Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Cell Rep ; 39(4): 110721, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35476996

ABSTRACT

The resistance to transcription factor-mediated reprogramming into pluripotent stem cells is one of the distinctive features of cancer cells. Here we dissect the profiles of reprogramming factor binding and the subsequent transcriptional response in cancer cells to reveal its underlying mechanisms. Using clear cell sarcomas (CCSs), we show that the driver oncogene EWS/ATF1 misdirects the reprogramming factors to cancer-specific enhancers and thereby impairs the transcriptional response toward pluripotency that is otherwise provoked. Sensitization to the reprogramming cue is observed in other cancer types when the corresponding oncogenic signals are pharmacologically inhibited. Exploiting this oncogene dependence of the transcriptional "stiffness," we identify mTOR signaling pathways downstream of EWS/ATF1 and discover that inhibiting mTOR activity substantially attenuates the propagation of CCS cells in vitro and in vivo. Our results demonstrate that the early transcriptional response to cell fate perturbations can be a faithful readout to identify effective therapeutics targets in cancer cells.


Subject(s)
Oncogenes , Sarcoma, Clear Cell , Humans , Sarcoma, Clear Cell/genetics , Signal Transduction , TOR Serine-Threonine Kinases , Transcription Factors/genetics
2.
Nat Metab ; 4(2): 254-268, 2022 02.
Article in English | MEDLINE | ID: mdl-35145326

ABSTRACT

ß cells have a limited capacity for regeneration, which predisposes towards diabetes. Here, we show that, of the MYC family members, Mycl plays a key role in proliferation of pancreatic endocrine cells. Genetic ablation of Mycl causes a reduction in the proliferation of pancreatic endocrine cells in neonatal mice. By contrast, the expression of Mycl in adult mice stimulates the proliferation of ß and α cells, and the cells persist after withdrawal of Mycl expression. A subset of the expanded α cells give rise to insulin-producing cells after this withdrawal. Transient Mycl expression in vivo is sufficient to normalize the hyperglycaemia of diabetic mice. In vitro expression of Mycl similarly provokes active replication in islet cells, even in those from aged mice. Finally, we show that MYCL stimulates the division of human adult cadaveric islet cells. Our results demonstrate that the induction of Mycl alone expands the functional ß-cell population, which may provide a regenerative strategy for ß cells.


Subject(s)
Diabetes Mellitus, Experimental , Glucagon-Secreting Cells , Insulin-Secreting Cells , Islets of Langerhans , Animals , Glucagon-Secreting Cells/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Pancreatic Hormones/metabolism
3.
Biochem Biophys Res Commun ; 599: 43-50, 2022 04 09.
Article in English | MEDLINE | ID: mdl-35168063

ABSTRACT

The cyclin-dependent kinase inhibitor p16Ink4a plays a central role in cellular senescence in vitro. Although previous studies suggested cellular senescence is integrated in the systemic mechanisms of organismal aging, the localization and the dynamics of p16Ink4a in tissues remain poorly understood, which hinders uncovering the role of p16Ink4a under the in vivo context. One of the reasons is due to the lack of reliable reagents; as we also demonstrate here that commonly used antibodies raised against human p16INK4A barely recognize its murine ortholog. Here we generated a mouse model, in which the endogenous p16Ink4a is HA-tagged at its N-terminus, to explore the protein expression of p16Ink4a at the organismal level. p16Ink4a was induced at the protein level along the course of senescence in primary embryonic fibroblasts derived from the mice, consistently to its transcriptional level. Remarkably, however, p16Ink4a was not detected in the tissues of the mice exposed to pro-senescence conditions including genotoxic stress and activation of oncogenic signaling pathways, indicating that there is only subtle p16Ink4a proteins induced. These results in our mouse model highlight the need for caution in evaluating p16Ink4a protein expression in vivo.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Animals , Cross Reactions , Cyclin-Dependent Kinase Inhibitor p16/immunology , DNA Damage , Exons , Liver/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , NIH 3T3 Cells
4.
Nat Commun ; 12(1): 5041, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34413299

ABSTRACT

In vivo reprogramming provokes a wide range of cell fate conversion. Here, we discover that in vivo induction of higher levels of OSKM in mouse somatic cells leads to increased expression of primordial germ cell (PGC)-related genes and provokes genome-wide erasure of genomic imprinting, which takes place exclusively in PGCs. Moreover, the in vivo OSKM reprogramming results in development of cancer that resembles human germ cell tumors. Like a subgroup of germ cell tumors, propagated tumor cells can differentiate into trophoblasts. Moreover, these tumor cells give rise to induced pluripotent stem cells (iPSCs) with expanded differentiation potential into trophoblasts. Remarkably, the tumor-derived iPSCs are able to contribute to non-neoplastic somatic cells in adult mice. Mechanistically, DMRT1, which is expressed in PGCs, drives the reprogramming and propagation of the tumor cells in vivo. Furthermore, the DMRT1-related epigenetic landscape is associated with trophoblast competence of the reprogrammed cells and provides a therapeutic target for germ cell tumors. These results reveal an unappreciated route for somatic cell reprogramming and underscore the impact of reprogramming in development of germ cell tumors.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms/pathology , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Cellular Reprogramming/physiology , Epigenesis, Genetic , Female , Genomic Imprinting , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred ICR , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Transcription Factors/genetics
5.
J Reprod Dev ; 66(5): 459-467, 2020 Oct 13.
Article in English | MEDLINE | ID: mdl-32624547

ABSTRACT

PTBP1, a well-conserved RNA-binding protein, regulates cellular development by tuning posttranscriptional mRNA modification such as alternative splicing (AS) or mRNA stabilization. We previously revealed that the loss of Ptbp1 in spermatogonia causes the dysregulation of spermatogenesis, but the molecular mechanisms by which PTBP1 regulates spermatogonium homeostasis are unclear. In this study, changes of AS or transcriptome in Ptbp1-knockout (KO) germline stem cells (GSC), an in vitro model of proliferating spermatogonia, was determined by next generation sequencing. We identified more than 200 differentially expressed genes, as well as 85 genes with altered AS due to the loss of PTBP1. Surprisingly, no differentially expressed genes overlapped with different AS genes in Ptbp1-KO GSC. In addition, we observed that the mRNA expression of Nanos3, an essential gene for normal spermatogenesis, was significantly decreased in Ptbp1-KO spermatogonia. We also revealed that PTBP1 protein binds to Nanos3 mRNA in spermatogonia. Furthermore, Nanos3+/-;Ptbp1+/- mice exhibited abnormal spermatogenesis, which resembled the effects of germ cell-specific Ptbp1 KO, whereas no significant abnormality was observed in mice heterozygous for either gene alone. These data implied that PTBP1 regulates alternative splicing and transcriptome in spermatogonia under different molecular pathways, and contributes spermatogenesis, at least in part, in concert with NANOS3.


Subject(s)
Alternative Splicing , Gene Expression Regulation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , RNA-Binding Proteins/metabolism , Spermatogenesis/physiology , Spermatogonia/metabolism , Animals , Gene Deletion , Genes, Regulator , Germ Cells/cytology , Heterozygote , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA-Binding Proteins/genetics , RNA-Seq , Testis/metabolism , Transcriptome
6.
Nat Commun ; 11(1): 3199, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32581223

ABSTRACT

De novo establishment of DNA methylation is accomplished by DNMT3A and DNMT3B. Here, we analyze de novo DNA methylation in mouse embryonic fibroblasts (2i-MEFs) derived from DNA-hypomethylated 2i/L ES cells with genetic ablation of Dnmt3a or Dnmt3b. We identify 355 and 333 uniquely unmethylated genes in Dnmt3a and Dnmt3b knockout (KO) 2i-MEFs, respectively. We find that Dnmt3a is exclusively required for de novo methylation at both TSS regions and gene bodies of Polycomb group (PcG) target developmental genes, while Dnmt3b has a dominant role on the X chromosome. Consistent with this, tissue-specific DNA methylation at PcG target genes is substantially reduced in Dnmt3a KO embryos. Finally, we find that human patients with DNMT3 mutations exhibit reduced DNA methylation at regions that are hypomethylated in Dnmt3 KO 2i-MEFs. In conclusion, here we report a set of unique de novo DNA methylation target sites for both DNMT3 enzymes during mammalian development that overlap with hypomethylated sites in human patients.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Animals , Cell Differentiation/genetics , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Epigenetic Repression/genetics , Female , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mutation , Organ Specificity , Polycomb-Group Proteins , Transcription Initiation Site , DNA Methyltransferase 3B
7.
Nat Commun ; 10(1): 3999, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31488818

ABSTRACT

Clear cell sarcoma (CCS) is a rare soft tissue sarcoma caused by the EWS/ATF1 fusion gene. Here, we established induced pluripotent stem cells (iPSCs) from EWS/ATF1-controllable murine CCS cells harboring sarcoma-associated genetic abnormalities. Sarcoma-iPSC mice develop secondary sarcomas immediately after EWS/ATF1 induction, but only in soft tissue. EWS/ATF1 expression induces oncogene-induced senescence in most cell types in sarcoma-iPSC mice but prevents it in sarcoma cells. We identify Tppp3-expressing cells in peripheral nerves as a cell-of-origin for these sarcomas. We show cell type-specific recruitment of EWS/ATF1 to enhancer regions in CCS cells. Finally, epigenetic silencing at these enhancers induces senescence and inhibits CCS cell growth through altered EWS/ATF1 binding. Together, we propose that distinct responses to premature senescence are the basis for the cell type-specificity of cancer development.


Subject(s)
Activating Transcription Factor 1/genetics , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Clear Cell/genetics , Animals , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/metabolism , Disease Models, Animal , Exome/genetics , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Neoplasms, Experimental , Nervous System , S100 Calcium Binding Protein beta Subunit/genetics , Sarcoma, Clear Cell/pathology , Transcriptome
8.
Stem Cell Reports ; 12(5): 1113-1128, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31056481

ABSTRACT

CpG islands (CGIs) including those at imprinting control regions (ICRs) are protected from de novo methylation in somatic cells. However, many cancers often exhibit CGI hypermethylation, implying that the machinery is impaired in cancer cells. Here, we conducted a comprehensive analysis of CGI methylation during somatic cell reprogramming. Although most CGIs remain hypomethylated, a small subset of CGIs, particularly at several ICRs, was often de novo methylated in reprogrammed pluripotent stem cells (PSCs). Such de novo ICR methylation was linked with the silencing of reprogramming factors, which occurs at a late stage of reprogramming. The ICR-preferred CGI hypermethylation was similarly observed in human PSCs. Mechanistically, ablation of Dnmt3a prevented PSCs from de novo ICR methylation. Notably, the ICR-preferred CGI hypermethylation was observed in pediatric cancers, while adult cancers exhibit genome-wide CGI hypermethylation. These results may have important implications in the pathogenesis of pediatric cancers and the application of PSCs.


Subject(s)
Cellular Reprogramming/genetics , DNA Methylation/genetics , Genomic Imprinting/genetics , Pluripotent Stem Cells/metabolism , Adult , Animals , Cells, Cultured , CpG Islands/genetics , Epigenesis, Genetic/genetics , Female , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Mice, 129 Strain , Mice, Inbred ICR , Pluripotent Stem Cells/cytology
9.
Cell Rep ; 26(10): 2608-2621.e6, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30840885

ABSTRACT

Atypical teratoid/rhabdoid tumor (AT/RT), which harbors SMARCB1 mutation and exhibits a characteristic histology of rhabdoid cells, has a poor prognosis because of the lack of effective treatments. Here, we establish human SMARCB1-deficient pluripotent stem cells (hPSCs). SMARCB1-deficient hPSC-derived neural progenitor-like cells (NPLCs) efficiently give rise to brain tumors when transplanted into the mouse brain. Notably, activation of an embryonic stem cell (ESC)-like signature confers a rhabdoid histology in SMARCB1-deficient NPLC-derived tumors and causes a poor prognosis. Consistently, we find the activation of the ESC-like gene expression signature and an ESC-like DNA methylation landscape in clinical specimens of AT/RT. Finally, we identify candidate genes that maintain the activation of the ESC-like signature and the growth of AT/RT cells. Collectively, SMARCB1-deficient hPSCs offer the human models for AT/RT, which uncover the role of the activated ESC-like signature in the poor prognosis and unique histology of AT/RT.


Subject(s)
Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Rhabdoid Tumor/drug therapy , Rhabdoid Tumor/genetics , Animals , Cell Culture Techniques , Humans , Mice , Transfection , Xenograft Model Antitumor Assays
10.
Cancer Sci ; 110(3): 926-938, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30588718

ABSTRACT

The emergence of clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 technology has dramatically advanced how we manipulate the genome. Regarding in vivo experiments, Cas9-transgenic animals could provide efficient and complex genome editing. However, this potential has not been fully realized partly due to a lack of convenient platforms and limited examples of successful disease modeling. Here, we devised two doxycycline (Dox)-inducible Cas9 platforms that efficiently enable conditional genome editing at multiple loci in vitro and in vivo. In these platforms, we took advantage of a site-specific multi-segment cloning strategy for rapid and easy integration of multiple single guide (sg)RNAs. We found that a platform containing rtTA at the Rosa26 locus and TRE-Cas9 together with multiple sgRNAs at the Col1a1 locus showed higher efficiency of inducible insertions and deletions (indels) with minimal leaky editing. Using this platform, we succeeded to model Wilms' tumor and the progression of intestinal adenomas with multiple mutations including an activating mutation with a large genomic deletion. Collectively, the established platform should make complicated disease modeling in the mouse easily attainable, extending the range of in vivo experiments in various biological fields including cancer research.


Subject(s)
Adenoma/genetics , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Intestinal Neoplasms/genetics , Kidney Neoplasms/genetics , RNA, Guide, Kinetoplastida/genetics , Wilms Tumor/genetics , Adenoma/pathology , Animals , Female , Gene Editing/methods , Intestinal Neoplasms/pathology , Kidney Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mutation/genetics , Wilms Tumor/pathology
11.
Nat Commun ; 9(1): 2081, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802314

ABSTRACT

The faithful shutdown of the somatic program occurs in the early stage of reprogramming. Here, we examined the effect of in vivo reprogramming on Kras-induced cancer development. We show that the transient expression of reprogramming factors (1-3 days) in pancreatic acinar cells results in the transient repression of acinar cell enhancers, which are similarly observed in pancreatitis. We next demonstrate that Kras and p53 mutations are insufficient to induce ERK signaling in the pancreas. Notably, the transient expression of reprogramming factors in Kras mutant mice is sufficient to induce the robust and persistent activation of ERK signaling in acinar cells and rapid formation of pancreatic ductal adenocarcinoma. In contrast, the forced expression of acinar cell-related transcription factors inhibits the pancreatitis-induced activation of ERK signaling and development of precancerous lesions in Kras-mutated acinar cells. These results underscore a crucial role of dedifferentiation-associated epigenetic regulations in the initiation of pancreatic cancers.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic , Female , Humans , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Mouse Embryonic Stem Cells , Mutation , Pancreas/cytology , Pancreas/pathology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Stomach/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , alpha-Fetoproteins/metabolism
12.
J Agric Food Chem ; 56(3): 1122-30, 2008 Feb 13.
Article in English | MEDLINE | ID: mdl-18181570

ABSTRACT

This study showed that gluten proteins were extracted with distilled water from dough prepared in the presence of NaCl. To elucidate the interrelationship of NaCl and gluten proteins in dough, the extracted proteins were characterized. These proteins were primarily found to be soluble gliadin monomers by N-terminal amino acid sequencing and analytical ultracentrifugation. Extracted proteins were aggregated by the addition of NaCl at concentrations of >10 mM. A decrease in beta-turn structures, which expose tryptophan residues to an aqueous environment in the presence of NaCl, was revealed by Fourier transform infrared analysis and scanning of fluorescence spectra. In addition, cross-linking experiments with disuccinimidyl tartrate showed that a large amount of protein was cross-linked in the dough only in the presence of NaCl. These results suggest that both interactions and distances between proteins were altered by the addition of NaCl.


Subject(s)
Glutens/chemistry , Glutens/drug effects , Sodium Chloride/pharmacology , Amino Acid Sequence , Bread/analysis , Gliadin/analysis , Gliadin/chemistry , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL