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










Publication year range
1.
Biomaterials ; 307: 122522, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38428092

ABSTRACT

Cellular skin substitutes such as epidermal constructs have been developed for various applications, including wound healing and skin regeneration. These cellular models are mostly derived from primary cells such as keratinocytes and fibroblasts in a two-dimensional (2D) state, and further development of three-dimensional (3D) cultured organoids is needed to provide insight into the in vivo epidermal phenotype and physiology. Here, we report the development of epidermal organoids (EpiOs) generated from induced pluripotent stem cells (iPSCs) as a novel epidermal construct and its application as a source of secreted biomolecules recovered by extracellular vesicles (EVs) that can be utilized for cell-free therapy of regenerative medicine. Differentiated iPSC-derived epidermal organoids (iEpiOs) are easily cultured and expanded through multiple organoid passages, while retaining molecular and functional features similar to in vivo epidermis. These mature iEpiOs contain epidermal stem cell populations and retain the ability to further differentiate into other skin compartment lineages, such as hair follicle stem cells. By closely recapitulating the epidermal structure, iEpiOs are expected to provide a more relevant microenvironment to influence cellular processes and therapeutic response. Indeed, iEpiOs can generate high-performance EVs containing high levels of the angiogenic growth factor VEGF and miRNAs predicted to regulate cellular processes such as proliferation, migration, differentiation, and angiogenesis. These EVs contribute to target cell proliferation, migration, and angiogenesis, providing a promising therapeutic tool for in vivo wound healing. Overall, the newly developed iEpiOs strategy as an organoid-based approach provides a powerful model for studying basic and translational skin research and may also lead to future therapeutic applications using iEpiOs-secreted EVs.


Subject(s)
Extracellular Vesicles , Pluripotent Stem Cells , Epidermis , Cell Differentiation , Organoids , Regeneration
2.
Cell Mol Life Sci ; 81(1): 142, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38485770

ABSTRACT

Thioredoxin interacting protein (Txnip) is a stress-responsive factor regulating Trx1 for redox balance and involved in diverse cellular processes including proliferation, differentiation, apoptosis, inflammation, and metabolism. However, the biological role of Txnip function in stem cell pluripotency has yet to be investigated. Here, we reveal the novel functions of mouse Txnip in cellular reprogramming and differentiation onset by involving in glucose-mediated histone acetylation and the regulation of Oct4, which is a fundamental component of the molecular circuitry underlying pluripotency. During reprogramming or PSC differentiation process, cellular metabolic and chromatin remodeling occur in order to change its cellular fate. Txnip knockout promotes induced pluripotency but hinders initial differentiation by activating pluripotency factors and promoting glycolysis. This alteration affects the intracellular levels of acetyl-coA, a final product of enhanced glycolysis, resulting in sustained histone acetylation on active PSC gene regions. Moreover, Txnip directly interacts with Oct4, thereby repressing its activity and consequently deregulating Oct4 target gene transcriptions. Our work suggests that control of Txnip expression is crucial for cell fate transitions by modulating the entry and exit of pluripotency.


Subject(s)
Cellular Reprogramming , Histones , Animals , Mice , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/genetics , Histones/metabolism , Protein Processing, Post-Translational , Thioredoxins/genetics , Thioredoxins/metabolism
3.
EMBO Mol Med ; 14(1): e14678, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34842355

ABSTRACT

Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli (EHEC) are the major virulence factors responsible for hemorrhagic colitis, which can lead to life-threatening systemic complications including acute renal failure (hemolytic uremic syndrome) and neuropathy. Here, we report that O-GlcNAcylation, a type of post-translational modification, was acutely increased upon induction of endoplasmic reticulum (ER) stress in host cells by Stxs. Suppression of the abnormal Stx-mediated increase in O-GlcNAcylation effectively inhibited apoptotic and inflammatory responses in Stx-susceptible cells. The protective effect of O-GlcNAc inhibition for Stx-mediated pathogenic responses was also verified using three-dimensional (3D)-cultured spheroids or organoids mimicking the human kidney. Treatment with an O-GlcNAcylation inhibitor remarkably improved the major disease symptoms and survival rate for mice intraperitoneally injected with a lethal dose of Stx. In conclusion, this study elucidates O-GlcNAcylation-dependent pathogenic mechanisms of Stxs and demonstrates that inhibition of aberrant O-GlcNAcylation is a potential approach to treat Stx-mediated diseases.


Subject(s)
Escherichia coli Infections , Hemolytic-Uremic Syndrome , Animals , Endoplasmic Reticulum Stress , Hemolytic-Uremic Syndrome/pathology , Kidney/pathology , Mice , Shiga Toxin/metabolism , Shiga Toxins
4.
Cancers (Basel) ; 13(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298635

ABSTRACT

2-oxoglutarate and iron-dependent oxygenase domain-containing protein 1 (OGFOD1) expression is upregulated in a variety of cancers and has been related to poor prognosis. However, despite this significance to cancer progression, the precise oncogenic mechanism of OGFOD1 is not understood. We demonstrated that OGFOD1 plays a role in enhancing the transcriptional activity of RNA polymerase II in breast cancer cells. OGFOD1 directly binds to the C-terminal domain of RNA polymerase II to alter phosphorylation status. The elimination of OGFOD1 resulted in decreased tumor development. Additionally, cell cycle-dependent kinase 7 and cell cycle-dependent kinase 9, critical enzymes for activating RNA polymerase II, phosphorylated serine 256 of OGFOD1, whereas a non-phosphorylated mutant OGFOD1 failed to enhance transcriptional activation and tumor growth. Consequently, OGFOD1 helps promote tumor growth by enhancing RNA polymerase II, whereas simultaneous phosphorylation of OGFOD1 by CDK enzymes is essential in stimulating RNA polymerase II-mediated transcription both in vitro and in vivo, and expression of target genes.

5.
Nucleic Acids Res ; 46(22): 11759-11775, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30335163

ABSTRACT

Constitutive heterochromatin undergoes a dynamic clustering and spatial reorganization during myogenic differentiation. However the detailed mechanisms and its role in cell differentiation remain largely elusive. Here, we report the identification of a muscle-specific long non-coding RNA, ChRO1, involved in constitutive heterochromatin reorganization. ChRO1 is induced during terminal differentiation of myoblasts, and is specifically localized to the chromocenters in myotubes. ChRO1 is required for efficient cell differentiation, with global impacts on gene expression. It influences DNA methylation and chromatin compaction at peri/centromeric regions. Inhibition of ChRO1 leads to defects in the spatial fusion of chromocenters, and mislocalization of H4K20 trimethylation, Suv420H2, HP1, MeCP2 and cohesin. In particular, ChRO1 specifically associates with ATRX/DAXX/H3.3 complex at chromocenters to promote H3.3 incorporation and transcriptional induction of satellite repeats, which is essential for chromocenter clustering. Thus, our results unveil a mechanism involving a lncRNA that plays a role in large-scale heterochromatin reorganization and cell differentiation.


Subject(s)
Carrier Proteins/genetics , Heterochromatin/chemistry , Histones/genetics , Intracellular Signaling Peptides and Proteins/genetics , Muscle Development/genetics , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , X-linked Nuclear Protein/genetics , Animals , CRISPR-Cas Systems , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Co-Repressor Proteins , Female , Gene Editing , Gene Expression Regulation, Developmental , HEK293 Cells , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Molecular Chaperones , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , NIH 3T3 Cells , Nuclear Proteins/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic , X-linked Nuclear Protein/metabolism , Cohesins
6.
Nucleic Acids Res ; 46(13): 6592-6607, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29846698

ABSTRACT

Transcription factors and chromatin remodeling proteins control the transcriptional variability for ESC lineage commitment. During ESC differentiation, chromatin modifiers are recruited to the regulatory regions by transcription factors, thereby activating the lineage-specific genes or silencing the transcription of active ESC genes. However, the underlying mechanisms that link transcription factors to exit from pluripotency are yet to be identified. In this study, we show that the Ctbp2-interacting zinc finger proteins, Zfp217 and Zfp516, function as linkers for the chromatin regulators during ESC differentiation. CRISPR-Cas9-mediated knock-outs of both Zfp217 and Zfp516 in ESCs prevent the exit from pluripotency. Both zinc finger proteins regulate the Ctbp2-mediated recruitment of the NuRD complex and polycomb repressive complex 2 (PRC2) to active ESC genes, subsequently switching the H3K27ac to H3K27me3 during ESC differentiation for active gene silencing. We therefore suggest that some zinc finger proteins orchestrate to control the concise epigenetic states on active ESC genes during differentiation, resulting in natural lineage commitment.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Gene Silencing , Trans-Activators/physiology , Alcohol Oxidoreductases/metabolism , Animals , Cells, Cultured , Co-Repressor Proteins , Embryonic Stem Cells/cytology , Humans , Mice , Nerve Tissue Proteins/metabolism , Transcription, Genetic
7.
Exp Mol Med ; 49(10): e385, 2017 10 13.
Article in English | MEDLINE | ID: mdl-29026198

ABSTRACT

The canonical Wnt pathway is critical for embryonic stem cell (ESC) pluripotency and aberrant control of ß-catenin leads to failure of exit from pluripotency and lineage commitments. Hence, maintaining the appropriate level of ß-catenin is important for the decision to commit to the appropriate lineage. However, how ß-catenin links to core transcription factors in ESCs remains elusive. C-terminal-binding protein (CtBP) in Drosophila is essential for Wnt-mediated target gene expression. In addition, Ctbp acts as an antagonist of ß-catenin/TCF activation in mammals. Recently, Ctbp2, a core Oct4-binding protein in ESCs, has been reported to play a key role in ESC pluripotency. However, the significance of the connection between Ctbp2 and ß-catenin with regard to ESC pluripotency remains elusive. Here, we demonstrate that C-terminal-binding protein 2 (Ctbp2) associates with major components of the ß-catenin destruction complex and limits the accessibility of ß-catenin to core transcription factors in undifferentiated ESCs. Ctbp2 knockdown leads to stabilization of ß-catenin, which then interacts with core pluripotency-maintaining factors that are occupied by Ctbp2, leading to incomplete exit from pluripotency. These findings suggest a suppressive function for Ctbp2 in reducing the protein level of ß-catenin, along with priming its position on core pluripotency genes to hinder ß-catenin deposition, which is central to commitment to the appropriate lineage.


Subject(s)
Cell Self Renewal/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , beta Catenin/metabolism , Alcohol Oxidoreductases , Animals , Binding Sites , Cell Line , Co-Repressor Proteins , Embryonic Stem Cells , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , Mice , Models, Biological , Nucleotide Motifs , Protein Binding , Protein Stability , RNA, Small Interfering/genetics
8.
Cell Metab ; 24(3): 494-501, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27476977

ABSTRACT

Embryonic stem cells (ESCs) undergo coordinated epigenetic and metabolic changes to differentiate properly. However, the precise mechanisms by which these alterations are fine-tuned in the early stages of differentiation have not been identified. In this study, we demonstrate that phosphoserine aminotransferase 1 (Psat1), an Oct4/Sox2/Nanog (OSN) target protein, regulates changes in α-ketoglutarate (α-KG), determining the fate of mouse ESCs (mESCs). Maintaining Psat1 levels was essential for mESC self-renewal and pluripotency. Moderate knockdown (KD) of Psat1 in mESCs lowered DNA 5'-hydroxymethylcytosine (5'-hmC) and increased histone methylation levels by downregulating permissive amounts of α-KG, ultimately accelerating differentiation. We found that intracellular α-KG declined transiently during differentiation and that its dysregulation by treatment with dimethyl-α-KG impeded differentiation. Further, by in vivo teratoma formation assay, pluripotency of Psat1 KD mESCs was impaired, especially into the ectodermal lineage. Thus, we have established how Psat1 is regulated in maintaining intracellular α-KG levels and determining the fate of mESCs.


Subject(s)
Cell Differentiation , Ketoglutaric Acids/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Transaminases/metabolism , Animals , DNA Methylation , Histones/metabolism , Intracellular Space/metabolism , Mice , Time Factors , Transcription Factors/metabolism
9.
Elife ; 5: e10877, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26880562

ABSTRACT

Pluripotency transcription programs by core transcription factors (CTFs) might be reset during M/G1 transition to maintain the pluripotency of embryonic stem cells (ESCs). However, little is known about how CTFs are governed during cell cycle progression. Here, we demonstrate that the regulation of Oct4 by Aurora kinase b (Aurkb)/protein phosphatase 1 (PP1) during the cell cycle is important for resetting Oct4 to pluripotency and cell cycle genes in determining the identity of ESCs. Aurkb phosphorylates Oct4(S229) during G2/M phase, leading to the dissociation of Oct4 from chromatin, whereas PP1 binds Oct4 and dephosphorylates Oct4(S229) during M/G1 transition, which resets Oct4-driven transcription for pluripotency and the cell cycle. Aurkb phosphor-mimetic and PP1 binding-deficient mutations in Oct4 alter the cell cycle, effect the loss of pluripotency in ESCs, and decrease the efficiency of somatic cell reprogramming. Our findings provide evidence that the cell cycle is linked directly to pluripotency programs in ESCs.


Subject(s)
Aurora Kinase B/metabolism , Cell Cycle , Embryonic Stem Cells/physiology , Octamer Transcription Factor-3/metabolism , Protein Phosphatase 1/metabolism , Animals , Mice , Phosphorylation , Protein Processing, Post-Translational
10.
Stem Cells ; 33(8): 2442-55, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25944056

ABSTRACT

For cells to exit from pluripotency and commit to a lineage, the circuitry of a core transcription factor (CTF) network must be extinguished in an orderly manner through epigenetic modifications. However, how this choreographed epigenetic remodeling at active embryonic stem cell (ESC) genes occurs during differentiation is poorly understood. In this study, we demonstrate that C-terminal binding protein 2 (Ctbp2) regulates nucleosome remodeling and deacetylation (NuRD)-mediated deacetylation of H3K27 and facilitates recruitment of polycomb repressive complex 2 (PRC2)-mediated H3K27me3 in active ESC genes for exit from pluripotency during differentiation. By genomewide analysis, we found that Ctbp2 resides in active ESC genes and co-occupies regions with ESC CTFs in undifferentiated ESCs. Furthermore, ablation of Ctbp2 effects inappropriate gene silencing in ESCs by sustaining high levels of H3K27ac and impeding H3K27me3 in active ESC genes, thereby sustaining ESC maintenance during differentiation. Thus, Ctbp2 preoccupies regions in active genes with the NuRD complex in undifferentiated ESCs that are directed toward H3K27me3 by PRC2 to induce stable silencing, which is pivotal for natural lineage commitment.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenesis, Genetic/physiology , Histones/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mouse Embryonic Stem Cells/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Alcohol Oxidoreductases , Animals , Cell Line , Chromatin Assembly and Disassembly/physiology , Co-Repressor Proteins , DNA-Binding Proteins/genetics , Histones/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Nucleosomes/genetics , Nucleosomes/metabolism , Phosphoproteins/genetics , Repressor Proteins/genetics
11.
Oncotarget ; 6(23): 19528-41, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-25909288

ABSTRACT

2-oxogluatrate and Fe(II)-dependent oxygenase domain-containing protein 1 (OGFOD1) was recently revealed to be a proline hydroxylase of RPS23 for translational termination. However, OGFOD1 is nuclear, whereas translational termination occurs in the cytoplasm, raising the possibility of another function of OGFOD1 in the nucleus. In this study, we demonstrate that OGFOD1 is involved in cell cycle regulation. OGFOD1 knockdown in MDA-MB-231 breast cancer cells significantly impeded cell proliferation and resulted in the accumulation of G1 and G2/M cells by decreasing the mRNA levels of G1/S transition- and G2/M-related transcription factors and their target genes. We also confirmed that OGFOD1 is highly expressed in breast cancer tissues by bioinformatic analysis and immunohistochemistry. Thus, we propose that OGFOD1 is required for breast cancer cell proliferation and is associated with poor prognosis in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Proliferation , Nuclear Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly , Computational Biology , Databases, Genetic , Female , G1 Phase Cell Cycle Checkpoints , G2 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Immunohistochemistry , In Situ Hybridization , Kaplan-Meier Estimate , MCF-7 Cells , Nuclear Proteins/genetics , Prognosis , RNA Interference , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
12.
Nucleic Acids Res ; 43(9): 4505-16, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25813038

ABSTRACT

Post-translational modifications of core histones affect various cellular processes, primarily through transcription. However, their relationship with the termination of transcription has remained largely unknown. In this study, we show that DNA damage-activated AKT phosphorylates threonine 45 of core histone H3 (H3-T45). By genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) analysis, H3-T45 phosphorylation was distributed throughout DNA damage-responsive gene loci, particularly immediately after the transcription termination site. H3-T45 phosphorylation pattern showed close-resemblance to that of RNA polymerase II C-terminal domain (CTD) serine 2 phosphorylation, which establishes the transcription termination signal. AKT1 was more effective than AKT2 in phosphorylating H3-T45. Blocking H3-T45 phosphorylation by inhibiting AKT or through amino acid substitution limited RNA decay downstream of mRNA cleavage sites and decreased RNA polymerase II release from chromatin. Our findings suggest that AKT-mediated phosphorylation of H3-T45 regulates the processing of the 3' end of DNA damage-activated genes to facilitate transcriptional termination.


Subject(s)
DNA Damage , Histones/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Termination, Genetic , Cell Line , HeLa Cells , Histones/chemistry , Humans , MCF-7 Cells , Phosphorylation , Threonine/metabolism , Transcription Initiation Site
SELECTION OF CITATIONS
SEARCH DETAIL
...