Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nature ; 633(8030): 678-685, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39112713

ABSTRACT

Oncogenic RAS-induced senescence (OIS) is an autonomous tumour suppressor mechanism associated with premalignancy1,2. Achieving this phenotype typically requires a high level of oncogenic stress, yet the phenotype provoked by lower oncogenic dosage remains unclear. Here we develop oncogenic RAS dose-escalation models in vitro and in vivo, revealing a RAS dose-driven non-linear continuum of downstream phenotypes. In a hepatocyte OIS model in vivo, ectopic expression of NRAS(G12V) does not induce tumours, in part owing to OIS-driven immune clearance3. Single-cell RNA sequencing analyses reveal distinct hepatocyte clusters with typical OIS or progenitor-like features, corresponding to high and intermediate levels of NRAS(G12V), respectively. When titred down, NRAS(G12V)-expressing hepatocytes become immune resistant and develop tumours. Time-series monitoring at single-cell resolution identifies two distinct tumour types: early-onset aggressive undifferentiated and late-onset differentiated hepatocellular carcinoma. The molecular signature of each mouse tumour type is associated with different progenitor features and enriched in distinct human hepatocellular carcinoma subclasses. Our results define the oncogenic dosage-driven OIS spectrum, reconciling the senescence and tumour initiation phenotypes in early tumorigenesis.


Subject(s)
Carcinogenesis , Cellular Senescence , Hepatocytes , Liver Neoplasms , Oncogene Protein p21(ras) , Animals , Female , Humans , Male , Mice , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Phenotype , Single-Cell Gene Expression Analysis
2.
Chembiochem ; 24(1): e202200450, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36336658

ABSTRACT

The protein high mobility group A1 (HMGA1) is an important regulator of chromatin organization and function. However, the mechanisms by which it exerts its biological function are not fully understood. Here, we report that the HMGA isoform, HMGA1a, nucleates into foci that display liquid-like properties in the nucleus, and that the protein readily undergoes phase separation to form liquid condensates in vitro. By bringing together machine-leaning modelling, cellular and biophysical experiments and multiscale simulations, we demonstrate that phase separation of HMGA1a is promoted by protein-DNA interactions, and has the potential to be modulated by post-transcriptional effects such as phosphorylation. We further show that the intrinsically disordered C-terminal tail of HMGA1a significantly contributes to its phase separation through electrostatic interactions via AT hooks 2 and 3. Our work sheds light on HMGA1 phase separation as an emergent biophysical factor in regulating chromatin structure.


Subject(s)
Chromatin , HMGA1a Protein , Chromatin/metabolism , HMGA1a Protein/genetics , HMGA1a Protein/chemistry , HMGA1a Protein/metabolism , Cell Nucleus/metabolism , DNA/metabolism , Phosphorylation
3.
Genes Dev ; 27(16): 1800-8, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23964094

ABSTRACT

Senescence is a stress-responsive form of stable cell cycle exit. Senescent cells have a distinct gene expression profile, which is often accompanied by the spatial redistribution of heterochromatin into senescence-associated heterochromatic foci (SAHFs). Studying a key component of the nuclear lamina lamin B1 (LMNB1), we report dynamic alterations in its genomic profile and their implications for SAHF formation and gene regulation during senescence. Genome-wide mapping reveals that LMNB1 is depleted during senescence, preferentially from the central regions of lamina-associated domains (LADs), which are enriched for Lys9 trimethylation on histone H3 (H3K9me3). LMNB1 knockdown facilitates the spatial relocalization of perinuclear H3K9me3-positive heterochromatin, thus promoting SAHF formation, which could be inhibited by ectopic LMNB1 expression. Furthermore, despite the global reduction in LMNB1 protein levels, LMNB1 binding increases during senescence in a small subset of gene-rich regions where H3K27me3 also increases and gene expression becomes repressed. These results suggest that LMNB1 may contribute to senescence in at least two ways due to its uneven genome-wide redistribution: first, through the spatial reorganization of chromatin and, second, through gene repression.


Subject(s)
Cellular Senescence/genetics , Chromatin Assembly and Disassembly/genetics , Heterochromatin/metabolism , Lamin Type B/metabolism , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Gene Expression Regulation , Heterochromatin/chemistry , Histones/metabolism , Lamin Type B/genetics , Protein Binding , Protein Structure, Tertiary
4.
Mol Cell ; 47(2): 203-14, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22795131

ABSTRACT

The expansion of repressive epigenetic marks has been implicated in heterochromatin formation during embryonic development, but the general applicability of this mechanism is unclear. Here we show that nuclear rearrangement of repressive histone marks H3K9me3 and H3K27me3 into nonoverlapping structural layers characterizes senescence-associated heterochromatic foci (SAHF) formation in human fibroblasts. However, the global landscape of these repressive marks remains unchanged upon SAHF formation, suggesting that in somatic cells, heterochromatin can be formed through the spatial repositioning of pre-existing repressively marked histones. This model is reinforced by the correlation of presenescent replication timing with both the subsequent layered structure of SAHFs and the global landscape of the repressive marks, allowing us to integrate microscopic and genomic information. Furthermore, modulation of SAHF structure does not affect the occupancy of these repressive marks, nor vice versa. These experiments reveal that high-order heterochromatin formation and epigenetic remodeling of the genome can be discrete events.


Subject(s)
Chromatin/chemistry , Heterochromatin/chemistry , Histones/metabolism , Bromodeoxyuridine/pharmacology , Cellular Senescence , Chromosomes/ultrastructure , Epigenesis, Genetic , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Gene Silencing , Genome , Genome-Wide Association Study , Histones/chemistry , Humans , Laser Scanning Cytometry/methods , Microscopy, Fluorescence/methods
5.
PLoS Genet ; 11(3): e1005053, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25790137

ABSTRACT

The downstream functions of the DNA binding tumor suppressor p53 vary depending on the cellular context, and persistent p53 activation has recently been implicated in tumor suppression and senescence. However, genome-wide information about p53-target gene regulation has been derived mostly from acute genotoxic conditions. Using ChIP-seq and expression data, we have found distinct p53 binding profiles between acutely activated (through DNA damage) and chronically activated (in senescent or pro-apoptotic conditions) p53. Compared to the classical 'acute' p53 binding profile, 'chronic' p53 peaks were closely associated with CpG-islands. Furthermore, the chronic CpG-island binding of p53 conferred distinct expression patterns between senescent and pro-apoptotic conditions. Using the p53 targets seen in the chronic conditions together with external high-throughput datasets, we have built p53 networks that revealed extensive self-regulatory 'p53 hubs' where p53 and many p53 targets can physically interact with each other. Integrating these results with public clinical datasets identified the cancer-associated lipogenic enzyme, SCD, which we found to be directly repressed by p53 through the CpG-island promoter, providing a mechanistic link between p53 and the 'lipogenic phenotype', a hallmark of cancer. Our data reveal distinct phenotype associations of chronic p53 targets that underlie specific gene regulatory mechanisms.


Subject(s)
DNA Methylation/genetics , DNA-Binding Proteins/genetics , Protein Interaction Maps/genetics , Tumor Suppressor Protein p53/genetics , Aging/genetics , Apoptosis/genetics , Cell Line , CpG Islands/genetics , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Phenotype , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Tumor Suppressor Protein p53/metabolism
6.
Genes Dev ; 23(7): 798-803, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19279323

ABSTRACT

As a stress response, senescence is a dynamic process involving multiple effector mechanisms whose combination determines the phenotypic quality. Here we identify autophagy as a new effector mechanism of senescence. Autophagy is activated during senescence and its activation is correlated with negative feedback in the PI3K-mammalian target of rapamycin (mTOR) pathway. A subset of autophagy-related genes are up-regulated during senescence: Overexpression of one of those genes, ULK3, induces autophagy and senescence. Furthermore, inhibition of autophagy delays the senescence phenotype, including senescence-associated secretion. Our data suggest that autophagy, and its consequent protein turnover, mediate the acquisition of the senescence phenotype.


Subject(s)
Aging/physiology , Autophagy/physiology , Mitosis/physiology , Feedback, Physiological/physiology , Gene Expression Regulation , Humans , Immunohistochemistry , Microtubule-Associated Proteins/metabolism , Neoplasms/physiopathology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases
7.
Bioinformatics ; 30(15): 2155-61, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24711654

ABSTRACT

MOTIVATION: In metabolomics, the goal is to identify and measure the concentrations of different metabolites (small molecules) in a cell or a biological system. The metabolites form an important layer in the complex metabolic network, and the interactions between different metabolites are often of interest. It is crucial to perform proper normalization of metabolomics data, but current methods may not be applicable when estimating interactions in the form of correlations between metabolites. We propose a normalization approach based on a mixed model, with simultaneous estimation of a correlation matrix. We also investigate how the common use of a calibration standard in nuclear magnetic resonance (NMR) experiments affects the estimation of correlations. RESULTS: We show with both real and simulated data that our proposed normalization method is robust and has good performance when discovering true correlations between metabolites. The standardization of NMR data is shown in simulation studies to affect our ability to discover true correlations to a small extent. However, comparing standardized and non-standardized real data does not result in any large differences in correlation estimates. AVAILABILITY AND IMPLEMENTATION: Source code is freely available at https://sourceforge.net/projects/metabnorm/ CONTACT: alexandra.jauhiainen@ki.se SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Metabolomics/methods , Data Interpretation, Statistical , Humans , Magnetic Resonance Spectroscopy , Metabolic Networks and Pathways , Metabolomics/standards , Reference Standards
8.
Methods Mol Biol ; 2784: 215-225, 2024.
Article in English | MEDLINE | ID: mdl-38502489

ABSTRACT

DNA fluorescence in situ hybridization (FISH) enables the visualization of chromatin architecture and the interactions between genomic loci at a single-cell level, complementary to genome-wide methods such as Hi-C. DNA FISH uses fluorescent-labeled DNA probes targeted to the loci of interest, allowing for the analysis of their spatial positioning and proximity with microscopy. Here, we describe an optimized experimental procedure for DNA FISH, from probe design and sample preparation through imaging and image quantification. This protocol can be readily applied to querying the spatial positioning of genomic loci of interest.


Subject(s)
Chromatin , DNA , In Situ Hybridization, Fluorescence/methods , DNA/genetics , Chromatin/genetics , Chromosomes , DNA Probes/genetics , Fluorescent Dyes
9.
Nat Commun ; 15(1): 6891, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134516

ABSTRACT

HMGA1 is an abundant non-histone chromatin protein that has been implicated in embryonic development, cancer, and cellular senescence, but its specific role remains elusive. Here, we combine functional genomics approaches with graph theory to investigate how HMGA1 genomic deposition controls high-order chromatin networks in an oncogene-induced senescence model. While the direct role of HMGA1 in gene activation has been described previously, we find little evidence to support this. Instead, we show that the heterogeneous linear distribution of HMGA1 drives a specific 3D chromatin organization. HMGA1-dense loci form highly interactive networks, similar to, but independent of, constitutive heterochromatic loci. This, coupled with the exclusion of HMGA1-poor chromatin regions, leads to coordinated gene regulation through the repositioning of genes. In the absence of HMGA1, the whole process is largely reversed, but many regulatory interactions also emerge, amplifying the inflammatory senescence-associated secretory phenotype. Such HMGA1-mediated fine-tuning of gene expression contributes to the heterogeneous nature of senescence at the single-cell level. A similar 'buffer' effect of HMGA1 on inflammatory signalling is also detected in lung cancer cells. Our study reveals a mechanism through which HMGA1 modulates chromatin compartmentalization and gene regulation in senescence and beyond.


Subject(s)
Cellular Senescence , Chromatin , HMGA1a Protein , Humans , Cell Line, Tumor , Chromatin/metabolism , Chromatin/genetics , Gene Expression Regulation , Gene Regulatory Networks , HMGA1a Protein/metabolism , HMGA1a Protein/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology
10.
Cancer Res ; 84(18): 2968-2984, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39037766

ABSTRACT

Perturbation of cell polarity is a hallmark of pancreatic ductal adenocarcinoma (PDAC) progression. Scribble (SCRIB) is a well-characterized polarity regulator that has diverse roles in the pathogenesis of human neoplasms. To investigate the impact of SCRIB deficiency in PDAC development and progression, Scrib expression was genetically ablated in well-established mouse models of PDAC. Scrib loss in combination with KrasG12D did not influence development of pancreatic intraepithelial neoplasms in mice. However, Scrib deletion cooperated with KrasG12D and concomitant Trp53 heterozygous deletion to promote invasive PDAC and metastatic dissemination, leading to reduced overall survival. Immunohistochemical and transcriptome analyses revealed that Scrib-null tumors display a pronounced reduction of collagen content and an abundance of cancer-associated fibroblasts (CAF). Mechanistically, IL1α levels were reduced in Scrib-deficient tumors, and Scrib knockdown downregulated IL1α in mouse PDAC organoids (mPDO), which impaired CAF activation. Furthermore, Scrib loss increased YAP activation in mPDOs and established PDAC cell lines, enhancing cell survival. Clinically, SCRIB expression was decreased in human PDAC, and SCRIB mislocalization was associated with poorer patient outcome. These results indicate that SCRIB deficiency enhances cancer cell survival and remodels the tumor microenvironment to accelerate PDAC development and progression, establishing the tumor suppressor function of SCRIB in advanced pancreatic cancer. Significance: SCRIB loss promotes invasive pancreatic cancer development via both cell-autonomous and non-cell-autonomous processes and is associated with poorer outcomes, denoting SCRIB as a tumor suppressor and potential biomarker for the prediction of recurrence.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Tumor Suppressor Proteins , Animals , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Mice , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Humans , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Cell Line, Tumor , Membrane Proteins/genetics , Membrane Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Interleukin-1alpha/metabolism , Interleukin-1alpha/genetics , Organoids/metabolism , Organoids/pathology , Mice, Knockout , Neoplasm Metastasis , Gene Expression Regulation, Neoplastic , Tumor Microenvironment , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/deficiency
11.
Nat Aging ; 2(1): 31-45, 2022 01.
Article in English | MEDLINE | ID: mdl-37118356

ABSTRACT

Senescence is a fate-determined state, accompanied by reorganization of heterochromatin. Although lineage-appropriate genes can be temporarily repressed through facultative heterochromatin, stable silencing of lineage-inappropriate genes often involves the constitutive heterochromatic mark, histone H3 lysine 9 trimethylation (H3K9me3). The fate of these heterochromatic genes during senescence is unclear. In the present study, we show that a small number of lineage-inappropriate genes, exemplified by the LCE2 skin genes, are derepressed during senescence from H3K9me3 regions in fibroblasts. DNA FISH experiments reveal that these gene loci, which are condensed at the nuclear periphery in proliferative cells, are decompacted during senescence. Decompaction of the locus is not sufficient for LCE2 expression, which requires p53 and C/EBPß signaling. NLRP3, which is predominantly expressed in macrophages from an open topologically associated domain (TAD), is also derepressed in senescent fibroblasts due to the local disruption of the H3K9me3-rich TAD that contains it. NLRP3 has been implicated in the amplification of inflammatory cytokine signaling in senescence and aging, highlighting the functional relevance of gene induction from 'permissive' H3K9me3 regions in senescent cells.


Subject(s)
Heterochromatin , Histones , Heterochromatin/genetics , Histones/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Cellular Senescence/genetics , Gene Expression
12.
Proc Natl Acad Sci U S A ; 105(16): 6103-8, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18413597

ABSTRACT

Adenovirus E1A drives oncogenesis by targeting key regulatory pathways that are critical for cellular growth control. The interaction of E1A with p400 is essential for many E1A activities, but the downstream target of this interaction is unknown. Here, we present evidence that the oncoprotein transcription factor Myc is the target of this interaction. We show that E1A stabilizes Myc protein via p400 and promotes the coassociation of Myc and p400 at Myc target genes, leading to their transcriptional induction. We also show that E1A requires Myc for its ability to activate Myc-dependent gene expression and induce apoptosis, and that forced expression of Myc is sufficient to rescue the activity of an E1A-mutant defective in p400 binding. Together, these findings establish that Myc, via p400, is an essential downstream target of E1A.


Subject(s)
Adenovirus E1A Proteins/metabolism , Cell Transformation, Viral , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Adenovirus E1A Proteins/genetics , Cell Line , Chromatin Immunoprecipitation , Humans , Proto-Oncogene Proteins c-myc/genetics
13.
Nat Commun ; 11(1): 6049, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33247104

ABSTRACT

Senescence is a state of stable proliferative arrest, generally accompanied by the senescence-associated secretory phenotype, which modulates tissue homeostasis. Enhancer-promoter interactions, facilitated by chromatin loops, play a key role in gene regulation but their relevance in senescence remains elusive. Here, we use Hi-C to show that oncogenic RAS-induced senescence in human diploid fibroblasts is accompanied by extensive enhancer-promoter rewiring, which is closely connected with dynamic cohesin binding to the genome. We find de novo cohesin peaks often at the 3' end of a subset of active genes. RAS-induced de novo cohesin peaks are transcription-dependent and enriched for senescence-associated genes, exemplified by IL1B, where de novo cohesin binding is involved in new loop formation. Similar IL1B induction with de novo cohesin appearance and new loop formation are observed in terminally differentiated macrophages, but not TNFα-treated cells. These results suggest that RAS-induced senescence represents a cell fate determination-like process characterised by a unique gene expression profile and 3D genome folding signature, mediated in part through cohesin redistribution on chromatin.


Subject(s)
Cell Cycle Proteins/metabolism , Cellular Senescence/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Transcription, Genetic , CCCTC-Binding Factor/metabolism , Cell Differentiation/genetics , Cell Line , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genetic Loci , Genome , Humans , Interleukin-1/genetics , Macrophages/cytology , Promoter Regions, Genetic , Protein Binding/drug effects , Tumor Necrosis Factor-alpha/pharmacology , ras Proteins/metabolism , Cohesins
14.
Methods Mol Biol ; 1534: 89-98, 2017.
Article in English | MEDLINE | ID: mdl-27812870

ABSTRACT

Oncogene-induced senescence (OIS) is a highly dynamic process, involving several different effector mechanisms, the multitude and combination of which likely determines the quality of the phenotype (Pérez-Mancera et al., Nat Rev Cancer 14:547-558, 2014). Autophagy, a cellular degradation process, has been proposed to be one of these senescence effectors, although its functional relevance seems highly context dependent (Hoare et al., Semin Cancer Biol 21:397-404, 2011). A number of methods for monitoring autophagy are available, and several excellent protocols have been published in this journal (Klionsky et al., Autophagy 8:445-544, 2012; Tooze et al., Methods Mol Biol 1270:155-165, 2015; Tabata et al., Methods Mol Biol 931:449-466, 2013; Young and Tooze, Methods Mol Biol 445:147-157, 2008). The same principles apply to models of OIS in culture. Thus, in this chapter, we describe how to generate OIS cells using human diploid fibroblasts (HDFs), the best-characterized cell model of OIS, and how to detect autophagy, particularly focusing on immunofluorescence methods.


Subject(s)
Autophagy/genetics , Cellular Senescence/genetics , Microscopy, Fluorescence , Oncogenes/genetics , Biomarkers , Female , Fibroblasts/metabolism , Fluorescent Antibody Technique , Gene Expression , Genetic Vectors/genetics , Humans , Microscopy, Fluorescence/methods , Retroviridae/genetics , Transduction, Genetic
15.
Metabolomics ; 11(6): 1848-1863, 2015.
Article in English | MEDLINE | ID: mdl-26491426

ABSTRACT

To investigate metabolic changes during cellular transformation, we used a 1H NMR based metabolite-metabolite correlation analysis (MMCA) method, which permits analysis of homeostatic mechanisms in cells at the steady state, in an inducible cell transformation model. Transcriptomic data were used to further explain the results. Transformed cells showed many more metabolite-metabolite correlations than control cells. Some had intuitively plausible explanations: a shift from glycolysis to amino acid oxidation after transformation was accompanied by a strongly positive correlation between glucose and glutamine and a strongly negative one between lactate and glutamate; there were also many correlations between the branched chain amino acids and the aromatic amino acids. Others remain puzzling: after transformation strong positive correlations developed between choline and a group of five amino acids, whereas the same amino acids showed negative correlations with phosphocholine, a membrane phospholipid precursor. MMCA in conjunction with transcriptome analysis has opened a new window into the metabolome.

16.
Mol Biol Cell ; 26(17): 2971-85, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26133385

ABSTRACT

Cellular senescence is a widespread stress response and is widely considered to be an alternative cancer therapeutic goal. Unlike apoptosis, senescence is composed of a diverse set of subphenotypes, depending on which of its associated effector programs are engaged. Here we establish a simple and sensitive cell-based prosenescence screen with detailed validation assays. We characterize the screen using a focused tool compound kinase inhibitor library. We identify a series of compounds that induce different types of senescence, including a unique phenotype associated with irregularly shaped nuclei and the progressive accumulation of G1 tetraploidy in human diploid fibroblasts. Downstream analyses show that all of the compounds that induce tetraploid senescence inhibit Aurora kinase B (AURKB). AURKB is the catalytic component of the chromosome passenger complex, which is involved in correct chromosome alignment and segregation, the spindle assembly checkpoint, and cytokinesis. Although aberrant mitosis and senescence have been linked, a specific characterization of AURKB in the context of senescence is still required. This proof-of-principle study suggests that our protocol is capable of amplifying tetraploid senescence, which can be observed in only a small population of oncogenic RAS-induced senescence, and provides additional justification for AURKB as a cancer therapeutic target.


Subject(s)
Aurora Kinase B/antagonists & inhibitors , Polyploidy , Protein Kinase Inhibitors/pharmacology , Aurora Kinase B/genetics , Cell Division , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Nucleus/genetics , Cellular Senescence/drug effects , Cellular Senescence/genetics , Chromosome Segregation , Cytokinesis/genetics , HeLa Cells , High-Throughput Screening Assays/methods , Humans , Mitosis/drug effects , Mitosis/genetics , Phenotype , Small Molecule Libraries/pharmacology
17.
Ultrasound Med Biol ; 29(10): 1415-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14597338

ABSTRACT

Atherosclerotic diseases are thought to be less frequent in Asians compared with Caucasians. Unlike Caucasians, nearly half of Asians have a functional deficiency in the low Km aldehyde dehydrogenase (ALDH2), a key enzyme in alcohol metabolism, which potentially modifies the prevalence of atherosclerosis. This study examined the associations between ALDH2 genotypes ("typical homo," "hetero" or "atypical homo") and carotid atherosclerosis in 304 Japanese patients. As a measure of carotid atherosclerosis, plaque score (PS) was evaluated by B-mode ultrasonography. Age- and sex-adjusted PS was lower in "atypical homo" genotype patients (2.7 +/- 1.2 [mean +/- standard error], n=21) (p<0.05) and tended to be lower in "hetero" patients (4.5 +/- 0.5, n=116) (p=0.07) compared with "typical homo" patients (5.7 +/- 0.4, n=167). When we controlled for traditional cardiovascular risk factors and alcohol intake, the "atypical homo" genotype was found to be associated with lower PS (beta=-0.13, p<0.05). Based on these findings, the ALDH2 genotypes seem to be associated with the severity of carotid atherosclerosis, potentially modifying the prevalence of atherosclerosis in Asians.


Subject(s)
Aldehyde Dehydrogenase/genetics , Asian People/genetics , Carotid Artery Diseases/genetics , Genetic Predisposition to Disease , Aged , Cardiovascular Diseases/etiology , Carotid Artery Diseases/diagnostic imaging , Carotid Artery Diseases/enzymology , Echoencephalography/methods , Ethanol/administration & dosage , Female , Genotype , Humans , Japan , Male , Middle Aged , Risk Factors , Severity of Illness Index
18.
Methods Mol Biol ; 965: 1-13, 2013.
Article in English | MEDLINE | ID: mdl-23296648

ABSTRACT

It has been 50 years since cellular senescence was first described in human diploid fibroblasts (HDFs), yet its mechanism as well as its physiological and clinical implications are still not fully appreciated. Recent progress suggests that cellular senescence is a collective phenotype, composed of complex networks of effector programs. The balance and quality within the effector network varies depending on the cell type, the nature of the stress as well as the context. Therefore, understanding each of these effectors in the context of the whole network will be necessary in order to fully understand senescence as a whole. Furthermore, searching for new effector programs of senescence will help to define this heterogeneous and complex phenotype according to cellular contexts.


Subject(s)
Cellular Senescence , Phenotype , Animals , Autophagy , Gene Expression Regulation , Humans , Transcription, Genetic/genetics , ras Proteins/metabolism
19.
Autophagy ; 7(11): 1387-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21799306

ABSTRACT

Evidence for a connection between lysosomes and mTOR is emerging. Seminal work from the Sabatini laboratory has shown that mTOR can be recruited to the lysosomal surface in response to amino acids, in a Rag GTPase-dependent manner, to become activated by Rheb. However the biological significance of this is not fully understood. Recent work from our laboratory has shown that lysosomes spatially link mTOR and autophagy forming a cytoplasmic compartment in close proximity to the Golgi apparatus (GA) during oncogenic Ras-induced senescence. The TOR-autophagy spatial coupling compartment (TASCC) is enriched for autolysosomes, but largely excludes autophagosomes. Our data suggest that mTOR, which is a positive regulator of protein synthesis, is recruited, in part, by the amino acid-rich environment surrounding the autolysosomes. This then facilitates protein synthesis at the nearby rER-GA system, reinforcing lysosome and autophagy biogenesis. Proper TASCC formation contributes to the production of secretory proteins, which also utilizes the rER-GA system. Since mTOR inhibits autophagy during the initial stages of autophagosome formation, TASCC formation is likely to facilitate autophagy by sequestering mTOR, suggesting that the TASCC is a self-enhancing structure.


Subject(s)
Autophagy , Cellular Senescence , TOR Serine-Threonine Kinases/metabolism , Cell Compartmentation , Fibroblasts/cytology , Fibroblasts/metabolism , Golgi Apparatus/metabolism , Humans , Lysosomes/metabolism , Time Factors
20.
Aging Cell ; 10(1): 158-71, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21078114

ABSTRACT

Cellular senescence is an effective tumor-suppressive mechanism that causes a stable proliferative arrest in cells with potentially oncogenic alterations. Here, we have investigated the role of the p33ING1 tumor suppressor in the regulation of cellular senescence in human primary fibroblasts. We show that p33ING1 triggers a senescent phenotype in a p53-dependent fashion. Also, endogenous p33ING1 protein accumulates in chromatin in oncogene-senescent fibroblasts and its silencing by RNA interference impairs senescence triggered by oncogenes. Notably, the ability to induce senescence is lost in a mutant version of p33ING1 present in human tumors. Using specific point mutants, we further show that recognition of the chromatin mark H3K4me3 is essential for induction of senescence by p33ING1. Finally, we demonstrate that ING1-induced senescence is associated to a specific genetic signature with a strong representation of chemokine and cytokine signaling factors, which significantly overlaps with that of oncogene-induced senescence. In summary, our results identify ING1 as a critical epigenetic regulator of cellular senescence in human fibroblasts and highlight its role in control of gene expression in the context of this tumor-protective response.


Subject(s)
Cellular Senescence/genetics , Chromatin/metabolism , Epigenomics , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Biomarkers/metabolism , Cell Culture Techniques , Chemokines/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Genes, Regulator/physiology , Humans , Inhibitor of Growth Protein 1 , Intracellular Signaling Peptides and Proteins/metabolism , Microarray Analysis , Nuclear Proteins/metabolism , Point Mutation , Protein Binding/genetics , RNA Interference , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL