repression is probably indirect. This miR cluster is frequently down-regulated or deleted in human cancer, however, the targets in B cells are unknown. Together these data indicate that EBNA3A and EBNA3C contribute to B cell transformation by inhibiting multiple tumour suppressor proteins, not only by direct repression of protein-encoding genes, but also by the manipulation of host long non-coding pri-miRs and miRs.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , MicroRNAs/genetics , B-Lymphocytes/virology , Blotting, Western , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Epstein-Barr Virus Nuclear Antigens/metabolism , Flow Cytometry , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunoprecipitation , MicroRNAs/biosynthesis , Oncogenes , Real-Time Polymerase Chain Reaction
12.
Epigenetics ; 10(8): 692-7, 2015.
Article in English | MEDLINE | ID: mdl-26091021

ABSTRACT

Altered placental function as a consequence of aberrant imprinted gene expression may be one mechanism mediating the association between low birth weight and increased cardiometabolic disease risk. Imprinted gene expression is regulated by epigenetic mechanisms, particularly DNA methylation (5mC) at differentially methylated regions (DMRs). While 5-hydroxymethylcytosine (5hmC) is also present at DMRs, many techniques do not distinguish between 5mC and 5hmC. Using human placental samples, we show that the expression of the imprinted gene CDKN1C associates with birth weight. Using specific techniques to map 5mC and 5hmC at DMRs controlling the expression of CDKN1C and the imprinted gene IGF2, we show that 5mC enrichment at KvDMR and DMR0, and 5hmC enrichment within the H19 gene body, associate positively with birth weight. Importantly, the presence of 5hmC at imprinted DMRs may complicate the interpretation of DNA methylation studies in placenta; future studies should consider using techniques that distinguish between, and permit quantification of, both modifications.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , DNA Methylation/genetics , Epigenesis, Genetic , Genomic Imprinting , 5-Methylcytosine/metabolism , Birth Weight/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cytosine/analogs & derivatives , Cytosine/metabolism , Female , Gene Expression Regulation, Developmental , Humans , Insulin-Like Growth Factor II/genetics , Placenta/metabolism , Pregnancy
13.
Pathol Biol (Paris) ; 63(3): 122-5, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25979571

ABSTRACT

Angiotensin II (Ang II) plays a pivotal role in the pathogenesis of cardiac fibrosis and long noncoding RNAs (lncRNAs) have been found to be involved in human diseases. The roles of Ang II receptors (AT1 and AT2) have been controversial. Our previous studies revealed that Ang II dynamically down-regulated the expression of lncRNA-NR024118 and Cdkn1c in adult rat cardiac fibroblasts. However, up to now, whether the decrease of lncRNA-NR024118 and Cdkn1c induced by Ang II is mediated by AT1 or AT2 has never been illustrated. In order to reveal which subtype of Ang II receptors mediate the decrease of lncRNA-NR024118 and Cdkn1c induced by Ang II, we studied the expression of NR024118 and Cdkn1c with different receptor blockers in Ang II-treated adult rat cardiac fibroblasts. In this study, we found that losartan (AT1 blocker) nearly completely reversed the decrease of lncRNA-NR024118 and partly reversed the decrease of Cdkn1c induced by Ang II in adult rat cardiac fibroblasts, while AT2 blocker (PD123319) did not show effect to the level of lncRNA-NR024118 and Cdkn1c. In conclusion, our current studies showed that the decrease of lncRNA-NR024118 and Cdkn1c induced by Ang II is mediated by AT1 receptor-dependent not AT2 receptor-dependent, which is helpful to understand the molecular mechanism of Ang II receptors in adult rat cardiac fibroblasts.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II/pharmacology , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Fibroblasts/drug effects , Losartan/pharmacology , Myocardium/cytology , RNA, Long Noncoding/biosynthesis , Receptor, Angiotensin, Type 1/physiology , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p57/genetics , Down-Regulation/drug effects , Fibroblasts/metabolism , Imidazoles/pharmacology , Male , Pyridines/pharmacology , RNA, Long Noncoding/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 2/drug effects
14.
Nat Neurosci ; 18(5): 657-65, 2015 May.
Article in English | MEDLINE | ID: mdl-25821910

ABSTRACT

The mechanism by which adult neural stem cells (NSCs) are established during development is unclear. In this study, analysis of cell cycle progression by examining retention of a histone 2B (H2B)-GFP fusion protein revealed that, in a subset of mouse embryonic neural progenitor cells (NPCs), the cell cycle slows between embryonic day (E) 13.5 and E15.5 while other embryonic NPCs continue to divide rapidly. By allowing H2B-GFP expressed at E9.5 to become diluted in dividing cells until the young adult stage, we determined that a majority of NSCs in the young adult subependymal zone (SEZ) originated from these slowly dividing embryonic NPCs. The cyclin-dependent kinase inhibitor p57 is highly expressed in this embryonic subpopulation, and the deletion of p57 impairs the emergence of adult NSCs. Our results suggest that a substantial fraction of adult SEZ NSCs is derived from a slowly dividing subpopulation of embryonic NPCs and identify p57 as a key factor in generating this embryonic origin of adult SEZ NSCs.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/physiology , Nerve Tissue Proteins/physiology , Neural Stem Cells/cytology , Neurogenesis , Telencephalon/cytology , Animals , Cell Cycle , Cell Division , Cell Lineage , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/genetics , Genes, Reporter , Histones/biosynthesis , Histones/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Recombinant Fusion Proteins/metabolism , Telencephalon/embryology
15.
J Laryngol Otol ; 129(2): 168-73, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25695279

ABSTRACT

BACKGROUND: Genetic alteration of cyclin-dependent kinase inhibitors has been associated with carcinogenesis mechanisms in various organs. OBJECTIVE: This study aimed to evaluate the expression and mutational analysis of Cip/Kip family cyclin-dependent kinase inhibitors (p21CIP1/WAF1, p27KIP1 and p57KIP2) in early glottic cancer. METHODS: Expressions of Cip/Kip family and p53 were determined by quantitative reverse transcription polymerase chain reaction and densitometry. For the analysis of p21 inactivation, sequence alteration was assessed using single-strand conformational polymorphism polymerase chain reaction. Additionally, the inactivation mechanism of p27 and p57 were investigated using DNA methylation analysis. RESULTS: Reduced expression of p27 and p57 were detected in all samples, whereas the expression of p21 was incompletely down-regulated in 6 of 11 samples. Additionally, single-strand conformational polymorphism polymerase chain reaction analysis showed the p53 mutation at exon 6. Methylation of p27 and p57 was detected by DNA methylation assay. CONCLUSION: Our results suggest that the Cip/Kip family may have a role as a molecular mechanism of carcinogenesis in early glottic cancer.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , Glottis/pathology , Head and Neck Neoplasms/genetics , Laryngeal Neoplasms/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , DNA Mutational Analysis/methods , Glottis/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/pathology , Polymorphism, Single-Stranded Conformational , Reverse Transcriptase Polymerase Chain Reaction/methods , Squamous Cell Carcinoma of Head and Neck
16.
Oncogene ; 34(14): 1822-30, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-24837369

ABSTRACT

Aberrant de novo methylation of DNA is considered an important mediator of tumorigenesis. To investigate the role of de novo DNA methyltransferase 3a (Dnmt3a) in intestinal tumor development, we analyzed the expression of Dnmt3a in murine colon crypts, murine colon adenomas and human colorectal cancer using RNA fluorescence in situ hybridization (FISH), quantitative PCR and immunostaining. Following conditional deletion of Dnmt3a in the colon of APC((Min/+)) mice, we analyzed tumor numbers, genotype of macroadenomas and laser dissected microadenomas, global and regional DNA methylation and gene expression. Our results showed increased Dnmt3a expression in colon adenomas of APC((Min/+)) mice and human colorectal cancer samples when compared with control tissue. Interestingly, in tumor tissue, RNA FISH analysis showed highest Dnmt3a expression in Lgr5-positive stem/progenitor cells. Deletion of Dnmt3a in APC((Min/+)) mice reduced colon tumor numbers by ~40%. Remaining adenomas and microadenomas almost exclusively contained the non-recombined Dnmt3a allele; no tumors composed of the inactivated Dnmt3a allele were detected. DNA methylation was reduced at the Oct4, Nanog, Tff2 and Cdkn1c promoters and expression of the tumor-suppressor genes Tff2 and Cdkn1c was increased. In conclusion, our results show that Dnmt3a is predominantly expressed in the stem/progenitor cell compartment of tumors and that deletion of Dnmt3a inhibits the earliest stages of intestinal tumor development.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Neoplastic Stem Cells/pathology , Adenoma/pathology , Animals , Cell Transformation, Neoplastic/genetics , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/genetics , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methyltransferase 3A , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucins/biosynthesis , Mucins/genetics , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Peptides/genetics , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Trefoil Factor-2
17.
Growth Factors ; 32(2): 53-62, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24580070

ABSTRACT

Insulin-like growth factor-1 (IGF-1) is known to promote proliferation in many cell types including c-kit(pos) cardiac stem cells (CSCs). Downstream signaling pathways of IGF-1 induced CSC proliferation have not been investigated. An important downstream target of IGF-1/Akt-1 signaling is FoxO3a, a key negative regulator of cell-cycle progression. We studied the effect of IGF-1 on proliferation of c-kit(pos) murine CSCs and found that IGF-1-mediated cell proliferation is associated with FoxO3a phosphorylation and inactivation of its transcriptional activity. PI3 inhibitors LY294002 and Wortmannin abolished the effect of IGF-1 on FoxO3a phosphorylation indicating that FoxO3a phosphorylation is mediated by PI3/Akt-1 pathway. In cells with FoxO3a translocation to the cytoplasm, there is decreased expression of cell-cycle inhibitors such as p27(kip1) and p57(kip2) and increased expression of CyclinD1. Our study provides evidence that IGF-1 induced CSC proliferation could be the result of FoxO3a inactivation and its downstream effect on cell-cycle regulators.


Subject(s)
Forkhead Transcription Factors/metabolism , Insulin-Like Growth Factor I/pharmacology , Myocardium/cytology , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/metabolism , Active Transport, Cell Nucleus , Androstadienes/pharmacology , Animals , Cell Cycle , Cell Proliferation/drug effects , Cells, Cultured , Chromones/pharmacology , Cyclin D1/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Mice , Mice, Inbred BALB C , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/biosynthesis , Signal Transduction/drug effects , Stem Cells/cytology , Wortmannin
18.
Sci Rep ; 4: 4012, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24500246

ABSTRACT

The quiescent (G0) phase of the cell cycle is the reversible phase from which the cells exit from the cell cycle. Due to the difficulty of defining the G0 phase, quiescent cells have not been well characterized. In this study, a fusion protein consisting of mVenus and a defective mutant of CDK inhibitor, p27 (p27K(-)) was shown to be able to identify and isolate a population of quiescent cells and to effectively visualize the G0 to G1 transition. By comparing the expression profiles of the G0 and G1 cells defined by mVenus-p27K(-), we have identified molecular features of quiescent cells. Quiescence is also an important feature of many types of stem cells, and mVenus-p27K(-)-transgenic mice enabled the detection of the quiescent cells with muscle stem cell markers in muscle in vivo. The mVenus-p27K(-) probe could be useful in investigating stem cells as well as quiescent cells.


Subject(s)
Bacterial Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , G1 Phase/genetics , Luminescent Proteins/genetics , Recombinant Fusion Proteins/genetics , Resting Phase, Cell Cycle/genetics , 3T3 Cells , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Gene Expression , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Retinoblastoma Protein/biosynthesis , Selenium-Binding Proteins/biosynthesis , Selenium-Binding Proteins/genetics , Stem Cells/cytology , Tumor Suppressor Protein p53/biosynthesis
19.
Dev Biol ; 387(1): 49-63, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24412371

ABSTRACT

Geminin is a protein involved in both DNA replication and cell fate acquisition. Although it is essential for mammalian preimplantation development, its role remains unclear. In one study, ablation of the geminin gene (Gmnn) in mouse preimplantation embryos resulted in apoptosis, suggesting that geminin prevents DNA re-replication, whereas in another study it resulted in differentiation of blastomeres into trophoblast giant cells (TGCs), suggesting that geminin regulates trophoblast specification and differentiation. Other studies concluded that trophoblast differentiation into TGCs is regulated by fibroblast growth factor-4 (FGF4), and that geminin is required to maintain endocycles. Here we show that ablation of Gmnn in trophoblast stem cells (TSCs) proliferating in the presence of FGF4 closely mimics the events triggered by FGF4 deprivation: arrest of cell proliferation, formation of giant cells, excessive DNA replication in the absence of DNA damage and apoptosis, and changes in gene expression that include loss of Chk1 with up-regulation of p57 and p21. Moreover, FGF4 deprivation of TSCs reduces geminin to a basal level that is required for maintaining endocycles in TGCs. Thus, geminin acts both like a component of the FGF4 signal transduction pathway that governs trophoblast proliferation and differentiation, and geminin is required to maintain endocycles.


Subject(s)
Fibroblast Growth Factor 4/metabolism , Geminin/metabolism , Giant Cells/metabolism , Trophoblasts/metabolism , Animals , Apoptosis/genetics , Cell Differentiation , Cell Proliferation , Checkpoint Kinase 1 , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , DNA Damage/genetics , DNA Replication/genetics , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Fibroblast Growth Factor 4/genetics , Geminin/genetics , Gene Expression Regulation, Developmental , Giant Cells/cytology , Mice , Mice, Transgenic , Protein Kinases/deficiency , Protein Kinases/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Trophoblasts/cytology , Up-Regulation
20.
Mod Pathol ; 27(2): 238-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23887308

ABSTRACT

Immunohistochemical analysis of cyclin-dependent kinase inhibitor 1C (CDKN1C, p57, Kip2) expression and molecular genotyping accurately classify hydatidiform moles into complete and partial types and distinguish these from non-molar specimens. Characteristics of a prospective series of all potentially molar specimens encountered in a large gynecologic pathology practice are summarized. Initially, all specimens were subjected to both analyses; this was later modified to triage cases for genotyping based on p57 results: p57-negative cases diagnosed as complete hydatidiform moles without genotyping; all p57-positive cases genotyped. Of the 678 cases, 645 were definitively classified as complete hydatidiform mole (201), partial hydatidiform mole (158), non-molar (272), and androgenetic/biparental mosaic (14); 33 were unsatisfactory, complex, or problematic. Of the 201 complete hydatidiform moles, 104 were p57-negative androgenetic and an additional 95 were p57-negative (no genotyping), 1 was p57-positive (retained maternal chromosome 11) androgenetic, and 1 was p57-non-reactive androgenetic; 90 (85%) of the 106 genotyped complete hydatidiform moles were monospermic and 16 were dispermic. Of the 158 partial hydatidiform moles, 155 were diandric triploid, with 154 p57-positive, 1 p57-negative (loss of maternal chromosome 11), and 1 p57-non-reactive; 3 were triandric tetraploid, with 2 p57-positive and 1 p57-negative (loss of maternal chromosome 11). Of 155 diandric triploid partial hydatidiform moles, 153 (99%) were dispermic and 2 were monospermic. Of the 272 non-molar specimens, 259 were p57-positive biparental diploid, 5 were p57-positive digynic triploid, 2 were p57-negative biparental diploid (no morphological features of biparental hydatidiform mole), and 6 were p57-non-reactive biparental diploid. Of the 14 androgenetic/biparental mosaics with discordant p57 expression, 6 were uniformly mosaic and 8 had a p57-negative androgenetic molar component. p57 expression is highly correlated with genotyping, serves as a reliable marker for diagnosis of complete hydatidiform moles, and identifies androgenetic cell lines in mosaic conceptions. Cases with aberrant and discordant p57 expression can be correctly classified by genotyping.


Subject(s)
Biomarkers, Tumor/analysis , Cyclin-Dependent Kinase Inhibitor p57/genetics , Hydatidiform Mole/genetics , Uterine Neoplasms/genetics , Adult , Cyclin-Dependent Kinase Inhibitor p57/analysis , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Female , Genotype , Humans , Hydatidiform Mole/metabolism , Immunohistochemistry , Pregnancy , Uterine Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL