Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Physiol Biochem ; 52(4): 822-837, 2019.
Article in English | MEDLINE | ID: mdl-30946557

ABSTRACT

BACKGROUND/AIMS: Lung fibrosis is associated with lung tissue contraction due to abnormal accumulation of myofibroblasts, which aggressively promote the fibrotic process. Transforming growth factor (TGF)-ß signaling in fibroblasts promotes extracellular matrix (ECM) synthesis and fibroblast migration and differentiation into myofibroblasts. Inhibition of extracellular signal-regulated kinase (ERK)5 blocks lung fibroblast activation by suppressing TGF-ß signaling. Here, we examined the effects of an ERK5 inhibitor on TGF-ß1-induced fibrosis in lung fibroblasts. METHODS: The effects of ERK5 inhibition following TGF-ß1 exposure were evaluated in lung fibroblasts isolated from fibrotic human lung tissues. Fibroblast-mediated collagen gel contraction and fibroblast migration towards fibronectin were assessed. Phenotypic differences in fibrotic fibroblasts were examined using the cap analysis gene expression method for genome-wide quantification of promoter activity. RESULTS: TGF-ß1stimulated contraction of collagen gels, fibroblast migration, and α-smooth muscle actin and fibronectin expression, and Smad3 phosphorylation were increased in fibrotic fibroblasts as compared to normal lung fibroblasts. Treatment with the ERK5 inhibitor blocked these responses to a greater extent in fibroblasts from patients with usual interstitial pneumonia as compared to nonspecific interstitial pneumonia, independent of bone morphogenetic protein/Smad1 regulation. Moreover, 223 genes including fibulin-5 -which is involved in the TGF-ß1-ERK5 signaling network- were upregulated in fibrotic fibroblasts, and ECM regulation was found to be enriched in the Reactome analysis. CONCLUSION: ERK5 inhibition attenuated the high sensitivity of fibrotic fibroblasts to TGF-ß1/Smad3 signaling. Thus, the ERK5 pathway components and fibulin-5 are potential therapeutic targets to prevent lung fibrosis progression.


Subject(s)
Mitogen-Activated Protein Kinase 7/metabolism , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Actins/metabolism , Aged , Aniline Compounds/pharmacology , Biomarkers/metabolism , Cell Movement/drug effects , Chemotaxis/drug effects , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibronectins/metabolism , Humans , Indoles/pharmacology , Male , Middle Aged , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Pulmonary Fibrosis/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Smad3 Protein/metabolism , Up-Regulation/drug effects
2.
Neuron ; 101(3): 472-485.e7, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30638745

ABSTRACT

Minimal sets of transcription factors can directly reprogram somatic cells into neurons. However, epigenetic remodeling during neuronal reprogramming has not been well reconciled with transcriptional regulation. Here we show that NeuroD1 achieves direct neuronal conversion from mouse microglia both in vitro and in vivo. Exogenous NeuroD1 initially occupies closed chromatin regions associated with bivalent trimethylation of histone H3 at lysine 4 (H3K4me3) and H3K27me3 marks in microglia to induce neuronal gene expression. These regions are resolved to a monovalent H3K4me3 mark at later stages of reprogramming to establish the neuronal identity. Furthermore, the transcriptional repressors Scrt1 and Meis2 are induced as NeuroD1 target genes, resulting in a decrease in the expression of microglial genes. In parallel, the microglial epigenetic signature in promoter and enhancer regions is erased. These findings reveal NeuroD1 pioneering activity accompanied by global epigenetic remodeling for two sequential events: onset of neuronal property acquisition and loss of the microglial identity during reprogramming.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cellular Reprogramming , Epigenesis, Genetic , Microglia/cytology , Neurons/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Corpus Striatum/cytology , Female , HEK293 Cells , Histone Code , Histones/chemistry , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Microglia/metabolism , Neurons/metabolism
3.
Sci Rep ; 7(1): 14225, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29079795

ABSTRACT

Single cell transcriptome analysis of a cancer tissue can provide objective assessment of subtype population or the activation of each of various microenvironment component cells. In this study, we applied our newly developed technique of single cell analysis to the myometrial infiltration side (M-side) and the endometrial side (E-side) of a human endometrioid adenocarcinoma with squamous differentiation tissues. We also analyzed spherogenic cultures derived from the same tissue to identify putative regulators of stemness in vivo. Cancer cells in the E-side were highly malignant compared with those in the M-side. Many cells on the E-side were positive for spheroid-specific tumorigenesis-related markers including SOX2. In addition, there were higher numbers of epithelial-to-mesenchymal transition (EMT) cells in the E-side compared with the M-side. This study identified a site containing cells with high malignant potential such as EMT and cancer stem-like cells in cancer tissues. Finally, we demonstrate that established endometrioid adenocarcinoma subtype classifiers were variably expressed across individual cells within a tumor. Thus, such intratumoral heterogeneity may be related to prognostic implications.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Gene Expression Profiling , Single-Cell Analysis , Adenocarcinoma/diagnosis , Adenocarcinoma/immunology , Adult , Chemokines/genetics , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/immunology , Female , Humans , Middle Aged , Neoplasm Staging , Prognosis , Spheroids, Cellular/pathology
4.
PLoS One ; 5(10): e13284, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20967262

ABSTRACT

From the ENCODE project, it is realized that almost every base of the entire human genome is transcribed. One class of transcripts resulting from this arises from the conjoined gene, which is formed by combining the exons of two or more distinct (parent) genes lying on the same strand of a chromosome. Only a very limited number of such genes are known, and the definition and terminologies used for them are highly variable in the public databases. In this work, we have computationally identified and manually curated 751 conjoined genes (CGs) in the human genome that are supported by at least one mRNA or EST sequence available in the NCBI database. 353 representative CGs, of which 291 (82%) could be confirmed, were subjected to experimental validation using RT-PCR and sequencing methods. We speculate that these genes are arising out of novel functional requirements and are not merely artifacts of transcription, since more than 70% of them are conserved in other vertebrate genomes. The unique splicing patterns exhibited by CGs reveal their possible roles in protein evolution or gene regulation. Novel CGs, for which no transcript is available, could be identified in 80% of randomly selected potential CG forming regions, indicating that their formation is a routine process. Formation of CGs is not only limited to human, as we have also identified 270 CGs in mouse and 227 in drosophila using our approach. Additionally, we propose a novel mechanism for the formation of CGs. Finally, we developed a database, ConjoinG, which contains detailed information about all the CGs (800 in total) identified in the human genome. In summary, our findings reveal new insights about the functionality of CGs in terms of another possible mechanism for gene regulation and genomic evolution and the mechanism leading to their formation.


Subject(s)
Gene Expression Regulation , Animals , Chromosome Mapping , Databases, Genetic , Eukaryotic Cells , Exons , Expressed Sequence Tags , Genome, Human , Humans , Mice , Molecular Sequence Data , RNA Splicing , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
BMC Res Notes ; 1: 95, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18957096

ABSTRACT

BACKGROUND: SH-SY5Y cells exhibit a neuronal phenotype when treated with all-trans retinoic acid (RA), but the molecular mechanism of activation in the signalling pathway mediated by phosphatidylinositol 3-kinase (PI3K) is unclear. To investigate this mechanism, we compared the gene expression profiles in SK-N-SH cells and two subtypes of SH-SY5Y cells (SH-SY5Y-A and SH-SY5Y-E), each of which show a different phenotype during RA-mediated differentiation. FINDINGS: SH-SY5Y-A cells differentiated in the presence of RA, whereas RA-treated SH-SY5Y-E cells required additional treatment with brain-derived neurotrophic factor (BDNF) for full differentiation. After exposing cells to a PI3K inhibitor, LY294002, we identified 386 genes and categorised these genes into two clusters dependent on the PI3K signalling pathway during RA-mediated differentiation in SH-SY5Y-A cells. Transcriptional regulation of the gene cluster, including 158 neural genes, was greatly reduced in SK-N-SH cells and partially impaired in SH-SY5Y-E cells, which is consistent with a defect in the neuronal phenotype of these cells. Additional stimulation with BDNF induced a set of neural genes that were down-regulated in RA-treated SH-SY5Y-E cells but were abundant in differentiated SH-SY5Y-A cells. CONCLUSION: We identified gene clusters controlled by PI3K- and TRKB-mediated signalling pathways during the differentiation of two subtypes of SH-SY5Y cells. The TRKB-mediated bypass pathway compensates for impaired neural function generated by defects in several signalling pathways, including PI3K in SH-SY5Y-E cells. Our expression profiling data will be useful for further elucidation of the signal transduction-transcriptional network involving PI3K or TRKB.

6.
BMC Pharmacol ; 7: 10, 2007 Aug 20.
Article in English | MEDLINE | ID: mdl-17705877

ABSTRACT

BACKGROUND: Protein-protein interactions (PPIs) are challenging but attractive targets for small chemical drugs. Whole PPIs, called the 'interactome', have been emerged in several organisms, including human, based on the recent development of high-throughput screening (HTS) technologies. Individual PPIs have been targeted by small drug-like chemicals (SDCs), however, interactome data have not been fully utilized for exploring drug targets due to the lack of comprehensive methodology for utilizing these data. Here we propose an integrative in silico approach for discovering candidates for drug-targetable PPIs in interactome data. RESULTS: Our novel in silico screening system comprises three independent assessment procedures: i) detection of protein domains responsible for PPIs, ii) finding SDC-binding pockets on protein surfaces, and iii) evaluating similarities in the assignment of Gene Ontology (GO) terms between specific partner proteins. We discovered six candidates for drug-targetable PPIs by applying our in silico approach to original human PPI data composed of 770 binary interactions produced by our HTS yeast two-hybrid (HTS-Y2H) assays. Among them, we further examined two candidates, RXRA/NRIP1 and CDK2/CDKN1A, with respect to their biological roles, PPI network around each candidate, and tertiary structures of the interacting domains. CONCLUSION: An integrative in silico approach for discovering candidates for drug-targetable PPIs was applied to original human PPIs data. The system excludes false positive interactions and selects reliable PPIs as drug targets. Its effectiveness was demonstrated by the discovery of the six promising candidate target PPIs. Inhibition or stabilization of the two interactions may have potential therapeutic effects against human diseases.


Subject(s)
Drug Delivery Systems/methods , Pharmaceutical Preparations/metabolism , Protein Interaction Mapping/methods , Drug Evaluation, Preclinical/methods , Humans , Pharmaceutical Preparations/chemistry , Protein Binding/physiology , Protein Structure, Secondary/physiology , Technology, Pharmaceutical/methods
7.
Proc Natl Acad Sci U S A ; 103(21): 8131-6, 2006 May 23.
Article in English | MEDLINE | ID: mdl-16698922

ABSTRACT

Cdc7 kinase, conserved through evolution, is known to be essential for mitotic DNA replication. The role of Cdc7 in meiotic recombination was suggested in Saccharomyces cerevisiae, but its precise role has not been addressed. Here, we report that Hsk1, the Cdc7-related kinase in Schizosaccharomyces pombe, plays a crucial role during meiosis. In a hsk1 temperature-sensitive strain (hsk1-89), meiosis is arrested with one nucleus state before meiosis I in most of the cells and meiotic recombination frequency is reduced by one order of magnitude, whereas premeiotic DNA replication is delayed but is apparently completed. Strikingly, formation of meiotic dsDNA breaks (DSBs) are largely impaired in the mutant, and Hsk1 kinase activity is essential for these processes. Deletion of all three checkpoint kinases, namely Cds1, Chk1, and Mek1, does not restore DSB formation, meiosis, or Cdc2 activation, which is suppressed in hsk1-89, suggesting that these aberrations are not caused by known checkpoint pathways but that Hsk1 may regulate DSB formation and meiosis. Whereas transcriptional induction of some rec genes and horsetail movement are normal, chromatin remodeling at ade6-M26, a recombination hotspot, which is prerequisite for subsequent DSB formation at this locus, is not observed in hsk1-89. These results indicate unique and essential roles of Hsk1 kinase in the initiation of meiotic recombination and meiosis.


Subject(s)
Cell Cycle Proteins/physiology , Meiosis , Protein Serine-Threonine Kinases/physiology , Schizosaccharomyces pombe Proteins/physiology , Bromodeoxyuridine/pharmacology , Chromatin/chemistry , DNA Damage , Mutation , Recombination, Genetic , Schizosaccharomyces , Schizosaccharomyces pombe Proteins/chemistry , Temperature , Transcription, Genetic
8.
Nature ; 440(7083): 497-500, 2006 Mar 23.
Article in English | MEDLINE | ID: mdl-16554811

ABSTRACT

Chromosome 11, although average in size, is one of the most gene- and disease-rich chromosomes in the human genome. Initial gene annotation indicates an average gene density of 11.6 genes per megabase, including 1,524 protein-coding genes, some of which were identified using novel methods, and 765 pseudogenes. One-quarter of the protein-coding genes shows overlap with other genes. Of the 856 olfactory receptor genes in the human genome, more than 40% are located in 28 single- and multi-gene clusters along this chromosome. Out of the 171 disorders currently attributed to the chromosome, 86 remain for which the underlying molecular basis is not yet known, including several mendelian traits, cancer and susceptibility loci. The high-quality data presented here--nearly 134.5 million base pairs representing 99.8% coverage of the euchromatic sequence--provide scientists with a solid foundation for understanding the genetic basis of these disorders and other biological phenomena.


Subject(s)
Chromosomes, Human, Pair 11 , Sequence Analysis, DNA , DNA , Gene Expression , Genes , Humans , Molecular Sequence Data , Physical Chromosome Mapping , Receptors, Odorant/genetics
10.
DNA Res ; 9(3): 89-97, 2002 Jun 30.
Article in English | MEDLINE | ID: mdl-12168953

ABSTRACT

We recently helped to complete the sequence of human chromosome 21 at a very high level of accuracy. Using this sequence we identified two novel genes, designated DSCR9 and DSCR10, in the so-called Down Syndrome Critical Region (DSCR) by computational gene prediction and subsequent cDNA cloning. Both DSCR9 and DSCR10 are expressed preferentially in testis and encode functionally unknown proteins with 149 and 87 amino acid residues, respectively. Zoo blot analysis suggested that both genes are exclusive to primate genomes such as chimpanzee, gorilla, orangutan, crab-eating monkey and African green monkey but are not present in other non-primate mammals including mouse, dog, cat, and chicken. Comparative genomic sequence analysis of DSCR9 and DSCR10 with the corresponding mouse syntenic region confirmed the lack of these genes in the mouse. These results strongly suggest that DSCR9 and DSCR10 have emerged as a new class of gene in the primate lineage during evolution.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Genes , Primates/genetics , Amino Acid Sequence , Animals , Base Sequence , Cats , Cattle , Cercopithecinae , Chickens , Cloning, Molecular , Dogs , Humans , Male , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Swine , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...