Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 338
Filter
Add more filters

Publication year range
1.
Pharmacology ; 106(3-4): 137-142, 2021.
Article in English | MEDLINE | ID: mdl-32980839

ABSTRACT

The dental follicle is part of the tooth germ, and isolated stem cells from this tissue (dental follicle cells; DFCs) are considered, for example, for regenerative medicine and immunotherapies. However somatic stem cells can also improve pharmaceutical research. Cell proliferation is limited by the induction of senescence, which, while reducing the therapeutic potential of DFCs for cell therapy, can also be used to study aging processes at the cellular level that can be used to test anti-aging pharmaceuticals. Unfortunately, very little is known about cellular senescence in DFCs. This review presents current knowledge about cellular senescence in DFCs.


Subject(s)
Cellular Senescence/physiology , Dental Sac/physiology , Cell Differentiation/physiology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Humans , Osteogenesis/physiology , Telomere/metabolism , Wnt-5a Protein/metabolism
2.
Am J Respir Cell Mol Biol ; 62(3): 319-330, 2020 03.
Article in English | MEDLINE | ID: mdl-31513752

ABSTRACT

Senescence of alveolar type II (ATII) cells, progenitors of the alveolar epithelium, is a pathological feature and contributes importantly to the pathogenesis of idiopathic pulmonary fibrosis. Despite recognition of the importance of ATII cell senescence in idiopathic pulmonary fibrosis pathogenesis, how ATII cell senescence is regulated and how senescent ATII cells contribute to lung fibrogenesis remain unclear. In this study, we show that TGF-ß1 (transforming growth factor-ß1), a most ubiquitous and potent profibrotic cytokine, induces plasminogen activator inhibitor-1 (PAI-1), a cell senescence and fibrosis mediator, and p16 as well as senescence, but not apoptosis, in primary mouse ATII cells. We also found that senescent ATII cells secrete various cytokines and chemokines, including IL-4 and IL-13, which stimulate the expression of genes associated with a profibrotic phenotype in alveolar macrophages. Similar responses were also observed in TGF-ß1-treated rat ATII (L2) and rat macrophage NR8383 cells. Deletion of PAI-1 or inhibition of PAI-1 activity with a small molecule PAI-1 inhibitor, however, blocks TGF-ß1-induced senescence as well as a senescence-associated secretory phenotype in ATII and L2 cells and, consequently, the stimulatory effects of the conditioned medium from senescent ATII/L2 cells on macrophages. Moreover, we show that silencing p16 ameliorates PAI-1 protein-induced ATII cell senescence and secretion of profibrotic mediators. Our data suggest that PAI-1 mediates TGF-ß1-induced ATII cell senescence and secretion of profibrotic mediators through inducing p16, and they also suggest that senescent ATII cells contribute to lung fibrogenesis in part by activating alveolar macrophages through secreting profibrotic and proinflammatory mediators.


Subject(s)
Alveolar Epithelial Cells/cytology , Cellular Senescence/physiology , Macrophage Activation/physiology , Macrophages, Alveolar/physiology , Serpin E2/physiology , Transforming Growth Factor beta1/physiology , Alveolar Epithelial Cells/metabolism , Animals , Cells, Cultured , Chemokines/metabolism , Culture Media, Conditioned/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Cytokines/metabolism , Genes, p16 , Mice , Mice, Knockout , Pulmonary Fibrosis/pathology , RNA Interference , RNA, Small Interfering/genetics , Rats , Serpin E2/deficiency , Serpin E2/genetics
3.
FASEB J ; 33(11): 12364-12373, 2019 11.
Article in English | MEDLINE | ID: mdl-31408372

ABSTRACT

Cellular senescence is a phenotypic state that contributes to age-related diseases through the secretion of matrix-degrading and inflammatory molecules. An emerging therapeutic strategy for osteoarthritis (OA) is to selectively eliminate senescent cells by initiating apoptosis. This study establishes a cartilage explant model of senescence induction and senolytic clearance using p16Ink4a expression as a biomarker of senescence. Growth-factor stimulation of explants increased the expression of p16Ink4a at both the mRNA and protein levels. Applying this culture system to cartilage from p16tdTom reporter mice (a knockin allele with tdTomato fluorescent protein regulated by the endogenous p16Ink4a promoter) demonstrated the emergence of a p16-high population that was quantified using flow cytometry for tdTomato. Cell sorting was used to separate chondrocytes based on tdTomato fluorescence and p16-high cells showed higher senescence-associated ß-galactosidase activity and increased gene expression of the senescence-associated secretory phenotype as compared with p16-low cells. The potential for effective senolysis within the cartilage extracellular matrix was assessed using navitoclax (ABT-263). Navitoclax treatment reduced the percentage of p16-high cells from 17.9 to 6.1% (mean of 13 matched pairs; P < 0.001) and increased cleaved caspase-3 confirmed apoptotic activity. Together, these findings establish a physiologically relevant cartilage explant model for testing the induction and elimination of senescent chondrocytes, which will support investigations of senolytic therapy for OA.-Sessions, G. A., Copp, M. E., Liu, J.-Y., Sinkler, M. A., D'Costa, S., Diekman, B. O. Controlled induction and targeted elimination of p16INK4a-expressing chondrocytes in cartilage explant culture.


Subject(s)
Cartilage, Articular/cytology , Cell Separation , Cellular Senescence , Chondrocytes/cytology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Aniline Compounds/pharmacology , Animals , Apoptosis , Cyclin-Dependent Kinase Inhibitor p16/analysis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Mice , Mice, Inbred C57BL , Osteoarthritis/therapy , Sulfonamides/pharmacology , Tissue Culture Techniques
4.
Zhonghua Fu Chan Ke Za Zhi ; 55(11): 784-790, 2020 Nov 25.
Article in Zh | MEDLINE | ID: mdl-33228350

ABSTRACT

Objective: To evaluate the value of p16INK4a detected by p16INK4a immunostaining as a new generation of cervical cytology for primary screening and secondary screening in population-based cervical cancer screening, and in improving cytological diagnosis. Methods: Between 2016 and 2018, 5 747 non-pregnant women aged 25-65 years with sexual history were recruited and underwent cervical cancer screening via high-risk (HR)-HPV/liquid-based cytological test (LCT) test in Shenzhen and surrounding areas. All slides were immuno-stained using p16INK4a technology, among them, 902 cases were offered p16INK4a detection during primary screening, and the remaining 4 845 cases were called-back by the virtue of abnormal HR-HPV and LCT results for p16INK4a staining. Participants with complete LCT examination, HR-HPV test, p16INK4a staining and histopathological examination results were included in this study. The performance of p16INK4a in primary and secondary screening, and in assisting cytology to detect high grade squamous intraepithelial lesion [HSIL, including cervical intraepithelial neoplasia (CIN) Ⅱ or Ⅲ] or worse [HSIL (CIN Ⅱ)+ or HSIL (CIN Ⅲ)+] were analyzed. Results: (1) One-thousand and ninety-seven cases with complete data of p16INK4a and histology were included. Pathological diagnosis: 995 cases of normal cervix, 37 cases of low grade squamous intraepithelial lesion (LSIL), 64 cases of HSIL and one case of cervical cancer were found. Among them, 65 cases of HSIL (CIN Ⅱ)+ and 34 cases of HSIL (CIN Ⅲ)+ were detected. The positive rate of p16INK4a in HSIL (CIN Ⅱ)+ was higher than that in CINⅠ or normal pathology (89.2% vs 10.2%; P<0.01). (2) p16INK4a as primary screening for HSIL (CIN Ⅱ)+ or HSIL (CIN Ⅲ)+ was equally sensitive to primary HR-HPV screening (89.2% vs 95.4%, 94.1% vs 94.1%; P>0.05), but more specific than HR-HPV screening (89.8% vs 82.5%, 87.7% vs 80.2%; P<0.05). p16INK4a was equally sensitive and similarly specific to cytology (≥LSIL; P>0.05). (3) The specificity of LCT adjunctive p16INK4a for detecting HSIL (CIN Ⅱ)+ or HSIL (CIN Ⅲ)+ were higher than that of LCT alone or adjunctive HR-HPV (P<0.01), while the sensitivity were similar (P>0.05). (4) p16INK4a staining as secondary screening: p16INK4a was significantly more specific (94.1% vs 89.7%, 91.9% vs 87.4%; P<0.01) and comparably sensitive (84.6% vs 90.8%, 88.2% vs 91.2%; P>0.05) to cytology for triaging primary HR-HPV screening. HPV 16/18 to colposcopy and triage other HR-HPV with p16INK4a was equally sensitive (88.2% vs 94.1%; P=0.500) and more specific (88.3% vs 83.0%; P<0.01) than HPV 16/18 to colposcopy and triage other HR-HPV with LCT≥ atypical squamous cells of undetermined significance (ASCUS), and the referral rate decreased (14.0% vs 19.4%; P=0.005). Conclusions: For primary screening, p16INK4a is equally specific to cytology and equally sensitive to HR-HPV screening. p16INK4a alone could be an efficient triage after primary HR-HPV screening. In addition, p16INK4a immunostaining could be used as an ancillary tool to cervical cytological diagnosis, and improves its accuracy in cervical cancer screening.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/analysis , Cyclin-Dependent Kinase Inhibitor p16/immunology , Early Detection of Cancer/methods , Immunohistochemistry/methods , Papillomavirus Infections/diagnosis , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Neoplasms/diagnosis , Adult , Aged , Biomarkers, Tumor/analysis , Cyclin-Dependent Kinase Inhibitor p16/physiology , Early Detection of Cancer/statistics & numerical data , Female , Human papillomavirus 16 , Human papillomavirus 18 , Humans , Middle Aged , Papillomaviridae/genetics , Papillomaviridae/isolation & purification , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , Pregnancy , Sensitivity and Specificity , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/virology , Vaginal Smears , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Dysplasia/virology
5.
Acta Biochim Biophys Sin (Shanghai) ; 49(11): 1015-1021, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29036495

ABSTRACT

Previous studies have shown that the differentiation potential declines with the age of progenitor cells and is linked to altered levels of senescence markers. The purpose of this study was to test whether senescence marker p16 affects age-related tenogenic differentiation in tendon stem/progenitor cells (TSPCs). Young and aged TSPCs were isolated from young/healthy and aged/degenerated human Achilles tendons, respectively. Cellular aging and capacity for tenogenic differentiation were examined. The results showed that the tenogenic differentiation capacity of TSPCs significantly decreases with advancing age. TSPCs from elderly donors showed upregulation of senescence-associated ß-galactosidase and p16 and concurrently a decrease in Type I collagen concentration and in the expressions of tendon-related markers: Scx, Tnmd, Bgn, Dcn, Col1, and Col3. Overexpression of p16 significantly inhibited tenogenic differentiation of young TSPCs. Analysis of the mechanism revealed that this effect is mediated by microRNA-217 and its target EGR1. These results indicated that p16 inhibits tenogenic differentiation of TSPCs via microRNA signaling pathways, which may serve as a potential target for the prevention or treatment in the future.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/physiology , Early Growth Response Protein 1/physiology , MicroRNAs/physiology , Signal Transduction/physiology , Stem Cells/metabolism , Tendons/cytology , Adolescent , Adult , Age Factors , Aged , Cell Differentiation , Humans , Middle Aged , Stem Cells/cytology , Young Adult
6.
Tumour Biol ; 37(6): 7625-32, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26687648

ABSTRACT

Phosphatase and tensin homolog (PTEN) and p16INK4a (p16) genes are tumor suppressor genes, associated with epigenetic alterations. PTEN and p16 promoter hypermethylation is a major epigenetic silencing mechanism leading to cancer. The cooperation between PTEN and p16 in pathogenesis of cancers suggest that their combination might be considered as potential molecular marker for specific subgroups of patients. Hence, the present study aimed to investigate whether PTEN and p16 promoter methylations were involved in oral squamous cell carcinoma (OSCC) in south Indian subjects. DNA methylation quantitative analyses of the two candidate tumor suppressor genes PTEN and p16 were performed by methylation-specific polymerase chain reaction (MSP). Fifty OSCC biopsy samples and their corresponding non-malignant portions as controls were studied comparatively. The methylation status was correlated with the clinical manifestations. Twelve out of 50 patients (24 %) were found to be methylated for PTEN gene, whereas methylation of the p16 gene occurred in 19 out of 50 cases (38 %). A statistically significant result was obtained (P = <0.0001 and 0.017) for both PTEN and p16 genes. PTEN and p16 promoter methylation may be the main mechanism leading to the low expression of PTEN and p16 genes indicating the progress of tumor development. Our data suggest that a low PTEN and p16 expression due to methylation may contribute to the cancer progression and could be useful for prognosis of OSCC. Therefore, analysis of promoter methylation in such genes may provide a biomarker valuable for early detection of oral cancer.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Methylation , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genes, p16 , Mouth Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Adult , Aged , Biomarkers, Tumor , Biopsy , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/pathology , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p16/analysis , Cyclin-Dependent Kinase Inhibitor p16/physiology , DNA, Neoplasm/chemistry , Early Detection of Cancer , Female , Humans , India/epidemiology , Male , Middle Aged , Models, Biological , Mouth Neoplasms/epidemiology , Mouth Neoplasms/pathology , PTEN Phosphohydrolase/analysis , PTEN Phosphohydrolase/physiology , Promoter Regions, Genetic/genetics , Risk Factors
7.
Nucleic Acids Res ; 42(7): e60, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24500204

ABSTRACT

Identifying differential features between conditions is a popular approach to understanding molecular features and their mechanisms underlying a biological process of particular interest. Although many tests for identifying differential expression of gene or gene sets have been proposed, there was limited success in developing methods for differential interactions of genes between conditions because of its computational complexity. We present a method for Evaluation of Dependency DifferentialitY (EDDY), which is a statistical test for differential dependencies of a set of genes between two conditions. Unlike previous methods focused on differential expression of individual genes or correlation changes of individual gene-gene interactions, EDDY compares two conditions by evaluating the probability distributions of dependency networks from genes. The method has been evaluated and compared with other methods through simulation studies, and application to glioblastoma multiforme data resulted in informative cancer and glioblastoma multiforme subtype-related findings. The comparison with Gene Set Enrichment Analysis, a differential expression-based method, revealed that EDDY identifies the gene sets that are complementary to those identified by Gene Set Enrichment Analysis. EDDY also showed much lower false positives than Gene Set Co-expression Analysis, a method based on correlation changes of individual gene-gene interactions, thus providing more informative results. The Java implementation of the algorithm is freely available to noncommercial users. Download from: http://biocomputing.tgen.org/software/EDDY.


Subject(s)
Gene Regulatory Networks , Cyclin-Dependent Kinase Inhibitor p16/physiology , Data Interpretation, Statistical , Gene Expression , Glioblastoma/classification , Glioblastoma/genetics , Humans , Tumor Suppressor Protein p53/physiology
8.
Fukuoka Igaku Zasshi ; 107(1): 12-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27333655

ABSTRACT

The expression of p16(INK4a) has been reported to induce cell-cycle arrest and cellular senescence. The p16(INK4a) expression has never been examined in human mast cells and mastocytosis. We immunohistologically examined the expression of p16(INK4a) and tryptase in 5 normal human skin and 4 mastocytosis. In normal mast cells, only 5.9 ± 3.4 (mean ± standard deviation) % of tryptase-positive mast cells coexpressed p16(INK4a). However, significantly higher percentage (86.0 ± 14.1%) of tryptase-positive tumor cells was immunoreactive to p16(INK4a) in all of 4 mastocytosis. The p16(INK4a) overexpression may induce the senescence of neoplastic mast cells to undergo spontaneous regression of mastocytosis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression , Urticaria Pigmentosa/genetics , Cell Cycle Checkpoints/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/physiology , Humans , Mast Cells/pathology , Mastocytosis, Cutaneous/genetics , Mastocytosis, Cutaneous/pathology , Neoplasm Regression, Spontaneous/genetics , Neoplasm Regression, Spontaneous/pathology , Skin/metabolism , Skin/pathology , Tryptases/genetics , Tryptases/metabolism , Urticaria Pigmentosa/pathology
9.
Br J Haematol ; 171(5): 818-29, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26403224

ABSTRACT

Acute lymphoblastic leukaemia (ALL) in infants is an intractable cancer in childhood. Although recent intensive chemotherapy progress has considerably improved ALL treatment outcome, disease cure is often accompanied by undesirable long-term side effects, and efficient, less toxic molecular targeting therapies have been anticipated. In infant ALL cells with KMT2A (MLL) fusion, the microRNA let-7b (MIRLET7B) is significantly downregulated by DNA hypermethylation of its promoter region. We show here that the expression of HMGA2, one of the oncogenes repressed by MIRLET7B, is reversely upregulated in infant ALL leukaemic cells, particularly in KMT2A-AFF1 (MLL-AF4) positive ALL. In addition to the suppression of MIRLET7B, KMT2A fusion proteins positively regulate the expression of HMGA2. HMGA2 is one of the negative regulators of CDKN2A gene, which encodes the cyclin-dependent kinase inhibitor p16(INK4A) . The HMGA2 inhibitor netropsin, when combined with demethylating agent 5-azacytidine, upregulated and sustained the expression of CDKN2A, which resulted in growth suppression of KMT2A-AFF1-expressing cell lines. This effect was more apparent compared to treatment with 5-azacytidine alone. These results indicate that the MIRLET7B-HMGA2-CDKN2A axis plays an important role in cell proliferation of leukaemic cells and could be a possible molecular target for the therapy of infant ALL with KMT2A-AFF1.


Subject(s)
HMGA2 Protein/antagonists & inhibitors , MicroRNAs/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Azacitidine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/physiology , DNA Methylation/drug effects , DNA-Binding Proteins/metabolism , Drug Synergism , Gene Knockdown Techniques , Genes, p16 , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/physiology , Humans , Infant , MicroRNAs/physiology , Molecular Targeted Therapy/methods , Myeloid-Lymphoid Leukemia Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/physiology , Netropsin/pharmacology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/drug effects , Transcriptional Elongation Factors , Up-Regulation
10.
Blood ; 121(17): 3402-12, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23440244

ABSTRACT

ABT-737 is a promising chemotherapeutic agent that promotes apoptosis by acting as a selective BH3 mimetic to neutralize Bcl-2-like family members. One shortcoming with its use is that Mcl-1, a member of the Bcl-2 family, is poorly inhibited by ABT-737 and thus is a major cause of resistance. We performed a short hairpin RNA (shRNA)-based drop-out screen to identify novel genes and pathways that could reverse resistance to ABT-737 treatment in Eµ-myc/Bcl-2 lymphoma cells engineered to rely on endogenous Mcl-1 for survival. Several drug-sensitive shRNAs were identified that were selectively depleted in the presence of ABT-737. Of these, 2 independent shRNAs targeting the RNA/DNA helicase Dhx9 were found to sensitize lymphomas to ABT-737 to an extent comparable to control Mcl-1 shRNAs. Although Dhx9 suppression sensitized both mouse and human cells to ABT-737 treatment, it did so without altering MCL-1 levels. Rather, loss of Dhx9 appeared to activate a p53-dependent apoptotic program, through aggravation of replicative stress, which was found to be both necessary and sufficient for the ABT-737-shDhx9 synthetic lethal relationship.


Subject(s)
Biphenyl Compounds/pharmacology , DEAD-box RNA Helicases/genetics , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Lymphoma/genetics , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Sulfonamides/pharmacology , Animals , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p16/physiology , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/metabolism , Genes, Modifier , Humans , Lymphoma/pathology , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism
11.
Blood ; 119(3): 717-26, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22101896

ABSTRACT

Alterations of the BM microenvironment have been shown to occur after chemoradiotherapy, during aging, and after genetic manipulations of telomere length. Nevertheless, whether BM stromal cells adopt senescent features in response to these events is unknown. In the present study, we provide evidence that exposure to ionizing radiation (IR) leads murine stromal BM cells to express senescence markers, namely senescence-associated ß-galactosidase and increased p16(INK4a)/p19(ARF) expression. Long (8 weeks) after exposure of mice to IR, we observed a reduction in the number of stromal cells derived from BM aspirates, an effect that we found to be absent in irradiated Ink4a/arf-knockout mice and to be mostly independent of the CFU potential of the stroma. Such a reduction in the number of BM stromal cells was specific, because stromal cells isolated from collagenase-treated bones were not reduced after IR. Surprisingly, we found that exposure to IR leads to a cellular nonautonomous and Ink4a/arf-dependent effect on lymphopoiesis. Overall, our results reveal the distinct sensitivity of BM stromal cell populations to IR and suggest that long-term residual damage to the BM microenvironment can influence hematopoiesis in an Ink4a/arf-dependent manner.


Subject(s)
ADP-Ribosylation Factor 1/physiology , Bone Marrow/radiation effects , Cellular Senescence/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/physiology , Homeostasis/radiation effects , Radiation, Ionizing , Stromal Cells/radiation effects , Animals , Apoptosis , Blotting, Western , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Differentiation , Cell Proliferation , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/radiation effects , Lymphopoiesis/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Stromal Cells/metabolism , Stromal Cells/pathology
12.
J Gastroenterol Hepatol ; 29(7): 1418-26, 2014.
Article in English | MEDLINE | ID: mdl-25123601

ABSTRACT

BACKGROUND AND AIM: The incidence of distal gastric adenocarcinoma has significantly decreased, but gastric cardia adenocarcinoma has been on the rise. Cardia adenocarcinoma might be a specific entity distinct from the carcinoma of the rest stomach. The aim was to explore putative differences in p16(INK) (4a) -retinoblastoma (Rb) pathway and INK4a/ARF methylation between gastric cardia and distal adenocarcinomas. METHODS: Ninety-six cardia adenocarcinomas and 79 distal samples were analyzed for comparing p16(INK) (4a) -Rb expressions, INK4a/ARF deletion, and methylation using immunohistochemistry, polymerase chain reaction, and methylation-specific polymerase chain reaction. RESULTS: The expression of p16(INK) (4a) in cardia adenocarcinoma (43.2%) was significantly lower than in distal cases (75.0%, P < 0.05). As well, cardia adenocarcinoma showed lower expression of p14(ARF) compared with distal cases (34.1% vs 57.5%, P < 0.05). The incidence of p16(INK) (4a) deletion was 20.5% and 15.0%, while p14(ARF) deletion was 18.2% and 10.0% in cardia and distal adenocarcinomas, respectively, showing no significant differences between two entities. However, the incidences of p14(ARF) and p16(INK) (4a) methylation in cardia adenocarcinoma were significantly higher than in distal samples (p14(ARF) : 61.5% vs 43.6%; p16(INK) (4a) : 73.1% vs 51.3%, P < 0.05). INK4a/ARF methylations were more prevalent in poorly differentiated cardia carcinoma compared with poorly differentiated distal cases. CONCLUSIONS: There were differences in p16(INK) (4a) -Rb immunotypes and INK4a/ARF methylation between two entities, indicating that cardia adenocarcinoma may be different in cell proliferation, differentiation, and gene biomarkers compared with distal gastric adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/physiology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach/anatomy & histology , Aged , Cell Proliferation , Cell Transformation, Neoplastic , Humans , Male , Methylation , Middle Aged
13.
Proc Natl Acad Sci U S A ; 108(5): 1919-24, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21245331

ABSTRACT

Epstein-Barr virus (EBV) nuclear antigen 3C (EBNA3C) and EBNA3A are each essential for EBV conversion of primary human B lymphocytes into continuously proliferating lymphoblast cell lines (LCLs) and for maintaining LCL growth. We now find that EBNA3C and EBNA3A's essential roles are to repress p16(INK4A) and p14(ARF). In the absence of EBNA3C or EBNA3A, p16(INK4A) and p14(ARF) expression increased and cell growth ceased. EBNA3C inactivation did not alter p16(INK4A) promoter CpG methylation, but reduced already low H3K27me3, relative to resting B cells, and increased H3K4me3 and H3-acetylation, linking EBNA3C inactivation to histone modifications associated with increased transcription. Importantly, knockdown of p16(INK4A) or p14(ARF) partially rescued LCLs from EBNA3C or EBNA3A inactivation-induced growth arrest and knockdown of both rescued LCL growth, confirming central roles for p16(INK4A) and p14(ARF) in LCL growth arrest following EBNA3C or EBNA3A inactivation. Moreover, blockade of p16(INK4A) and p14(ARF) effects on pRb and p53 by human papilloma virus type 16 E7 and E6 expression, sustained LCL growth after EBNA3C or EBNA3A inactivation. These data indicate that EBNA3C and EBNA3A joint repression of CDKN2A p16(INK4A) and p14(ARF) is essential for LCL growth.


Subject(s)
Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Epstein-Barr Virus Nuclear Antigens/physiology , Tumor Suppressor Protein p14ARF/physiology , Cell Line , CpG Islands , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , Gene Knockdown Techniques , Humans , Promoter Regions, Genetic , Tumor Suppressor Protein p14ARF/genetics
14.
Biochem Biophys Res Commun ; 432(1): 34-9, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23376716

ABSTRACT

Normal cells undergo a growth-arrested status that is produced by p53-dependent down-regulation of histone H2AX. Immortality is developed after abrogation of the H2AX-diminished state, which is associated with genomic instability (often with tetraploidy) and the induction of mutations in either the Arf or p53 gene. However, the role of Arf in control of H2AX expression and genome stability is still unclear. Here, we show that both Arf and p53 are required for the down-regulation of H2AX and formation of the growth-arrested state. Wild-type (WT) mouse embryonic fibroblasts (MEFs) subjected to tetraploidization with DNA lesions did not undergo mitotic catastrophe-associated cell death and stayed in a growth-arrested state, until immortality was attained with mutations in the Arf/p53 module and recovery of H2AX expression. Whereas tetraploidization was essential for immortalization of WT MEFs, this event was not required for immortalization of MEFs containing mutations in Arf/p53 and these cells still underwent mitotic catastrophe-associated cell death. Thus, WT MEFs are protected from immortalization with genome stability, which is abrogated with tetraploidization and mutation of either Arf or p53.


Subject(s)
Cell Cycle Checkpoints , Cyclin-Dependent Kinase Inhibitor p16/physiology , Diploidy , Genomic Instability , Tetraploidy , Tumor Suppressor Protein p53/physiology , 3T3 Cells , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Histones/metabolism , Mice , Mice, Knockout , Mitosis , Tumor Suppressor Protein p53/genetics
15.
Blood ; 118(9): 2556-66, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21636855

ABSTRACT

The CDKN2A locus, which contains the tumor suppressor gene p16(INK4a), is associated with an increased risk of age-related inflammatory diseases, such as cardiovascular disease and type 2 diabetes, in which macrophages play a crucial role. Monocytes can polarize toward classically (CAMϕ) or alternatively (AAMϕ) activated macrophages. However, the molecular mechanisms underlying the acquisition of these phenotypes are not well defined. Here, we show that p16(INK4a) deficiency (p16(-/-)) modulates the macrophage phenotype. Transcriptome analysis revealed that p16(-/-) BM-derived macrophages (BMDMs) exhibit a phenotype resembling IL-4-induced macrophage polarization. In line with this observation, p16(-/-) BMDMs displayed a decreased response to classically polarizing IFNγ and LPS and an increased sensitivity to alternative polarization by IL-4. Furthermore, mice transplanted with p16(-/-) BM displayed higher hepatic AAMϕ marker expression levels on Schistosoma mansoni infection, an in vivo model of AAMϕ phenotype skewing. Surprisingly, p16(-/-) BMDMs did not display increased IL-4-induced STAT6 signaling, but decreased IFNγ-induced STAT1 and lipopolysaccharide (LPS)-induced IKKα,ß phosphorylation. This decrease correlated with decreased JAK2 phosphorylation and with higher levels of inhibitory acetylation of STAT1 and IKKα,ß. These findings identify p16(INK4a) as a modulator of macrophage activation and polarization via the JAK2-STAT1 pathway with possible roles in inflammatory diseases.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/deficiency , Genes, p16 , Inflammation/genetics , Janus Kinase 2/physiology , Macrophage Activation , STAT1 Transcription Factor/physiology , Animals , Bone Marrow Transplantation , Cyclin-Dependent Kinase Inhibitor p16/physiology , Cytokines/biosynthesis , I-kappa B Kinase/physiology , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Liver/metabolism , Liver/pathology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/physiology , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Processing, Post-Translational , Radiation Chimera , STAT6 Transcription Factor/physiology , Schistosomiasis/immunology , Signal Transduction
16.
Blood ; 117(12): 3257-67, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21245485

ABSTRACT

Previous authors have suggested that tumor suppressor expression promotes aging while preventing cancer, but direct experimental support for this cancer-aging hypothesis has been elusive. Here, by using somatic, tissue-specific inactivation of the p16(INK4a) tumor suppressor in murine T- or B-lymphoid progenitors, we report that ablation of p16(INK4a) can either rescue aging or promote cancer in a lineage-specific manner. Deletion of p16(INK4a) in the T lineage ameliorated several aging phenotypes, including thymic involution, decreased production of naive T cells, reduction in homeostatic T-cell proliferation, and attenuation of antigen-specific immune responses. Increased T-cell neoplasia was not observed with somatic p16(INK4a) inactivation in T cells. In contrast, B lineage-specific ablation of p16(INK4a) was associated with a markedly increased incidence of systemic, high-grade B-cell neoplasms, which limited studies of the effects of somatic p16(INK4a) ablation on B-cell aging. Together, these data show that expression of p16(INK4a) can promote aging and prevent cancer in related lymphoid progeny of a common stem cell.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Lymphocytes/metabolism , Lymphocytes/physiology , Neoplasms/genetics , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/physiology , Gene Deletion , Gene Expression/physiology , Integrases/genetics , Integrases/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocytes/pathology , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/metabolism , Lymphoid Progenitor Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Organ Specificity/genetics
17.
Blood ; 118(9): 2541-50, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21757616

ABSTRACT

Dysfunction of AML1/Runx1, a transcription factor, plays a crucial role in the development of many types of leukemia. Additional events are often required for AML1 dysfunction to induce full-blown leukemia; however, a mechanistic basis of their cooperation is still elusive. Here, we investigated the effect of AML1 deficiency on the development of MLL-ENL leukemia in mice. Aml1 excised bone marrow cells lead to MLL-ENL leukemia with shorter duration than Aml1 intact cells in vivo. Although the number of MLL-ENL leukemia-initiating cells is not affected by loss of AML1, the proliferation of leukemic cells is enhanced in Aml1-excised MLL-ENL leukemic mice. We found that the enhanced proliferation is the result of repression of p19(ARF) that is directly regulated by AML1 in MLL-ENL leukemic cells. We also found that down-regulation of p19(ARF) induces the accelerated onset of MLL-ENL leukemia, suggesting that p19(ARF) is a major target of AML1 in MLL-ENL leukemia. These results provide a new insight into a role for AML1 in the progression of leukemia.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Gene Expression Regulation, Leukemic/genetics , Leukemia, Biphenotypic, Acute/genetics , Neoplasm Proteins/physiology , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Bone Marrow Transplantation , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Core Binding Factor Alpha 2 Subunit/biosynthesis , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/physiology , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Radiation Chimera , Recombinant Fusion Proteins/physiology , Transcription, Genetic
18.
Blood ; 117(24): 6617-26, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21518927

ABSTRACT

Oncogene-induced senescence (OIS) is a barrier for tumor development. Oncogene-dependent DNA damage and activation of the ARF/p53 pathway play a central role in OIS and, accordingly, ARF and p53 are frequently mutated in human cancer. A number of leukemia/lymphoma-initiating oncogenes, however, inhibit ARF/p53 and only infrequently select for ARF or p53 mutations, suggesting the involvement of other tumor-suppressive pathways. We report that NPM-ALK, the initiating oncogene of anaplastic large cell lymphomas (ALCLs), induces DNA damage and irreversibly arrests the cell cycle of primary fibroblasts and hematopoietic progenitors. This effect is associated with inhibition of p53 and is caused by activation of the p16INK4a/pRb tumor-suppressive pathway. Analysis of NPM-ALK lymphomagenesis in transgenic mice showed p16INK4a-dependent accumulation of senescent cells in premalignant lesions and decreased tumor latency in the absence of p16INK4a. Accordingly, human ALCLs showed no expression of either p16INK4a or pRb. Up-regulation of the histone-demethylase Jmjd3 and de-methylation at the p16INK4a promoter contributed to the effect of NPM-ALK on p16INK4a, which was transcriptionally regulated. These data demonstrate that p16INK4a/pRb may function as an alternative pathway of oncogene-induced senescence, and suggest that the reactivation of p16INK4a expression might be a novel strategy to restore the senescence program in some tumors.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/physiology , Lymphoma/genetics , Protein-Tyrosine Kinases/physiology , Retinoblastoma Protein/physiology , Animals , Cells, Cultured , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lymphoma/metabolism , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/prevention & control , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/physiology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology
19.
Nat Cell Biol ; 8(11): 1291-7, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17028578

ABSTRACT

The p16(INK4a) cyclin-dependent kinase inhibitor has a key role in establishing stable G1 cell-cycle arrest through activating the retinoblastoma (Rb) tumour suppressor protein pRb in cellular senescence. Here, we show that the p16(INK4a) /Rb-pathway also cooperates with mitogenic signals to induce elevated intracellular levels of reactive oxygen species (ROS), thereby activating protein kinase Cdelta (PKCdelta) in human senescent cells. Importantly, once activated by ROS, PKCdelta promotes further generation of ROS, thus establishing a positive feedback loop to sustain ROS-PKCdelta signalling. Sustained activation of ROS-PKCdelta signalling irreversibly blocks cytokinesis, at least partly through reducing the level of WARTS (also known as LATS1), a mitotic exit network (MEN) kinase required for cytokinesis, in human senescent cells. This irreversible cytokinetic block is likely to act as a second barrier to cellular immortalization ensuring stable cell-cycle arrest in human senescent cells. These results uncover an unexpected role for the p16(INK4a)-Rb pathway and provide a new insight into how senescent cell-cycle arrest is enforced in human cells.


Subject(s)
Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Retinoblastoma Protein/physiology , Signal Transduction/physiology , Acetophenones/pharmacology , Acetylcysteine/pharmacology , Benzopyrans/pharmacology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Line , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dose-Response Relationship, Drug , Humans , Immunoblotting , Models, Biological , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction/drug effects , Time Factors , Transfection
20.
J Immunol ; 187(12): 6527-38, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22095712

ABSTRACT

The innate immune system is the first line of defense against invading organisms, and TLRs are the main sensors of microbial components, initiating signaling pathways that induce the production of proinflammatory cytokines and type I IFNs. An antiviral action for the tumor suppressor alternative reading frame (ARF) has been reported; however, the precise role of ARF in innate immunity is unknown. In this study, we show that ARF plays an important role in regulation of inflammatory responses. In peritoneal macrophages and bone marrow-derived macrophages from ARF-deficient animals, the induction of proinflammatory cytokines and chemokines by TLR ligands was severely impaired. The altered responses of ARF(-/-) cells to TLR ligands result from aberrant activation of intracellular signaling molecules including MAPKs, IκBα degradation, and NF-κB activation. Additionally, animals lacking ARF were resistant to LPS-induced endotoxic shock. This impaired activation of inflammation in ARF(-/-) mice was not restricted to TLRs, as it was also shown in response to non-TLR signaling pathways. Thus, ARF(-/-) mice were also unable to trigger a proper inflammatory response in experimental peritonitis or in 12-O-tetradecanoylphorbol-13-acetate-induced edema. Overexpression of ARF, but not its downstream target p53, rescued the ARF-deficient phenotype, increasing TLR4 levels and restoring inflammatory reaction. An increase in the E2F1 protein levels observed in ARF(-/-) macrophages at basal condition and after LPS stimulation may be involved in the impaired response in this system, as E2F1 has been described as an inflammatory suppressor. These results indicate that tumor suppressor ARF is a new regulator of inflammatory cell signaling.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/physiology , Immunity, Innate , Inflammation Mediators/physiology , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/metabolism , Toll-Like Receptors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL