Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(7)2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38612707

ABSTRACT

Cancers harness embryonic programs to evade aging and promote survival. Normally, sequences at chromosome ends called telomeres shorten with cell division, serving as a countdown clock to limit cell replication. Therefore, a crucial aspect of cancerous transformation is avoiding replicative aging by activation of telomere repair programs. Mouse embryonic stem cells (mESCs) activate a transient expression of the gene Zscan4, which correlates with chromatin de-condensation and telomere extension. Head and neck squamous cell carcinoma (HNSCC) cancers reactivate ZSCAN4, which in turn regulates the phenotype of cancer stem cells (CSCs). Our study reveals a new role for human ZSCAN4 in facilitating functional histone H3 acetylation at telomere chromatin. Next-generation sequencing indicates ZSCAN4 enrichment at telomere chromatin. These changes correlate with ZSCAN4-induced histone H3 acetylation and telomere elongation, while CRISPR/Cas9 knockout of ZSCAN4 leads to reduced H3 acetylation and telomere shortening. Our study elucidates the intricate involvement of ZSCAN4 and its significant contribution to telomere chromatin remodeling. These findings suggest that ZSCAN4 induction serves as a novel link between 'stemness' and telomere maintenance. Targeting ZSCAN4 may offer new therapeutic approaches to effectively limit or enhance the replicative lifespan of stem cells and cancer cells.


Subject(s)
Histones , Telomere , Animals , Mice , Humans , Acetylation , Telomere/genetics , Chromatin/genetics , Aging
2.
Adv Sci (Weinh) ; 11(11): e2306683, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38183347

ABSTRACT

3D bioprinting holds great promise for meeting the increasing need for transplantable tissues and organs. However, slow printing, interlayer mixing, and the extended exposure of cells to non-physiological conditions in thick structures still hinder clinical applications. Here the DeepFreeze-3D (DF-3D) procedure and bioink for creating multilayered human-scale tissue mimetics is presented for the first time. The bioink is tailored to support stem cell viability, throughout the rapid freeform DF-3D biofabrication process. While the printer nozzle is warmed to room temperature, each layer solidifies at contact with the stage (-80 °C), or the subsequent layers, ensuring precise separation. After thawing, the encapsulated stem cells remain viable without interlayer mixing or delamination. The composed cell-laden constructs can be cryogenically stored and thawed when needed. Moreover, it is shown that under inductive conditions the stem cells differentiate into bone-like cells and grow for months after thawing, to form large tissue-mimetics in the scale of centimeters. This is important, as this approach allows the generation and storage of tissue mimetics in the size and thickness of human tissues. Therefore, DF-3D biofabrication opens new avenues for generating off-the-shelf human tissue analogs. It further holds the potential for regenerative treatments and for studying tissue pathologies caused by disease, tumor, or trauma.


Subject(s)
Bioprinting , Tissue Engineering , Humans , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Printing, Three-Dimensional , Bioprinting/methods , Bioengineering , Stem Cells
3.
OBM Geriat ; 6(2)2022.
Article in English | MEDLINE | ID: mdl-35812693

ABSTRACT

Telomeres consist of special features and proteins to protect the ends of each chromosome from deterioration and fusion. The telomeric DNA repeats are highly susceptible to oxidative damage that can accelerate telomere shortening and affect telomere integrity. Several DNA repair factors including MYH/MUTYH DNA glycosylase, its interacting partners Rad9/Rad1/Hus1 checkpoint clamp, and SIRT6 aging regulator, are associated with the telomeres. MYH prevents C:G to A:T mutation by removing adenine mispaired with a frequent oxidative DNA lesion, 8-oxoguanine. Here, we show that hMYH knockout (KO) human HEK-293T cells are more sensitive to H2O2 treatment, have higher levels of DNA strand breaks and shorter telomeres than the control hMYH +/+ cells. SIRT6 foci increase at both the global genome and at telomeric regions in H2O2-treated hMYH +/+ cells. However, in untreated hMYH KO HEK-293T cells, SIRT6 foci only increase at the global genome, but not at the telomeric regions. In addition, the hMYH KO HEK-293T cells have increased extra-chromosomal and intra-chromosomal telomeres compared to the control cells, even in the absence of H2O2 treatment. After H2O2 treatment, the frequency of extra-chromosomal telomeres increased in control HEK-293T cells. Remarkably, in H2O2-treated hMYH KO cells, the frequencies of extra-chromosomal telomeres, intra-chromosomal telomeres, and telomere fusions are further increased. We further found that the sensitivity to H2O2 and shortened telomeres of hMYH KO cells, are restored by expressing wild-type hMYH, and partially rescued by expressing hMYHQ324H mutant (defective in Hus1 interaction only), but not by expressing hMYHV315A mutant (defective in both SIRT6 and Hus1 interactions). Thus, MYH interactions with SIRT6 and Hus1 are critical for maintaining cell viability and telomeric stability. Therefore, the failure to coordinate 8-oxoG repair is detrimental to telomere integrity.

4.
Cell Mol Biol Lett ; 26(1): 38, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34407767

ABSTRACT

BACKGROUND: Neurological disorders are considered one of the greatest burdens to global public health and a leading cause of death. Stem cell therapies hold great promise for the cure of neurological disorders, as stem cells can serve as cell replacement, while also secreting factors to enhance endogenous tissue regeneration. Adult human multipotent stem cells (MSCs) reside on blood vessels, and therefore can be found in many tissues throughout the body, including palatine tonsils. Several studies have reported the capacity of MSCs to differentiate into, among other cell types, the neuronal lineage. However, unlike the case with embryonic stem cells, it is unclear whether MSCs can develop into mature neurons. METHODS: Human tonsillar MSCs (T-MSCs) were isolated from a small, 0.6-g sample, of tonsillar biopsies with high viability and yield as we recently reported. Then, these cells were differentiated by a rapid, multi-stage procedure, into committed, post-mitotic, neuron-like cells using defined conditions. RESULTS: Here we describe for the first time the derivation and differentiation of tonsillar biopsy-derived MSCs (T-MSCs), by a rapid, multi-step protocol, into post-mitotic, neuron-like cells using defined conditions without genetic manipulation. We characterized our T-MSC-derived neuronal cells and demonstrate their robust differentiation in vitro. CONCLUSIONS: Our procedure leads to a rapid neuronal lineage commitment and loss of stemness markers, as early as three days following neurogenic differentiation. Our studies identify biopsy-derived T-MSCs as a potential source for generating neuron-like cells which may have potential use for in vitro modeling of neurodegenerative diseases or cell replacement therapies.


Subject(s)
Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Neurons/cytology , Palatine Tonsil/cytology , Adult , Biopsy , Cell Differentiation/physiology , Cell Lineage , Cells, Cultured , Child , Child, Preschool , Female , Humans , Male , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Neurons/metabolism , Palatine Tonsil/metabolism , Palatine Tonsil/surgery , Young Adult
5.
Int J Mol Sci ; 21(16)2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32824412

ABSTRACT

Reduced NME1 expression in melanoma cell lines, mouse models of melanoma, and melanoma specimens in human patients is associated with increased metastatic activity. Herein, we investigate the role of NME1 in repair of double-stranded breaks (DSBs) and choice of double-strand break repair (DSBR) pathways in melanoma cells. Using chromatin immunoprecipitation, NME1 was shown to be recruited rapidly and directly to DSBs generated by the homing endonuclease I-PpoI. NME1 was recruited to DSBs within 30 min, in concert with recruitment of ataxia-telangiectasia mutated (ATM) protein, an early step in DSBR complex formation, as well as loss of histone 2B. NME1 was detected up to 5 kb from the break site after DSB induction, suggesting a role in extending chromatin reorganization away from the repair site. shRNA-mediated silencing of NME1 expression led to increases in the homologous recombination (HR) and non-homologous end-joining (NHEJ) pathways of double-strand break repair (DSBR), and reduction in the low fidelity, alternative-NHEJ (A-NHEJ) pathway. These findings suggest low expression of NME1 drives DSBR towards higher fidelity pathways, conferring enhanced genomic stability necessary for rapid and error-free proliferation in invasive and metastatic cells. The novel mechanism highlighted in the current study appears likely to impact metastatic potential and therapy-resistance in advanced melanoma and other cancers.


Subject(s)
Melanoma/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Recombinational DNA Repair , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Endodeoxyribonucleases/metabolism , Genomic Instability , Histones/metabolism , Humans , NM23 Nucleoside Diphosphate Kinases/genetics
6.
Oncogene ; 39(26): 4970-4982, 2020 06.
Article in English | MEDLINE | ID: mdl-32507861

ABSTRACT

Cancer stem cells (CSCs) are cells within tumors that maintain the ability to self-renew, drive tumor growth, and contribute to therapeutic resistance and cancer recurrence. In this study, we investigate the role of Zinc finger and SCAN domain containing 4 (ZSCAN4) in human head and neck squamous cell carcinoma (HNSCC). The murine Zscan4 is involved in telomere maintenance and genomic stability of mouse embryonic stem cells. Our data indicate that the human ZSCAN4 is enriched for, marks and is co-expressed with CSC markers in HNSCC. We show that transient ZSCAN4 induction for just 2 days increases CSC frequency both in vitro and in vivo and leads to upregulation of pluripotency and CSC factors. Importantly, we define for the first time the role of ZSCAN4 in altering the epigenetic profile and regulating the chromatin state. Our data show that ZSCAN4 leads to a functional histone 3 hyperacetylation at the promoters of OCT3/4 and NANOG, leading to an upregulation of CSC factors. Consistently, ZSCAN4 depletion leads to downregulation of CSC markers, decreased ability to form tumorspheres and severely affects tumor growth. Our study suggests that ZSCAN4 plays an important role in the maintenance of the CSC phenotype, indicating it is a potential therapeutic target in HNSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Transcription Factors/genetics , Acetylation , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/therapy , Histones/metabolism , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phenotype , RNA Interference , Transcription Factors/metabolism , Xenograft Model Antitumor Assays/methods
7.
Curr Issues Mol Biol ; 35: 85-98, 2020.
Article in English | MEDLINE | ID: mdl-31422934

ABSTRACT

Telomeres are a unique structure of DNA repeats covered by proteins at the ends of the chromosomes that protect the coding regions of the genome and function as a biological clock. They require a tight regulation of the factors covering and protecting their structure, as they are shortened with each cell division to limit the ability of cells to replicate uncontrollably. Additionally, they protect the chromosome ends from DNA damage responses and thereby, prevent genomic instability. Telomere dysfunction can lead to chromosomal abnormalities and cancer. Therefore, dysregulation of any of the factors that regulate the integrity of the telomeres will have implications to chromosomal stability, replicative lifespan and may lead to cell transformation. This review will cover the main factors participating in the normal function of the telomeres and how these are regulated by the ubiquitin and SUMO systems. Accumulating evidence indicate that the ubiquitin and SUMO pathways are significant regulators of the shelterin complex and other chromatin modifiers, which are important for telomere structure integrity. Furthermore, the crosstalk between these two pathways has been reported in telomeric DNA repair. A better understanding of the factors contributing to telomere biology, and how they are regulated, is important for the design of new strategies for cancer therapies and regenerative medicine.


Subject(s)
Chromatin/metabolism , DNA Replication/genetics , Neoplasms/metabolism , Sumoylation , Telomerase/metabolism , Telomere/metabolism , Ubiquitination , Animals , Chromatin/enzymology , DNA Repair , Humans , Neoplasms/enzymology , Neoplasms/genetics , Shelterin Complex , Telomerase/genetics , Telomere/genetics , Telomere-Binding Proteins/metabolism
8.
Biochem Biophys Res Commun ; 498(1): 72-78, 2018 03 25.
Article in English | MEDLINE | ID: mdl-29477841

ABSTRACT

Zscan4 is an early embryonic gene cluster expressed in mouse embryonic stem and induced pluripotent stem cells where it plays critical roles in genomic stability, telomere maintenance, and pluripotency. Zscan4 expression is transient, and characterized by infrequent high expression peaks that are quickly down-regulated, suggesting its expression is tightly controlled. However, little is known about the protein degradation pathway responsible for regulating the human ZSCAN4 protein levels. In this study we determine for the first time the ZSCAN4 protein half-life and degradation pathway, including key factors involved in the process, responsible for the regulation of ZSCAN4 stability. We demonstrate lysine 48 specific polyubiquitination and subsequent proteasome dependent degradation of ZSCAN4, which may explain how this key factor is efficiently cleared from the cells. Importantly, our data indicate an interaction between ZSCAN4 and the E3 ubiquitin ligase RNF20. Moreover, our results show that RNF20 depletion by gene knockdown does not affect ZSCAN4 transcription levels, but instead results in increased ZSCAN4 protein levels. Further, RNF20 depletion stabilizes the ZSCAN4 protein half-life, suggesting that RNF20 negatively regulates ZSCAN4 stability. Due to the significant cellular functions of ZSCAN4, our results have important implications in telomere regulation, stem cell biology, and cancer.


Subject(s)
DNA-Binding Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Cell Line , Half-Life , Humans , Lysine/metabolism , Polyubiquitin/metabolism , Protein Stability , Proteolysis , Ubiquitination
9.
Stem Cell Res Ther ; 8(1): 174, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28750664

ABSTRACT

BACKGROUND: Human adult stem cells hold the potential for the cure of numerous conditions and degenerative diseases. They possess major advantages over pluripotent stem cells as they can be derived from donors at any age, and therefore pose no ethical concerns or risk of teratoma tumor formation in vivo. Furthermore, they have a natural ability to differentiate and secrete factors that promote tissue healing without genetic manipulation. However, at present, clinical applications of adult stem cells are limited by a shortage of a reliable, standardized, and easily accessible tissue source which does not rely on specimens discarded from unrelated surgical procedures. METHOD: Human tonsil-derived mesenchymal progenitor cells (MPCs) were isolated from a small sample of tonsillar tissue (average 0.88 cm3). Our novel procedure poses a minimal mechanical and enzymatic insult to the tissue, and therefore leads to high cell viability and yield. We characterized these MPCs and demonstrated robust multipotency in vitro. We further show that these cells can be propagated and maintained in xeno-free conditions. RESULTS: We have generated tonsillar biopsy-derived MPC (T-MPC) lines from multiple donors across a spectrum of age, sex, and race, and successfully expanded them in culture. We characterized them by cell surface markers, as well as in vitro expansion and differentiation potential. Our procedure provides a robust yield of tonsillar biopsy-derived T-MPCs. CONCLUSIONS: Millions of MPCs can be harvested from a sample smaller than 1 g, which can be collected from a fully awake donor in an outpatient setting without the need for general anesthesia or hospitalization. Our study identifies tonsillar biopsy as an abundant source of adult MPCs for regenerative medicine.


Subject(s)
Cell Separation/methods , Multipotent Stem Cells/pathology , Palatine Tonsil/pathology , Biopsy , Female , Humans , Male
10.
Mol Biol Cell ; 28(3): 396-410, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27932492

ABSTRACT

MARCH5, an OMM-associated E3 ubiquitin ligase, controls mitochondrial function. Despite its importance, the mechanism and factors controlling MARCH5 activity are largely unknown. Here we report that the MARCH5 C-terminal domain plays a critical role in degradation of MARCH5 substrates, likely by facilitating release of ubiquitinated proteins from the OMM. We also found that the mitochondrial fission proteins Drp1 and Mff negatively regulate MARCH5's activity toward MiD49 and Mcl1. Knockouts of either Drp1 or Mff led to reduced expression, shorter half-lives, and increased ubiquitination of MiD49 and Mcl1. Effects of Mff and Drp1 depletion on degradation rates and ubiquitination of Mcl1 and MiD49 were eliminated in Drp1-/-/MARCH5-/- and Mff-/-/MARCH5-/- cells. Our data show that it is not mitochondrial morphology per se but rather Mff and Drp1 that directly control MARCH5. Consistently, we find that Mff is an integral component of the MARCH5/p97/Npl4 complex, which is also controlled by MARCH5's C-terminal domain. Furthermore, not only mitochondrial fission but also fusion is regulated through Mff and Drp1 protein activities. Thus, in addition to their canonical roles in mitochondrial fission, Mff and Drp1 also act as regulatory factors that control mitochondrial fission and fusion.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mitochondrial Dynamics/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology , Cell Culture Techniques , Dynamins , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/physiology , HCT116 Cells , Humans , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/physiology , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/physiology , Peptide Elongation Factors/metabolism , Peptide Elongation Factors/physiology , Ubiquitination
11.
BMC Mol Biol ; 16: 12, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26063178

ABSTRACT

BACKGROUND: SIRT6, a member of the NAD(+)-dependent histone/protein deacetylase family, regulates genomic stability, metabolism, and lifespan. MYH glycosylase and APE1 are two base excision repair (BER) enzymes involved in mutation avoidance from oxidative DNA damage. Rad9-Rad1-Hus1 (9-1-1) checkpoint clamp promotes cell cycle checkpoint signaling and DNA repair. BER is coordinated with the checkpoint machinery and requires chromatin remodeling for efficient repair. SIRT6 is involved in DNA double-strand break repair and has been implicated in BER. Here we investigate the direct physical and functional interactions between SIRT6 and BER enzymes. RESULTS: We show that SIRT6 interacts with and stimulates MYH glycosylase and APE1. In addition, SIRT6 interacts with the 9-1-1 checkpoint clamp. These interactions are enhanced following oxidative stress. The interdomain connector of MYH is important for interactions with SIRT6, APE1, and 9-1-1. Mutagenesis studies indicate that SIRT6, APE1, and Hus1 bind overlapping but different sequence motifs on MYH. However, there is no competition of APE1, Hus1, or SIRT6 binding to MYH. Rather, one MYH partner enhances the association of the other two partners to MYH. Moreover, APE1 and Hus1 act together to stabilize the MYH/SIRT6 complex. Within human cells, MYH and SIRT6 are efficiently recruited to confined oxidative DNA damage sites within transcriptionally active chromatin, but not within repressive chromatin. In addition, Myh foci induced by oxidative stress and Sirt6 depletion are frequently localized on mouse telomeres. CONCLUSIONS: Although SIRT6, APE1, and 9-1-1 bind to the interdomain connector of MYH, they do not compete for MYH association. Our findings indicate that SIRT6 forms a complex with MYH, APE1, and 9-1-1 to maintain genomic and telomeric integrity in mammalian cells.


Subject(s)
Cell Cycle Checkpoints , DNA Repair , DNA/metabolism , Sirtuins/metabolism , Amino Acid Motifs , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromatin/genetics , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Exonucleases/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Sirtuins/genetics , Telomere/metabolism
12.
Sci Rep ; 3: 1390, 2013.
Article in English | MEDLINE | ID: mdl-23462645

ABSTRACT

Networks of transcription factors (TFs) are thought to determine and maintain the identity of cells. Here we systematically repressed each of 100 TFs with shRNA and carried out global gene expression profiling in mouse embryonic stem (ES) cells. Unexpectedly, only the repression of a handful of TFs significantly affected transcriptomes, which changed in two directions/trajectories: one trajectory by the repression of either Pou5f1 or Sox2; the other trajectory by the repression of either Esrrb, Sall4, Nanog, or Tcfap4. The data suggest that the trajectories of gene expression change are already preconfigured by the gene regulatory network and roughly correspond to extraembryonic and embryonic fates of cell differentiation, respectively. These data also indicate the robustness of the pluripotency gene network, as the transient repression of most TFs did not alter the transcriptomes.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/genetics , Animals , Cluster Analysis , Gene Expression Profiling , Gene Silencing , Mice , Models, Biological , RNA Interference , Transcription Factors/metabolism , Transcriptome
13.
Nature ; 464(7290): 858-63, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20336070

ABSTRACT

Exceptional genomic stability is one of the hallmarks of mouse embryonic stem (ES) cells. However, the genes contributing to this stability remain obscure. We previously identified Zscan4 as a specific marker for two-cell embryo and ES cells. Here we show that Zscan4 is involved in telomere maintenance and long-term genomic stability in ES cells. Only 5% of ES cells express Zscan4 at a given time, but nearly all ES cells activate Zscan4 at least once during nine passages. The transient Zscan4-positive state is associated with rapid telomere extension by telomere recombination and upregulation of meiosis-specific homologous recombination genes, which encode proteins that are colocalized with ZSCAN4 on telomeres. Furthermore, Zscan4 knockdown shortens telomeres, increases karyotype abnormalities and spontaneous sister chromatid exchange, and slows down cell proliferation until reaching crisis by passage eight. Together, our data show a unique mode of genome maintenance in ES cells.


Subject(s)
Embryonic Stem Cells/metabolism , Genomic Instability , Telomere/genetics , Telomere/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cell Proliferation , Chromosome Aberrations , Embryonic Stem Cells/cytology , Embryonic Stem Cells/pathology , Gene Expression Regulation , Gene Knockdown Techniques , Karyotyping , Meiosis/genetics , Meiosis/physiology , Mice , Protein Transport , Recombination, Genetic/genetics , Sister Chromatid Exchange/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Up-Regulation
14.
Cell Stem Cell ; 5(4): 420-33, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19796622

ABSTRACT

To examine transcription factor (TF) network(s), we created mouse ESC lines, in each of which 1 of 50 TFs tagged with a FLAG moiety is inserted into a ubiquitously controllable tetracycline-repressible locus. Of the 50 TFs, Cdx2 provoked the most extensive transcriptome perturbation in ESCs, followed by Esx1, Sox9, Tcf3, Klf4, and Gata3. ChIP-Seq revealed that CDX2 binds to promoters of upregulated target genes. By contrast, genes downregulated by CDX2 did not show CDX2 binding but were enriched with binding sites for POU5F1, SOX2, and NANOG. Genes with binding sites for these core TFs were also downregulated by the induction of at least 15 other TFs, suggesting a common initial step for ESC differentiation mediated by interference with the binding of core TFs to their target genes. These ESC lines provide a fundamental resource to study biological networks in ESCs and mice.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Regulatory Networks/physiology , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , CDX2 Transcription Factor , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Chromatin Immunoprecipitation , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , GATA3 Transcription Factor/physiology , Gene Regulatory Networks/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Immunoprecipitation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/physiology , Mice , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Biochem Biophys Res Commun ; 341(2): 291-8, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16446152

ABSTRACT

Beta-cell replacement represents the ultimate cure for type 1 diabetes, however it is limited by availability of organ donors. Adult human islets are difficult to propagate in culture, and efforts to expand them result in dedifferentiation. Here we describe conditions for expansion of adult human islet cells, as well as a way for their redifferentiation. Most cells in islets isolated from human pancreata were induced to replicate within the first week of culture in expansion medium. Cells were propagated for 16 population doublings, without a change in replication rate or noticeable cell mortality, representing an expansion of over 65,000-fold. Replication was accompanied by a decrease in expression of key beta-cell genes. Shift of the cells to differentiation medium containing betacellulin resulted in redifferentiation, as manifested by restoration of beta-cell gene expression and insulin content. These methods may allow transplantation of functional islet cells from single donors into multiple recipients.


Subject(s)
Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Adult , Aged , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Dose-Response Relationship, Drug , Female , Glucose/pharmacology , Humans , Insulin/metabolism , Ischemia , Male , Microscopy, Confocal , Microscopy, Fluorescence , Middle Aged , Peptides/chemistry , Phenotype , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
16.
Diabetes ; 54(9): 2568-75, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123344

ABSTRACT

beta-Cell transplantation is viewed as a cure for type 1 diabetes; however, it is limited by the number of pancreas donors. Human stem cells offer the promise of an abundant source of insulin-producing cells, given the existence of methods for manipulating their differentiation. We have previously demonstrated that the expression of the beta-cell transcription factor pancreatic duodenal homeobox 1 (PDX-1) in human fetal liver cells activates multiple aspects of the beta-cell phenotype. These cells, termed FH-B-TPN cells, produce insulin, release insulin in response to physiological glucose levels, and replace beta-cell function in diabetic immunodeficient mice. However, they deviate from the normal beta-cell phenotype by the lack of expression of a number of beta-cell genes, the expression of non-beta-cell genes, and a lower insulin content. Here we aimed to promote differentiation of FH-B-TPN cells toward the beta-cell phenotype using soluble factors. Cells cultured with activin A in serum-free medium upregulated expression of NeuroD and Nkx2.2 and downregulated paired box homeotic gene 6 (PAX-6). Glucokinase and prohormone convertase 1/3 were also upregulated, whereas pancreatic polypeptide and glucagon as well as liver markers were downregulated. Insulin content was increased by up to 33-fold, to approximately 60% of the insulin content of normal beta-cells. The cells were shown to contain human C-peptide and release insulin in response to physiological glucose levels. Cell transplantation into immunodeficient diabetic mice resulted in the restoration of stable euglycemia. The cells continued to express insulin in vivo, and no cell replication was detected. Thus, the manipulation of culture conditions induced a significant and stable differentiation of FH-B-TPN cells toward the beta-cell phenotype, making them excellent candidates for beta-cell replacement in type 1 diabetes.


Subject(s)
Cell Differentiation/physiology , Hepatocytes/metabolism , Homeodomain Proteins/metabolism , Insulin/metabolism , Stem Cells/physiology , Trans-Activators/metabolism , Activins/pharmacology , Betacellulin , Exenatide , Gene Expression Regulation , Homeobox Protein Nkx-2.2 , Humans , Inhibin-beta Subunits/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Niacinamide/pharmacology , Nuclear Proteins , Peptides/pharmacology , Phenotype , Transcription Factors , Venoms/pharmacology
17.
Proc Natl Acad Sci U S A ; 100(12): 7253-8, 2003 Jun 10.
Article in English | MEDLINE | ID: mdl-12756298

ABSTRACT

Beta-cell replacement is considered to be the most promising approach for treatment of type 1 diabetes. Its application on a large scale is hindered by a shortage of cells for transplantation. Activation of insulin expression, storage, and regulated secretion in stem/progenitor cells offers novel ways to overcome this shortage. We explored whether fetal human progenitor liver cells (FH) could be induced to differentiate into insulin-producing cells after expression of the pancreatic duodenal homeobox 1 (Pdx1) gene, which is a key regulator of pancreatic development and insulin expression in beta cells. FH cells possess a considerable replication capacity, and this was further extended by introduction of the gene for the catalytic subunit of human telomerase. Immortalized FH cells expressing Pdx1 activated multiple beta-cell genes, produced and stored considerable amounts of insulin, and released insulin in a regulated manner in response to glucose. When transplanted into hyperglycemic immunodeficient mice, the cells restored and maintained euglycemia for prolonged periods. Quantitation of human C-peptide in the mouse serum confirmed that the glycemia was normalized by the transplanted human cells. This approach offers the potential of a novel source of cells for transplantation into patients with type 1 diabetes.


Subject(s)
Hepatocytes/transplantation , Homeodomain Proteins , Hyperglycemia/therapy , Insulin/biosynthesis , Stem Cell Transplantation/methods , Animals , Blood Glucose/metabolism , C-Peptide/blood , Cell Differentiation , DNA-Binding Proteins , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/therapy , Female , Gene Expression , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Hyperglycemia/blood , Mice , Mice, Inbred NOD , Mice, SCID , Stem Cells/cytology , Stem Cells/metabolism , Telomerase/genetics , Trans-Activators/genetics , Transduction, Genetic , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...