Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
2.
Cancer Cell ; 26(5): 653-67, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25517748

ABSTRACT

We isolated and analyzed, at single-nucleotide resolution, cancer-associated neochromosomes from well- and/or dedifferentiated liposarcomas. Neochromosomes, which can exceed 600 Mb in size, initially arise as circular structures following chromothripsis involving chromosome 12. The core of the neochromosome is amplified, rearranged, and corroded through hundreds of breakage-fusion-bridge cycles. Under selective pressure, amplified oncogenes are overexpressed, while coamplified passenger genes may be silenced epigenetically. New material may be captured during punctuated chromothriptic events. Centromeric corrosion leads to crisis, which is resolved through neocentromere formation or native centromere capture. Finally, amplification terminates, and the neochromosome core is stabilized in linear form by telomere capture. This study investigates the dynamic mutational processes underlying the life history of a special form of cancer mutation.


Subject(s)
Chromosomes, Human/genetics , Liposarcoma/genetics , Retroperitoneal Neoplasms/genetics , Aged , Carcinogenesis/genetics , Cell Line, Tumor , Centromere/genetics , Chromosome Aberrations , Female , Humans , Liposarcoma/pathology , Models, Genetic , Mutagenesis , Oncogenes , Retroperitoneal Neoplasms/pathology , Translocation, Genetic
3.
Chromosome Res ; 21(2): 101-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23580138

ABSTRACT

The first centromeric protein identified in any species was CENP-A, a divergent member of the histone H3 family that was recognised by autoantibodies from patients with scleroderma-spectrum disease. It has recently been suggested to rename this protein CenH3. Here, we argue that the original name should be maintained both because it is the basis of a long established nomenclature for centromere proteins and because it avoids confusion due to the presence of canonical histone H3 at centromeres.


Subject(s)
Autoantigens/genetics , Chromosomal Proteins, Non-Histone/genetics , Histones/genetics , Autoantigens/metabolism , Centromere , Centromere Protein A , Chromosomal Proteins, Non-Histone/metabolism , Histones/metabolism , Humans , Kinetochores , Scleroderma, Systemic/genetics , Terminology as Topic
4.
Nucleic Acids Res ; 41(8): 4447-58, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23444137

ABSTRACT

We have previously shown that α-thalassemia mental retardation X-linked (ATRX) and histone H3.3 are key regulators of telomeric chromatin in mouse embryonic stem cells. The function of ATRX and H3.3 in the maintenance of telomere chromatin integrity is further demonstrated by recent studies that show the strong association of ATRX/H3.3 mutations with alternative lengthening of telomeres in telomerase-negative human cancer cells. Here, we demonstrate that ATRX and H3.3 co-localize with the telomeric DNA and associated proteins within the promyelocytic leukemia (PML) bodies in mouse ES cells. The assembly of these telomere-associated PML bodies is most prominent at S phase. RNA interference (RNAi)-mediated knockdown of PML expression induces the disassembly of these nuclear bodies and a telomere dysfunction phenotype in mouse ES cells. Loss of function of PML bodies in mouse ES cells also disrupts binding of ATRX/H3.3 and proper establishment of histone methylation pattern at the telomere. Our study demonstrates that PML bodies act as epigenetic regulators by serving as platforms for the assembly of the telomeric chromatin to ensure a faithful inheritance of epigenetic information at the telomere.


Subject(s)
Cell Nucleus Structures/metabolism , Chromatin/metabolism , Embryonic Stem Cells/metabolism , Telomere/metabolism , Animals , Cell Line, Tumor , Cell Nucleus Structures/chemistry , DNA Helicases/analysis , DNA Repair , Epigenesis, Genetic , Histones/analysis , Humans , Mice , NIH 3T3 Cells , Nuclear Proteins/analysis , Nuclear Proteins/physiology , Phenotype , S Phase , X-linked Nuclear Protein
5.
PLoS Genet ; 8(9): e1002919, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22969435

ABSTRACT

Reduced DNA methylation has been reported in DICER1-deficient mouse ES cells. Reductions seen at pericentric satellite repeats have suggested that siRNAs are required for the proper assembly of heterochromatin. More recent studies have postulated that the reduced methylation is an indirect effect: the loss of Mir290 cluster miRNAs leads to upregulation of the transcriptional repressor RBL2 that targets the downregulation of DNA methyltransferase (Dnmt) genes. However, the observations have been inconsistent. We surmised that the inconsistency could be related to cell line "age," given that DNA methylation is lost progressively with passage in DNMT-deficient ES cells. We therefore subjected Dicer1(-/-) ES cells to two experimental regimes to rigorously test the level of functional DNMT activity. First, we cultured them for a prolonged period. If DNMT activity was reduced, further losses of methylation would occur. Second, we measured their DNMT activity in a rebound DNA methylation assay: DNA methylation was stripped from Cre/loxP conditionally mutant Dicer1 ES cells using a shRNA targeting Dnmt1 mRNA. Cre expression then converted these cells to Dicer1(-/-), allowing for DNMT1 recovery and forcing the cells to remethylate in the absence of RNAi. In both cases, we found functional DNMT activity to be normal. Finally, we also show that the level of RBL2 protein is not at excess levels in Dicer1(-/-) ES cells as has been assumed. These studies reveal that reduced functional DNMT activity is not a salient feature of DICER1-deficient ES cells. We suggest that the reduced DNA methylation sometimes observed in these cells could be due to stochastic alterations in DNA methylation patterns that could offer growth or survival advantages in culture, or to the dysregulation of pathways acting in opposition to the DNMT pathway.


Subject(s)
DEAD-box RNA Helicases/genetics , DNA Methylation , Embryonic Stem Cells/metabolism , Ribonuclease III/genetics , Animals , DEAD-box RNA Helicases/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Mice , MicroRNAs/metabolism , Retinoblastoma-Like Protein p130/metabolism , Ribonuclease III/metabolism
6.
Article in English | MEDLINE | ID: mdl-22719790

ABSTRACT

Tai chi exercise has been shown to improve physiological and psychosocial functions, well-being, quality of life, and disease conditions. The biological mechanisms by which tai chi exerts its holistic effects remain unknown. We investigated whether tai chi practice results in positive epigenetic changes at the molecular level. Design. The DNA methylation profiles of sixty CpG-dinucleotide marks in female tai chi practitioners (N = 237; 45-88 years old) who have been practising tai chi for three or more years were compared with those of age-matched control females (N = 263) who have never practised tai chi. Results. Six CpG marks originating from three different chromosomes reveal a significant difference (P < 0.05) between the two cohorts. Four marks show losses while two marks show gains in DNA methylation with age in the controls. In the tai chi cohort all six marks demonstrate significant slowing (by 5-70%) of the age-related methylation losses or gains observed in the controls, suggesting that tai chi practice may be associated with measurable beneficial epigenetic changes. Conclusions. The results implicate the potential use of DNA methylation as an epigenetic biomarker to better understand the biological mechanisms and the health and therapeutic efficacies of tai chi.

7.
Chromosoma ; 121(4): 327-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22527114

ABSTRACT

The centromere is a chromosomal structure that is essential for the accurate segregation of replicated eukaryotic chromosomes to daughter cells. In most centromeres, the underlying DNA is principally made up of repetitive DNA elements, such as tandemly repeated satellite DNA and retrotransposable elements. Paradoxically, for such an essential genomic region, the DNA is rapidly evolving both within and between species. In this review, we show that the centromere locus is a resilient structure that can undergo evolutionary cycles of birth, growth, maturity, death and resurrection. The birth phase is highlighted by examples in humans and other organisms where centromere DNA deletions or chromosome rearrangements can trigger the epigenetic assembly of neocentromeres onto genomic sites without typical features of centromere DNA. In addition, functional centromeres can be generated in the laboratory using various methodologies. Recent mapping of the foundation centromere mark, the histone H3 variant CENP-A, onto near-complete genomes has uncovered examples of new centromeres which have not accumulated centromere repeat DNA. During the growth period of the centromere, repeat DNA begins to appear at some, but not all, loci. The maturity stage is characterised by centromere repeat accumulation, expansions and contractions and the rapid evolution of the centromere DNA between chromosomes of the same species and between species. This stage provides inherent centromere stability, facilitated by repression of gene activity and meiotic recombination at and around the centromeres. Death to a centromere can result from genomic instability precipitating rearrangements, deletions, accumulation of mutations and the loss of essential centromere binding proteins. Surprisingly, ancestral centromeres can undergo resurrection either in the field or in the laboratory, via as yet poorly understood mechanisms. The underlying principle for the preservation of a centromeric evolutionary life cycle is to provide resilience and perpetuity for the all-important structure and function of the centromere.


Subject(s)
Biological Evolution , Centromere/genetics , Repetitive Sequences, Nucleic Acid , Animals , Centromere/chemistry , Histones/genetics , Histones/metabolism , Humans , Karyotype
8.
J Cell Sci ; 125(Pt 6): 1591-604, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22344259

ABSTRACT

In vertebrates, two condensin complexes exist, condensin I and condensin II, which have differing but unresolved roles in organizing mitotic chromosomes. To dissect accurately the role of each complex in mitosis, we have made and studied the first vertebrate conditional knockouts of the genes encoding condensin I subunit CAP-H and condensin II subunit CAP-D3 in chicken DT40 cells. Live-cell imaging reveals highly distinct segregation defects. CAP-D3 (condensin II) knockout results in masses of chromatin-containing anaphase bridges. CAP-H (condensin I)-knockout anaphases have a more subtle defect, with chromatids showing fine chromatin fibres that are associated with failure of cytokinesis and cell death. Super-resolution microscopy reveals that condensin-I-depleted mitotic chromosomes are wider and shorter, with a diffuse chromosome scaffold, whereas condensin-II-depleted chromosomes retain a more defined scaffold, with chromosomes more stretched and seemingly lacking in axial rigidity. We conclude that condensin II is required primarily to provide rigidity by establishing an initial chromosome axis around which condensin I can arrange loops of chromatin.


Subject(s)
Adenosine Triphosphatases/physiology , Chromosomes/genetics , DNA-Binding Proteins/physiology , Mitosis/genetics , Multiprotein Complexes/physiology , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Cell Line, Tumor , Chickens , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Knockout Techniques/methods , Multiprotein Complexes/deficiency , Multiprotein Complexes/genetics
9.
Proc Natl Acad Sci U S A ; 109(6): 1979-84, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22308327

ABSTRACT

Transcription of the centromeric regions has been reported to occur in G1 and S phase in different species. Here, we investigate whether transcription also occurs and plays a functional role at the mammalian centromere during mitosis. We show the presence of actively transcribing RNA polymerase II (RNAPII) and its associated transcription factors, coupled with the production of centromere satellite transcripts at the mitotic kinetochore. Specific inhibition of RNAPII activity during mitosis leads to a decrease in centromeric α-satellite transcription and a concomitant increase in anaphase-lagging cells, with the lagging chromosomes showing reduced centromere protein C binding. These findings demonstrate an essential role of RNAPII in the transcription of α-satellite DNA, binding of centromere protein C, and the proper functioning of the mitotic kinetochore.


Subject(s)
Centromere/metabolism , Mitosis , RNA Polymerase II/metabolism , Transcription, Genetic , Animals , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Mammalian/metabolism , DNA, Satellite/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Kinetochores/metabolism , Mice , Phosphoprotein Phosphatases , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
10.
Chromosoma ; 121(2): 169-79, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22080934

ABSTRACT

Although centromere protein B (CENP-B) is a highly conserved mammalian centromere protein, its function remains unknown. The presence of the protein is required to form artificial satellite DNA-based centromeres de novo, yet cenpb knockout mice are viable for multiple generations with no mitotic or meiotic defects, and the protein is not present at fully functional neocentromeres. Previous studies have suggested that the presence of functionally redundant paralogues of CENP-B may explain the lack of a phenotype in knockout mice, and the related Tigger-derived (TIGD) family of proteins has been implicated as the most likely candidate for such paralogues. Here, we describe an investigation of the centromere-binding properties of the three TIGD proteins most highly related to CENP-B through phylogenetic analysis through EGFP fusion studies and immunocytochemistry. Although two of the three proteins bound to human centromeres with low affinity when overexpressed as fusion proteins, the strongest candidate, TIGD3, demonstrated no native centromeric binding when using raised antibodies, either in human cells or in cenpb (-/-) mouse ES cells. We conclude that the existence of functional CENP-B paralogues is highly unlikely and that CENP-B acts alone at the centromere. Based on these data, we suggest a new, meiotic drive model of CENP-B action during centromere repositioning in evolution.


Subject(s)
Centromere Protein B/genetics , Centromere/genetics , Evolution, Molecular , Multigene Family/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Animals , Blotting, Western , Cloning, Molecular , Computational Biology , DNA Primers/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunohistochemistry , Mice , Mice, Knockout
11.
Headache ; 50(6): 965-72, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20618815

ABSTRACT

OBJECTIVE: To evaluate relative telomere length of female migraine patients. BACKGROUND: Migraine is a debilitating disorder affecting 6-28% of the population. Studies on the mechanisms of migraine have demonstrated genetic causes but the pathophysiology and subcellular effects of the disease remain poorly understood. Shortened telomere length is associated with age-related or chronic diseases, and induced stresses. Migraine attacks may impart significant stress on cellular function, thus this study investigates a correlation between shortening of telomeres and migraine. METHODS: Relative telomere length was measured using a previously described quantitative polymerase chain reaction method. A regression analysis was performed to assess differences in mean relative telomere length between migraine patients and healthy controls. RESULTS: The leukocyte telomeres of a cohort of 142 Caucasian female migraine subjects aged 18-77 years and 143 matched 17-77-year-old healthy control Caucasian women were examined. A significantly shorter relative telomere length was observed in the migraine group compared with the control group after adjusting for age and body mass index (P = .001). In addition, age of onset was observed to associate with the loss of relative telomere length, especially at early age of onset (<17 years old). No association was observed between relative telomere length and the severity and frequency of migraine attacks and the duration of migraine. CONCLUSION: Telomeres are shorter in migraine patients and there is more variation in telomere length in migraine patients.


Subject(s)
Migraine Disorders/genetics , Telomere/genetics , Adolescent , Adult , Aged , Female , Humans , Leukocytes, Mononuclear , Middle Aged , Polymerase Chain Reaction , Regression Analysis , White People
12.
Hum Mol Genet ; 19(8): 1618-32, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20118148

ABSTRACT

The fragile X syndrome (FXS) is caused by silencing of the fragile X mental retardation gene (FMR1) and the absence of its product, fragile X mental retardation protein (FMRP), resulting from CpG island methylation associated with large CGG repeat expansions (more than 200) termed full mutation (FM). We have identified a number of novel epigenetic markers for FXS using matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF MS), naming the most informative fragile X-related epigenetic element 1 (FREE1) and 2 (FREE2). Methylation of both regions was correlated with that of the FMR1 CpG island detected using Southern blot (FREE1 R = 0.97; P < 0.00001, n = 23 and FREE2 R = 0.93; P < 0.00001, n = 23) and negatively correlated with lymphocyte expression of FMRP (FREE1 R = -0.62; P = 0.01, n = 15 and FREE2 R = -0.55; P = 0.03, n = 15) in blood of partially methylated 'high functioning' FM males. In blood of FM carrier females, methylation of both markers was inversely correlated with the FMR1 activation ratio (FREE1 R = -0.93; P < 0.0001, n = 12 and FREE2 R = -0.95; P < 0.0001, n = 9). In a sample set of 49 controls, 18 grey zone (GZ 40-54 repeats), 22 premutation (PM 55-170 repeats) and 22 (affected) FXS subjects, the FREE1 methylation pattern was consistent between blood and chorionic villi as a marker of methylated FM alleles and could be used to differentiate FXS males and females from controls, as well as from carriers of GZ/PM alleles, but not between GZ and PM alleles and controls. Considering its high-throughput and specificity for pathogenic FM alleles, low cost and minimal DNA requirements, FREE MALDI-TOF MS offers a unique tool in FXS diagnostics and newborn population screening.


Subject(s)
DNA Methylation , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Transcriptional Activation , Aged , Alleles , Base Sequence , Cell Line , CpG Islands , Female , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Humans , Male , Middle Aged , Molecular Sequence Data
13.
Genome Res ; 20(3): 351-60, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20110566

ABSTRACT

ATRX (alpha thalassemia/mental retardation syndrome X-linked) belongs to the SWI2/SNF2 family of chromatin remodeling proteins. Besides the ATPase/helicase domain at its C terminus, it contains a PHD-like zinc finger at the N terminus. Mutations in the ATRX gene are associated with X-linked mental retardation (XLMR) often accompanied by alpha thalassemia (ATRX syndrome). Although ATRX has been postulated to be a transcriptional regulator, its precise roles remain undefined. We demonstrate ATRX localization at the telomeres in interphase mouse embryonic stem (ES) cells in synchrony with the incorporation of H3.3 during telomere replication at S phase. Moreover, we found that chromobox homolog 5 (CBX5) (also known as heterochromatin protein 1 alpha, or HP1 alpha) is also present at the telomeres in ES cells. We show by coimmunoprecipitation that this localization is dependent on the association of ATRX with histone H3.3, and that mutating the K4 residue of H3.3 significantly diminishes ATRX and H3.3 interaction. RNAi-knockdown of ATRX induces a telomere-dysfunction phenotype and significantly reduces CBX5 enrichment at the telomeres. These findings suggest a novel function of ATRX, working in conjunction with H3.3 and CBX5, as a key regulator of ES-cell telomere chromatin.


Subject(s)
DNA Helicases/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Pluripotent Stem Cells/metabolism , Telomere/metabolism , Adenosine Triphosphatases/chemistry , Animals , Chromatin/metabolism , Chromatin/physiology , Chromatin Assembly and Disassembly , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , DNA Helicases/genetics , DNA Replication/genetics , DNA Replication/physiology , Embryonic Stem Cells/metabolism , Genes , Histones/genetics , Humans , Intellectual Disability/genetics , Interphase/genetics , Mental Retardation, X-Linked/genetics , Mice , Mutation , Nuclear Proteins/genetics , X-linked Nuclear Protein , alpha-Thalassemia/genetics
14.
BMC Biochem ; 11: 50, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-21194474

ABSTRACT

BACKGROUND: Cell biologists face the need to rapidly analyse their proteins of interest in order to gain insight into their function. Often protein purification, cellular localisation and Western blot analyses can be multi-step processes, where protein is lost, activity is destroyed or effective antibodies have not yet been generated. AIM: To develop a method that simplifies the critical protein analytical steps of the laboratory researcher, leading to easy, efficient and rapid protein purification, cellular localisation and quantification. RESULTS: We have tagged the SMC2 subunit of the condensin complex with the Streptavidin-Binding Peptide (SBP), optimising and demonstrating the efficacious use of this tag for performing these protein analytical steps. Based on silver staining, and Western analysis, SBP delivered an outstanding specificity and purity of the condensin complex. We also developed a rapid and highly specific procedure to localise SBP-tagged proteins in cells in a single step procedure thus bypassing the need for using antibodies. Furthermore we have shown that the SBP tag can be used for isolating tagged proteins from chemically cross-linked cell populations for capturing DNA-protein interactions. CONCLUSIONS: The small 38-amino acid synthetic SBP offers the potential to successfully perform all four critical analytical procedures as a single step and should have a general utility for the study of many proteins and protein complexes.


Subject(s)
Adenosine Triphosphatases/metabolism , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/isolation & purification , Animals , Blotting, Western , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line, Tumor , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , Fluorescence , Humans , Microscopy, Fluorescence , Multiprotein Complexes/chemistry , Multiprotein Complexes/isolation & purification , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Streptavidin/chemistry , Streptavidin/metabolism
16.
Genome Res ; 19(12): 2202-13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19737860

ABSTRACT

The Y centromere sequence of house mouse, Mus musculus, remains unknown despite our otherwise significant knowledge of the genome sequence of this important mammalian model organism. Here, we report the complete molecular characterization of the C57BL/6J chromosome Y centromere, which comprises a highly diverged minor satellite-like sequence (designated Ymin) with higher-order repeat (HOR) sequence organization previously undescribed at mouse centromeres. The Ymin array is approximately 90 kb in length and resides within a single BAC clone that provides sequence information spanning an endogenous animal centromere for the first time. By exploiting direct patrilineal inheritance of the Y chromosome, we demonstrate stability of the Y centromere DNA structure spanning at least 175 inbred generations to beyond the time of domestication of the East Asian M.m. molossinus "fancy" mouse through which the Y chromosome was first introduced into the classical inbred laboratory mouse strains. Despite this stability, at least three unequal genetic exchange events have altered Ymin HOR unit length and sequence structure since divergence of the ancestral Mus musculus subspecies around 900,000 yr ago, with major turnover of the HOR arrays driving rapid divergence of sequence and higher-order structure at the mouse Y centromere. A comparative sequence analysis between the human and chimpanzee centromeres indicates a similar rapid divergence of the primate Y centromere. Our data point to a unique DNA sequence and organizational architecture for the mouse Y centromere that has evolved independently of all other mouse centromeres.


Subject(s)
Base Sequence , Biological Evolution , Centromere/genetics , DNA, Satellite/genetics , Y Chromosome/genetics , Animals , Chromosomes, Artificial, Bacterial , Female , Gene Library , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Pan troglodytes , Repetitive Sequences, Nucleic Acid , Sequence Analysis, DNA , Species Specificity , Time Factors
17.
Dev Cell ; 17(3): 305-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19758554

ABSTRACT

The conserved centromere protein C (CENP-C) is indispensable for kinetochore function. Yet its mechanism of action has remained elusive. In this issue of Developmental Cell, Tanaka et al. report that the fission yeast homolog, Cnp3, acts as a linker protein that fulfills a variety of different roles in the bi- and mono-orientation of chromosomes during mitosis and meiosis I.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Chromosomes/ultrastructure , Schizosaccharomyces/physiology , Centromere/ultrastructure , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Gene Expression Regulation, Fungal , Kinetochores/metabolism , Meiosis , Mitosis , Models, Biological , Models, Genetic , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism
18.
BMC Clin Pathol ; 9: 5, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19505339

ABSTRACT

BACKGROUND: Elevated levels of FMR1 mRNA in blood have been implicated in RNA toxicity associated with a number of clinical conditions. Due to the extensive inter-sample variation in the time lapse between the blood collection and RNA extraction in clinical practice, the resulting variation in mRNA quality significantly confounds mRNA analysis by real-time PCR. METHODS: Here, we developed an improved method to normalize for mRNA degradation in a sample set with large variation in rRNA quality, without sample omission. Initially, RNA samples were artificially degraded, and analyzed using capillary electrophoresis and real-time PCR standard curve method, with the aim of defining the best predictors of total RNA and mRNA degradation. RESULTS: We found that: (i) the 28S:18S ratio and RNA quality indicator (RQI) were good predictors of severe total RNA degradation, however, the greatest changes in the quantity of different mRNAs (FMR1, DNMT1, GUS, B2M and GAPDH) occurred during the early to moderate stages of degradation; (ii) chromatographic features for the 18S, 28S and the inter-peak region were the most reliable predictors of total RNA degradation, however their use for target gene normalization was inferior to internal control genes, of which GUS was the most appropriate. Using GUS for normalization, we examined in the whole blood the relationship between the FMR1 mRNA and CGG expansion in a non-coding portion of this gene, in a sample set (n = 30) with the large variation in rRNA quality. By combining FMR1 3' and 5' mRNA analyses the confounding impact of mRNA degradation on the correlation between FMR1 expression and CGG size was minimized, and the biological significance increased from p = 0.046 for the 5' FMR1 assay, to p = 0.018 for the combined FMR1 3' and 5' mRNA analysis. CONCLUSION: Our observations demonstrate that, through the use of an appropriate internal control and the direct analysis of multiple sites of target mRNA, samples that do not conform to the conventional rRNA criteria can still be utilized to obtain biologically/clinically relevant data. Although, this strategy clearly has application for improved assessment of FMR1 mRNA toxicity in blood, it may also have more general implications for gene expression studies in fresh and archival tissues.

20.
Genome Res ; 19(3): 404-14, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19196724

ABSTRACT

Little is known about the telomere chromatin dynamics of embryonic stem (ES) cell. Here, we demonstrate localization of histone H3.3 at interphase telomeres and enrichment of Ser31-phosphorylated H3.3 at metaphase telomeres in pluripotent mouse ES cells. Upon differentiation, telomeric H3.3S31P signal decreases, accompanied by increased association of heterochromatin repressive marks and decreased micrococcal nuclease sensitivity at the telomeres. H3.3 is recruited to the telomeres at late S/G2 phase, coinciding with telomere replication and processing. RNAi-depletion of H3.3 induces telomere-dysfunction phenotype, providing evidence for a role of H3.3 in the regulation of telomere chromatin integrity in ES cells. The distinctive changes in H3.3 distribution suggests the existence of a unique and functionally essential telomere chromatin in ES cells that undergoes dynamic differentiation-dependent remodeling during the process of differentiation.


Subject(s)
Chromatin/physiology , Embryonic Stem Cells/metabolism , Histones/metabolism , Telomere/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Child, Preschool , Chromatin/metabolism , DNA Replication/genetics , DNA Replication/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Histones/physiology , Humans , Interphase/genetics , Interphase/physiology , Mice , Mitosis/genetics , Mitosis/physiology , NIH 3T3 Cells , Phosphorylation , Protamine Kinase/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...