Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Geriatr Nurs ; 42(3): 650-656, 2021.
Article in English | MEDLINE | ID: mdl-33823423

ABSTRACT

The purpose of this study was to determine the effect of a group music intervention with rhythmic exercises program on cognitive function and mental health outcomes among Korean nursing home residents . A randomized controlled study design was employed, in which the experimental group (n = 20) received a biweekly group music intervention with rhythmic exercises program and a regular activities program for 12 weeks, and the control group (n = 20) received only a regular activities program provided by the nursing home. We measured cognitive function and mental health outcomes using the Mini-Mental State Examination-Korean Version (MMSE-K), Geriatric Depression Scale Short Form-Korean Version (GDSSF-K), Geriatric Anxiety Inventory-Korean Version (GAI-K), and Life Satisfaction Scale. ANCOVA was performed to examine the effect of the group music intervention on the study variables. The results suggested that the group music intervention with rhythmic exercises program improved cognitive function, depression, anxiety, and life satisfaction.


Subject(s)
Music Therapy , Aged , Cognition , Humans , Nursing Homes , Outcome Assessment, Health Care , Pilot Projects
2.
Mol Cancer Res ; 16(3): 439-452, 2018 03.
Article in English | MEDLINE | ID: mdl-29330289

ABSTRACT

The tumor suppressor BRCA1 regulates the DNA damage response (DDR) and other processes that remain incompletely defined. Among these, BRCA1 heterodimerizes with BARD1 to ubiquitylate targets via its N-terminal E3 ligase activity. Here, it is demonstrated that BRCA1 promotes oxidative metabolism by degrading Oct1 (POU2F1), a transcription factor with proglycolytic and tumorigenic effects. BRCA1 E3 ubiquitin ligase mutation skews cells toward a glycolytic metabolic profile while elevating Oct1 protein. CRISPR-mediated Oct1 deletion reverts the glycolytic phenotype. RNA sequencing (RNAseq) confirms deregulation of metabolic genes downstream of Oct1. BRCA1 mediates Oct1 ubiquitylation and degradation, and mutation of two ubiquitylated Oct1 lysines insulates the protein against BRCA1-mediated destabilization. Oct1 deletion in MCF-7 breast cancer cells does not perturb growth in standard culture, but inhibits growth in soft agar and xenograft assays. In primary breast cancer clinical specimens, Oct1 protein levels correlate positively with tumor aggressiveness and inversely with BRCA1. These results identify BRCA1 as an Oct1 ubiquitin ligase that catalyzes Oct1 degradation to promote oxidative metabolism and restrict tumorigenicity. Mol Cancer Res; 16(3); 439-52. ©2018 AACR.


Subject(s)
BRCA1 Protein/metabolism , Octamer Transcription Factor-1/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , BRCA1 Protein/genetics , Carbohydrate Metabolism , Cell Line, Tumor , Female , Heterografts , Humans , MCF-7 Cells , Metabolomics/methods , Mice , Mice, Inbred NOD , Mice, SCID
3.
Elife ; 62017 05 24.
Article in English | MEDLINE | ID: mdl-28537559

ABSTRACT

Embryonic stem cells co-express Oct4 and Oct1, a related protein with similar DNA-binding specificity. To study the role of Oct1 in ESC pluripotency and transcriptional control, we constructed germline and inducible-conditional Oct1-deficient ESC lines. ESCs lacking Oct1 show normal appearance, self-renewal and growth but manifest defects upon differentiation. They fail to form beating cardiomyocytes, generate neurons poorly, form small, poorly differentiated teratomas, and cannot generate chimeric mice. Upon RA-mediated differentiation, Oct1-deficient cells induce lineage-appropriate developmentally poised genes poorly while lineage-inappropriate genes, including extra-embryonic genes, are aberrantly expressed. In ESCs, Oct1 co-occupies a specific set of targets with Oct4, but does not occupy differentially expressed developmental targets. Instead, Oct1 occupies these targets as cells differentiate and Oct4 declines. These results identify a dynamic interplay between Oct1 and Oct4, in particular during the critical window immediately after loss of pluripotency when cells make the earliest developmental fate decisions.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/physiology , Octamer Transcription Factor-1/metabolism , Transcription, Genetic , Animals , Mice , Octamer Transcription Factor-3/metabolism
4.
Proc Natl Acad Sci U S A ; 112(31): E4236-45, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26199412

ABSTRACT

Dioxygenases of the TET (Ten-Eleven Translocation) family produce oxidized methylcytosines, intermediates in DNA demethylation, as well as new epigenetic marks. Here we show data suggesting that TET proteins maintain the consistency of gene transcription. Embryos lacking Tet1 and Tet3 (Tet1/3 DKO) displayed a strong loss of 5-hydroxymethylcytosine (5hmC) and a concurrent increase in 5-methylcytosine (5mC) at the eight-cell stage. Single cells from eight-cell embryos and individual embryonic day 3.5 blastocysts showed unexpectedly variable gene expression compared with controls, and this variability correlated in blastocysts with variably increased 5mC/5hmC in gene bodies and repetitive elements. Despite the variability, genes encoding regulators of cholesterol biosynthesis were reproducibly down-regulated in Tet1/3 DKO blastocysts, resulting in a characteristic phenotype of holoprosencephaly in the few embryos that survived to later stages. Thus, TET enzymes and DNA cytosine modifications could directly or indirectly modulate transcriptional noise, resulting in the selective susceptibility of certain intracellular pathways to regulation by TET proteins.


Subject(s)
5-Methylcytosine/metabolism , DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Proto-Oncogene Proteins/metabolism , Transcriptome/genetics , Animals , Biomarkers/metabolism , Blastocyst/metabolism , Blastomeres/metabolism , Blastomeres/pathology , Cell Lineage , Cholesterol/biosynthesis , DNA/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dioxygenases , Down-Regulation/genetics , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Mammalian/pathology , Genomic Imprinting , Hedgehog Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Repetitive Sequences, Nucleic Acid/genetics , Sequence Analysis, RNA , Signal Transduction/genetics
5.
Blood Res ; 50(2): 97-102, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26157779

ABSTRACT

BACKGROUND: Few clinical studies have clarified the prognostic factors that affect clinical outcomes for patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) after immunochemotherapy. METHODS: A total of 158 patients with relapsed or refractory DLBCL were enrolled. All patients underwent positron emission tomography/computed tomography (PET/CT) before and after salvage therapy. All enrolled patients previously received the ifosfamide, carboplatin, and etoposide regimen. Clinical outcomes were compared according to several factors (age ≥ 65 years, low age-adjusted International Prognostic Index [aa-IPI], maximum standardized uptake value [SUVmax] <6.0 on PET/CT, time to relapse ≥12 months, complete response after salvage therapy). A low aa-IPI, SUVmax <6.0, and time to relapse ≥ 12 months were independent prognostic factors for survival. RESULTS: In univariate analysis and multivariate analysis, SUVmax below 6.0 (P<0.001 for progression-free survival (PFS), P<0.001 for overall survival (OS)) and low aa-IPI (P<0.001 for PFS, P<0.001 for OS) were independent prognostic factors associated with favorable outcome. CONCLUSION: The aa-IPI and initial SUVmax were powerful prognostic factors in patients with relapsed or refractory DLBCL.

6.
Proc Natl Acad Sci U S A ; 111(4): 1361-6, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24474761

ABSTRACT

Dioxygenases of the Ten-Eleven Translocation (TET) family are 5-methylcytosine oxidases that convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) and further oxidation products in DNA. We show that Tet1 and Tet2 have distinct roles in regulating 5hmC in mouse embryonic stem cells (mESC). Tet1 depletion diminishes 5hmC levels at transcription start sites (TSS), whereas Tet2 depletion is predominantly associated with decreased 5hmC in gene bodies. Enrichment of 5hmC is observed at the boundaries of exons that are highly expressed, and Tet2 depletion results in substantial loss of 5hmC at these boundaries. In contrast, at promoter/TSS regions, Tet2 depletion results in increased 5hmC, potentially because of the redundant activity of Tet1. Together, the data point to a complex interplay between Tet1 and Tet2 in mESC, and to distinct roles for these two proteins in regulating promoter, exon, and polyadenylation site usage in cells.


Subject(s)
DNA-Binding Proteins/physiology , Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins/physiology , Animals , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , Exons , Mice , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Transcription, Genetic
7.
FASEB J ; 27(7): 2807-17, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23580612

ABSTRACT

The Oct1 transcription factor is a potent regulator of stress responses, metabolism, and tumorigenicity. Although Oct1 is regulated by phosphorylation and ubiquitination, the presence and importance of other modifications is unknown. Here we show that Oct1 is modified by O-linked ß-N-acetylglucosamine (O-GlcNAc) moieties. We map two sites of O-GlcNAcylation at positions T255 and S728 within human Oct1. Under anchorage-independent overgrowth conditions, Oct1 associates 3-fold more strongly with the Gadd45a promoter and mediates transcriptional repression. Increased binding correlates with quantitative reductions in Oct1 nuclear periphery-associated puncta, and a reduced association with lamin B1. The O-GlcNAc modification sites are important for both Gadd45a repression and anchorage-independent survival. In contrast to chronic overgrowth conditions, following acute nutrient starvation Oct1 mediates Gadd45a activation. The O-GlcNAc sites are also important for Gadd45a activation under these conditions. We also, for the first time, identify specific Oct1 ubiquitination sites. The findings suggest that Oct1 integrates metabolic and stress signals via O-GlcNAc modification to regulate target gene activity.


Subject(s)
Acetylglucosamine/metabolism , Transcription Factor Brn-3A/metabolism , Transcriptional Activation , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Cycle Proteins/genetics , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoblotting , Mice , Mice, Knockout , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor Brn-3A/genetics
8.
Cell Res ; 23(1): 6-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22868271

ABSTRACT

In zygotes, a global loss of DNA methylation occurs selectively in the paternal pronucleus before the first cell division, concomitantly with the appearance of modified forms of 5-methylcytosine. The adjacent maternal pronucleus and certain paternally-imprinted loci are protected from this process. Nakamura et al. recently clarified the molecular mechanism involved: PGC7/Stella/Dppa3 binds to dimethylated histone 3 lysine 9 (H3K9me2), thereby blocking the activity of the Tet3 methylcytosine oxidase in the maternal genome as well as at certain imprinted loci in the paternal genome.


Subject(s)
DNA Methylation , DNA-Binding Proteins/metabolism , Histones/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Zygote/metabolism , Animals , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins/genetics , Dioxygenases , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development , Female , Histones/genetics , Male , Mice , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Zygote/cytology
9.
PLoS Genet ; 8(11): e1003048, 2012.
Article in English | MEDLINE | ID: mdl-23144633

ABSTRACT

Defining master transcription factors governing somatic and cancer stem cell identity is an important goal. Here we show that the Oct4 paralog Oct1, a transcription factor implicated in stress responses, metabolic control, and poised transcription states, regulates normal and pathologic stem cell function. Oct1(HI) cells in the colon and small intestine co-express known stem cell markers. In primary malignant tissue, high Oct1 protein but not mRNA levels strongly correlate with the frequency of CD24(LO)CD44(HI) cancer-initiating cells. Reducing Oct1 expression via RNAi reduces the proportion of ALDH(HI) and dye efflux(HI) cells, and increasing Oct1 increases the proportion of ALDH(HI) cells. Normal ALDH(HI) cells harbor elevated Oct1 protein but not mRNA levels. Functionally, we show that Oct1 promotes tumor engraftment frequency and promotes hematopoietic stem cell engraftment potential in competitive and serial transplants. In addition to previously described Oct1 transcriptional targets, we identify four Oct1 targets associated with the stem cell phenotype. Cumulatively, the data indicate that Oct1 regulates normal and cancer stem cell function.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells , Organic Cation Transporter 1 , Stem Cells , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Biomarkers/metabolism , CD24 Antigen/metabolism , Colon/cytology , Colon/metabolism , HeLa Cells , Humans , Hyaluronan Receptors/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Organic Cation Transporter 1/genetics , Organic Cation Transporter 1/metabolism , Phenotype , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/metabolism
10.
PLoS One ; 6(8): e23872, 2011.
Article in English | MEDLINE | ID: mdl-21897860

ABSTRACT

BACKGROUND: Transcription factor Oct1 regulates multiple cellular processes. It is known to be phosphorylated during the cell cycle and by stress, however the upstream kinases and downstream consequences are not well understood. One of these modified forms, phosphorylated at S335, lacks the ability to bind DNA. Other modification states besides phosphorylation have not been described. METHODOLOGY/PRINCIPAL FINDINGS: We show that Oct1 is phosphorylated at S335 in the Oct1 DNA binding domain during M-phase by the NIMA-related kinase Nek6. Phospho-Oct1 is also ubiquitinated. Phosphorylation excludes Oct1 from mitotic chromatin. Instead, Oct1(pS335) concentrates at centrosomes, mitotic spindle poles, kinetochores and the midbody. Oct1 siRNA knockdown diminishes the signal at these locations. Both Oct1 ablation and overexpression result in abnormal mitoses. S335 is important for the overexpression phenotype, implicating this residue in mitotic regulation. Oct1 depletion causes defects in spindle morphogenesis in Xenopus egg extracts, establishing a mitosis-specific function of Oct1. Oct1 colocalizes with lamin B1 at the spindle poles and midbody. At the midbody, both proteins are mutually required to correctly localize the other. We show that phospho-Oct1 is modified late in mitosis by non-canonical K11-linked polyubiquitin chains. Ubiquitination requires the anaphase-promoting complex, and we further show that the anaphase-promoting complex large subunit APC1 and Oct1(pS335) interact. CONCLUSIONS/SIGNIFICANCE: These findings reveal mechanistic coupling between Oct1 phosphorylation and ubquitination during mitotic progression, and a role for Oct1 in mitosis.


Subject(s)
Mitosis , Octamer Transcription Factor-1/metabolism , Ubiquitination , Animals , HeLa Cells , Humans , Kinetochores/metabolism , Lamin Type B/metabolism , Mice , NIMA-Related Kinases , Octamer Transcription Factor-1/chemistry , Octamer Transcription Factor-1/deficiency , Organelles/metabolism , Phosphorylation , Polyubiquitin/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Serine/metabolism
11.
Genome Res ; 21(7): 1055-64, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21527551

ABSTRACT

The pluripotency control regions (PluCRs) are defined as genomic regions that are bound by POU5F1, SOX2, and NANOG in vivo. We utilized a high-throughput binding assay to record more than 270,000 different DNA/protein binding measurements along incrementally tiled windows of DNA within these PluCRs. This high-resolution binding map is then used to systematically define the context of POU factor binding, and reveals patterns of cooperativity and competition in the pluripotency network. The most prominent pattern is a pervasive binding competition between POU5F1 and the forkhead transcription factors. Like many transcription factors, POU5F1 is co-expressed with a paralog, POU2F1, that shares an apparently identical binding specificity. By analyzing thousands of binding measurements, we discover context effects that discriminate POU2F1 from POU5F1 binding. Proximal NANOG binding promotes POU5F1 binding, whereas nearby SOX2 binding favors POU2F1. We demonstrate by cross-species comparison and by chromatin immunoprecipitation (ChIP) that the contextual sequence determinants learned in vitro are sufficient to predict POU2F1 binding in vivo.


Subject(s)
Genome , Homeodomain Proteins/genetics , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-3/genetics , Animals , Binding Sites/genetics , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Chromosome Mapping , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation, Developmental , High-Throughput Nucleotide Sequencing , Homeodomain Proteins/metabolism , Humans , Octamer Transcription Factor-1/metabolism , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic , Protein Binding/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
12.
J Biol Chem ; 286(1): 450-9, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21051540

ABSTRACT

Little is known regarding how the Oct1 transcription factor regulates target gene expression. Using murine fibroblasts and two target genes, Polr2a and Ahcy, we show that Oct1 recruits the Jmjd1a/KDM3A lysine demethylase to catalyze the removal of the inhibitory histone H3K9 dimethyl mark and block repression. Using purified murine T cells and the Il2 target locus, and a colon cancer cell line and the Cdx2 target locus, we show that Oct1 recruits the NuRD chromatin-remodeling complex to promote a repressed state, but in a regulated manner can switch to a different capacity and mediate Jmjd1a recruitment to block repression. These findings indicate that Oct1 maintains repression through a mechanism involving NuRD and maintains poised gene expression states through an antirepression mechanism involving Jmjd1a. We propose that, rather than acting as a primary trigger of gene activation or repression, Oct1 is a switchable stabilizer of repressed and inducible states.


Subject(s)
Organic Cation Transporter 1/metabolism , Transcription, Genetic , Animals , Base Sequence , CDX2 Transcription Factor , Cell Line, Tumor , Fibroblasts/metabolism , Genetic Loci/genetics , Homeodomain Proteins/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects
13.
Trends Biochem Sci ; 34(10): 491-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19733480

ABSTRACT

It is a classic story of two related transcription factors. Oct4 is a potent regulator of pluripotency during early mammalian embryonic development, and is notable for its ability to convert adult somatic cells to pluripotency. The widely expressed Oct1 protein shares significant homology with Oct4, binds to the same sequences, regulates common target genes, and shares common modes of upstream regulation, including the ability to respond to cellular stress. Both proteins are also associated with malignancy, yet Oct1 cannot substitute for Oct4 in the generation of pluripotency. The molecular underpinnings of these phenomena are emerging, as are the consequences for adult stem cells and cancer, and thereby hangs a tale.


Subject(s)
Energy Metabolism , Neoplasms/metabolism , Octamer Transcription Factor-1/metabolism , Octamer Transcription Factor-3/metabolism , Stem Cells/physiology , Stress, Physiological , Animals , Cell Differentiation/physiology , Humans , Neoplasms/genetics , Octamer Transcription Factor-1/classification , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-3/classification , Octamer Transcription Factor-3/genetics , Phylogeny
14.
Genes Dev ; 23(2): 208-22, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19171782

ABSTRACT

Oct1 and Oct4 are homologous transcription factors with similar DNA-binding specificities. Here we show that Oct1 is dynamically phosphorylated in vivo following exposure of cells to oxidative and genotoxic stress. We further show that stress regulates the selectivity of both proteins for specific DNA sequences. Mutation of conserved phosphorylation target DNA-binding domain residues in Oct1, and Oct4 confirms their role in regulating binding selectivity. Using chromatin immunoprecipitation, we show that association of Oct4 and Oct1 with a distinct group of in vivo targets is inducible by stress, and that Oct1 is essential for a normal post-stress transcriptional response. Finally, using an unbiased Oct1 target screen we identify a large number of genes targeted by Oct1 specifically under conditions of stress, and show that several of these inducible Oct1 targets are also inducibly bound by Oct4 in embryonic stem cells following stress exposure.


Subject(s)
DNA Damage/physiology , Gene Expression Regulation , Octamer Transcription Factor-1/metabolism , Octamer Transcription Factor-3/metabolism , Oxidative Stress/physiology , Amino Acid Sequence , Animals , Dimerization , HeLa Cells , Humans , Inverted Repeat Sequences/genetics , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Octamer Transcription Factor-1/chemistry , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/genetics , Phosphorylation , Protein Binding , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL