Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cell ; 177(4): 910-924.e22, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30982595

ABSTRACT

The assembly of organized colonies is the earliest manifestation in the derivation or induction of pluripotency in vitro. However, the necessity and origin of this assemblance is unknown. Here, we identify human pluripotent founder cells (hPFCs) that initiate, as well as preserve and establish, pluripotent stem cell (PSC) cultures. PFCs are marked by N-cadherin expression (NCAD+) and reside exclusively at the colony boundary of primate PSCs. As demonstrated by functional analysis, hPFCs harbor the clonogenic capacity of PSC cultures and emerge prior to commitment events or phenotypes associated with pluripotent reprogramming. Comparative single-cell analysis with pre- and post-implantation primate embryos revealed hPFCs share hallmark properties with primitive endoderm (PrE) and can be regulated by non-canonical Wnt signaling. Uniquely informed by primate embryo organization in vivo, our study defines a subset of founder cells critical to the establishment pluripotent state.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cell Differentiation , Cell Lineage , Embryonic Development , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Single-Cell Analysis , Wnt Signaling Pathway
2.
Stem Cells ; 35(9): 2095-2102, 2017 09.
Article in English | MEDLINE | ID: mdl-28758276

ABSTRACT

Induced pluripotent stem cell reprogramming has provided critical insights into disease processes by modeling the genetics and related clinical pathophysiology. Human cancer represents highly diverse genetics, as well as inter- and intra-patient heterogeneity, where cellular model systems capable of capturing this disease complexity would be invaluable. Acute myeloid leukemia (AML) represents one of most heterogeneous cancers and has been divided into genetic subtypes correlated with unique risk stratification over the decades. Here, we report our efforts to induce pluripotency from the heterogeneous population of human patients that represents this disease in the clinic. Using robust optimized reprogramming methods, we demonstrate that reprogramming of AML cells harboring leukemic genomic aberrations is a rare event with the exception of those with de novo mixed-lineage leukemia (MLL) mutations that can be reprogrammed and model drug responses in vitro. Our findings indicate that unlike hematopoietic cells devoid of genomic aberrations, AML cells harboring driver mutations are refractory to reprogramming. Expression of MLL fusion proteins in AML cells did not contribute to induced reprogramming success, which continued to select for patient derived cells devoid of AML patient-specific aberrations. Our study reveals that unanticipated blockades to achieving pluripotency reside within the majority of transformed AML patient cells. Stem Cells 2017;35:2095-2102.


Subject(s)
Cellular Reprogramming , Hematopoiesis , Induced Pluripotent Stem Cells/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation/genetics , Bone Marrow Cells/pathology , Humans , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism
3.
Int J Mol Sci ; 18(9)2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28902128

ABSTRACT

Evaluation of the extent and nature of induced pluripotent stem cell (iPSC) genetic instability is important for both basic research and future clinical use. As previously demonstrated regarding embryonic stem cells, such DNA aberrations might affect the differentiation capacity of the cells and increase their tumorigenicity. Here, we first focus on the contribution of multiple DNA damage response pathways during cellular reprogramming. We then discuss the origin and mechanisms responsible for the modification of genetic material in iPSCs (pre-existing variations in somatic cells, mutations induced by reprogramming factors, and mutations induced by culture expansion) and deepen the possible functional consequences of genetic variations in these cells. Lastly, we present some recent improvements of iPSC generation methods aimed at obtaining cells with fewer genetic variations.


Subject(s)
Cellular Reprogramming/physiology , Genomic Instability , Induced Pluripotent Stem Cells/physiology , Cellular Reprogramming Techniques/methods , Chromosomal Instability , DNA Damage/physiology , DNA Repair/physiology , Gene Dosage , Genetic Variation , Humans , Mutation , Oxidative Stress/physiology
4.
Stem Cells ; 30(7): 1414-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22628289

ABSTRACT

Phosphorylation of histone H2AX (γH2AX) is known to be the earliest indicator of DNA double-strand breaks. Recently, it has been shown that mouse embryonic stem cells (mESCs) have very high basal levels of γH2AX, even when they have not been exposed to genotoxic agents. As the specialized role of high basal γH2AX levels in pluripotent stem cells is still debated, we investigated whether H2AX phosphorylation is important in maintaining self-renewal of these cells. Here, we report that not only mESCs but also mouse-induced pluripotent stem cells (miPSCs), have high basal levels of γH2AX. We show that basal γH2AX levels decrease upon ESC and iPSC differentiation and increase when the cells are treated with self-renewal-enhancing small molecules. We observe that self-renewal activity is highly compromised in H2AX-/- cells and that it can be restored in these cells through reconstitution with a wild-type, but not a phospho-mutated, H2AX construct. Taken together, our findings suggest a novel function of H2AX that expands the knowledge of this histone variant beyond its role in DNA damage and into a new specialized biological function in mouse pluripotent stem cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Histones/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , DNA Damage/genetics , Flow Cytometry , Histones/genetics , Mice , Microscopy, Confocal , Phosphorylation
5.
Int J Mol Sci ; 14(2): 2617-36, 2013 Jan 28.
Article in English | MEDLINE | ID: mdl-23358251

ABSTRACT

Recent studies have shown that mouse embryonic stem cells (mESCs) rely on a distinctive genome caretaking network. In this review, we will discuss how mESCs functionally respond to DNA damage and describe several modifications in mESC DNA damage response, which accommodate dynamic cycling and preservation of genetic information. Subsequently, we will discuss how the transition from mESCs to adult stem/progenitor cells can be involved in the decline of tissue integrity and function in the elderly.

6.
Cell Chem Biol ; 30(7): 780-794.e8, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37379846

ABSTRACT

Overlapping principles of embryonic and tumor biology have been described, with recent multi-omics campaigns uncovering shared molecular profiles between human pluripotent stem cells (hPSCs) and adult tumors. Here, using a chemical genomic approach, we provide biological evidence that early germ layer fate decisions of hPSCs reveal targets of human cancers. Single-cell deconstruction of hPSCs-defined subsets that share transcriptional patterns with transformed adult tissues. Chemical screening using a unique germ layer specification assay for hPSCs identified drugs that enriched for compounds that selectively suppressed the growth of patient-derived tumors corresponding exclusively to their germ layer origin. Transcriptional response of hPSCs to germ layer inducing drugs could be used to identify targets capable of regulating hPSC specification as well as inhibiting adult tumors. Our study demonstrates properties of adult tumors converge with hPSCs drug induced differentiation in a germ layer specific manner, thereby expanding our understanding of cancer stemness and pluripotency.


Subject(s)
Neoplasms , Pluripotent Stem Cells , Humans , Cell Differentiation/physiology , Neoplasms/drug therapy , Neoplasms/genetics , Genomics
7.
Small ; 8(20): 3192-200, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-22821625

ABSTRACT

Highly bright and photostable cyanine dye-doped silica nanoparticles, IRIS Dots, are developed, which can efficiently label human mesenchymal stem cells (hMSCs). The application procedure used to label hMSCs is fast (2 h), the concentration of IRIS Dots for efficient labeling is low (20 µg mL(-1) ), and the labeled cells can be visualized by flow cytometry, confocal microscopy, and transmission electron microscopy. Labeled hMSCs are unaffected in their viability and proliferation, as well as stemness surface marker expression and differentiation capability into osteocytes. Moreover, this is the first report that shows nonfunctionalized IRIS Dots can discriminate between live and early-stage apoptotic stem cells (both mesenchymal and embryonic) through a distinct external cell surface distribution. On the basis of biocompatibility, efficient labeling, and apoptotic discrimination potential, it is suggested that IRIS Dots can serve as a promising stem cell tracking agent.


Subject(s)
Nanoparticles/chemistry , Optical Imaging/methods , Silicon Dioxide/chemistry , Stem Cells , Apoptosis/physiology , Cells, Cultured , Flow Cytometry , Humans , Microscopy, Confocal
8.
Drug Discov Today ; 27(12): 103407, 2022 12.
Article in English | MEDLINE | ID: mdl-36243303

ABSTRACT

The discovery and development of effective drugs for cancer patients has seen limited success in the clinic from phase I trials onward. The high attrition rate of current drug development approaches requires careful evaluation to provide a better understanding of the factors that correlate with or predict positive clinical outcomes. Here, we examine pre-clinical drug development approaches and conduct a meta-analysis of 2918 clinical studies involving 466 unique drugs tested in clinical trials for acute myeloid leukemia (AML). Our goal was to determine whether there are key shared pre-clinical characteristics that ultimately relate to successful or unsuccessful drugs in patients. We provide an evidence-based recommendation for the use of phenotypic drug discovery rather than other methods during pre-clinical development. Although our analysis was limited to AML, similar analyses are likely to be informative for other tumor-specific drug discovery campaigns, informing and improving the foundational discovery screens and platforms for other cancers.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/drug therapy , Drug Discovery
9.
F1000Res ; 10: 964, 2021.
Article in English | MEDLINE | ID: mdl-34909193

ABSTRACT

Data from the Istituto Superiore di Sanità (ISS) emphasized by the media indicate that COVID-19 vaccination reduces related infections, hospitalizations and deaths. However, a comparison showed significantly more hospitalizations and intensive care unit accesses in the corresponding months and days in 2021 versus 2020 and no significant differences in deaths. The combination of non-alternative hypotheses may help explain the discrepancy between the results in the entire population and the vaccination's success claimed by the ISS in reducing infections, serious cases and deaths: a bias: counting as unvaccinated also "those vaccinated with 1 dose in the two weeks following the inoculation", and as incompletely vaccinated also "those vaccinated with 2 doses within two weeks of the 2nd inoculation".a systematic error: counting as unvaccinated also "vaccinated with 1 dose in the two weeks following the inoculation", and as incompletely vaccinated also "vaccinated with 2 doses within two weeks of the 2nd inoculation". Many reports show an increase in COVID-19 cases in these time-windows, and related data should be separated levels of protective effectiveness in vaccinated people, often considered stable, actually show signs of progressive reduction over time, which could contribute to reducing the overall population resultunvaccinated people show more severe disease than in 2020, supporting also in humans the theory of imperfect vaccines, which offer less resistance to the entry of germs than the resistance later encountered inside the human body. This favors the selection of more resistant and virulent mutants, that can be spread by vaccinated people. This damages first the unvaccinated people, but ultimately the whole community. An open scientific debate is needed to discuss these hypotheses, following the available evidence (as well as to discuss the inconsistent theory of unvaccinated young people as reservoirs of viruses/mutants), to assess the long-term and community impact of different vaccination strategies.


Subject(s)
COVID-19 , Adolescent , COVID-19 Vaccines , Hospitalization , Humans , SARS-CoV-2 , Vaccination
10.
Cell Rep ; 34(10): 108818, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691101

ABSTRACT

Histone variants (HVs) are a subfamily of epigenetic regulators implicated in embryonic development, but their role in human stem cell fate remains unclear. Here, we reveal that the phosphorylation state of the HV H2A.X (γH2A.X) regulates self-renewal and differentiation of human pluripotent stem cells (hPSCs) and leukemic progenitors. As demonstrated by CRISPR-Cas deletion, H2A.X is essential in maintaining normal hPSC behavior. However, reduced levels of γH2A.X enhances hPSC differentiation toward the hematopoietic lineage with concomitant inhibition of neural development. In contrast, activation and sustained levels of phosphorylated H2A.X enhance hPSC neural fate while suppressing hematopoiesis. This controlled lineage bias correlates to occupancy of γH2A.X at genomic loci associated with ectoderm versus mesoderm specification. Finally, drug modulation of H2A.X phosphorylation overcomes differentiation block of patient-derived leukemic progenitors. Our study demonstrates HVs may serve to regulate pluripotent cell fate and that this biology could be extended to somatic cancer stem cell control.


Subject(s)
Cell Self Renewal/physiology , Histones/metabolism , Neoplastic Stem Cells/cytology , Pluripotent Stem Cells/cytology , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Lineage , Ectoderm/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/deficiency , Histones/genetics , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mesoderm/metabolism , Neoplastic Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Nucleosomes/metabolism , Phosphorylation , Pluripotent Stem Cells/metabolism
11.
Cell Rep ; 34(11): 108845, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33730576

ABSTRACT

Identifying precise targets of individual cancers remains challenging. Chronic lymphocytic leukemia (CLL) represents the most common adult hematologic malignancy, and trisomy 12 (tri12) represents a quarter of CLL patients. We report that tri12 human pluripotent stem cells (hPSCs) allow for the identification of gene networks and targets specific to tri12, which are controlled by comparative normal PSCs. Identified targets are upregulated in tri12 leukemic cells from a cohort of 159 patients with monoclonal B cell lymphocytosis and CLL. tri12 signaling patterns significantly influence progression-free survival. Actionable targets are identified using high-content drug testing and functionally validated in an additional 44 CLL patient samples. Using xenograft models, interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitor is potent and selective against human tri12 CLL versus healthy patient-derived xenografts. Our study uses hPSCs to uncover targets from genetic aberrations and apply them to cancer. These findings provide immediate translational potential as biomarkers and targets for therapeutic intervention.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Pluripotent Stem Cells/metabolism , Trisomy/genetics , Adult , Aged , Aged, 80 and over , Animals , Cell Line , Disease Progression , Female , Gene Dosage , Gene Regulatory Networks , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mice, Inbred NOD , Middle Aged , Models, Genetic , Reproducibility of Results , Xenograft Model Antitumor Assays
12.
Cell Chem Biol ; 28(10): 1394-1406.e10, 2021 10 21.
Article in English | MEDLINE | ID: mdl-33979648

ABSTRACT

Natural products (NPs) encompass a rich source of bioactive chemical entities. Here, we used human cancer stem cells (CSCs) in a chemical genomics campaign with NP chemical space to interrogate extracts from diverse strains of actinomycete for anti-cancer properties. We identified a compound (McM25044) capable of selectively inhibiting human CSC function versus normal stem cell counterparts. Biochemical and molecular studies revealed that McM025044 exerts inhibition on human CSCs through the small ubiquitin-like modifier (SUMO) cascade, found to be hyperactive in a variety of human cancers. McM025044 impedes the SUMOylation pathway via direct targeting of the SAE1/2 complex. Treatment of patient-derived CSCs resulted in reduced levels of SUMOylated proteins and suppression of progenitor and stem cell capacity measured in vitro and in vivo. Our study overcomes a barrier in chemically inhibiting oncogenic SUMOylation activity and uncovers a unique role for SAE2 in the biology of human cancers.


Subject(s)
Neoplastic Stem Cells/metabolism , Ubiquitin-Activating Enzymes/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Biological Products/therapeutic use , Cell Line, Tumor , Cell Self Renewal , Cell Survival/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mice , Molecular Docking Simulation , Neoplastic Stem Cells/cytology , RNA Interference , RNA, Small Interfering/metabolism , Sumoylation/drug effects , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics
13.
BMC Cancer ; 9: 281, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19674456

ABSTRACT

BACKGROUND: Current chemotherapy of human cancers focuses on the DNA damage pathway to induce a p53-mediated cellular response leading to either G1 arrest or apoptosis. However, genotoxic treatments may induce mutations and translocations that result in secondary malignancies or recurrent disease. In addition, about 50% of human cancers are associated with mutations in the p53 gene. Nongenotoxic activation of apoptosis by targeting specific molecular pathways thus provides an attractive therapeutic approach. METHODS: Normal and leukemic cells were evaluated for their sensitivity to 5, 6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB) through cell viability and caspase activation tests. The apoptotic pathway induced by DRB was analysed by immunfluorescence and immunoblot analysis. H2AX phosphorylation and cell cycle analysis were performed to study the dependance of apoptosis on DNA damage and DNA replication, respectively. To investigate the role of p53 in DRB-induced apoptosis, specific p53 inhibitors were used. Statistical analysis on cell survival was performed with the test of independence. RESULTS: Here we report that DRB, an inhibitor of the transcriptional cyclin-dependent kinases (CDKs) 7 and 9, triggers DNA replication-independent apoptosis in normal and leukemic human cells regardless of their p53 status and without inducing DNA damage. Our data indicate that (i) in p53-competent cells, apoptosis induced by DRB relies on a cytosolic accumulation of p53 and subsequent Bax activation, (ii) in the absence of p53, it may rely on p73, and (iii) it is independent of ATM and NBS1 proteins. Notably, even apoptosis-resistant leukemic cells such as Raji were sensitive to DRB. CONCLUSION: Our results indicate that DRB represents a potentially useful cancer chemotherapeutic strategy that employs both the p53-dependent and -independent apoptotic pathways without inducing genotoxic stress, thereby decreasing the risk of secondary malignancies.


Subject(s)
Apoptosis/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA Replication/drug effects , Dichlororibofuranosylbenzimidazole/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cells, Cultured , Humans , Leukemia/drug therapy , Leukemia/genetics , Leukemia/metabolism , Leukemia/physiopathology , Lymphocytes/cytology , Lymphocytes/drug effects , Tumor Suppressor Protein p53/genetics
14.
Mol Immunol ; 45(2): 328-37, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17659780

ABSTRACT

V(D)J recombination is a mechanism peculiar to the somatic rearrangement of antigen receptor genes. It requires both expression of the RAG-1 and RAG-2 recombinases and accessibility of the substrate to its recombinase and post-cleavage/DNA repair stage. TCR revision is a genetic correction mechanism that changes T cell specificity by re-activating V(D)J recombination in peripheral T cells. This process is now well described in both normal or pathological murine and human settings. Many of its features, such as the question of whether it occurs in truly mature T cells, remain to be elucidated. Its occurrence in human CD8+ T cells is also an open question. We have therefore established an in vitro model of TCR revision in mature human CD8+ T cells to determine whether down-regulation of the TCR/CD3 complex from the cell surface in the presence of IL7 as a factor favouring chromatin remodelling initiates a TCR revision pathway. Only mature CD8+ T cells carrying already-formed antigen receptors were used. CD8+ T cells treated with anti-CD3 and IL7 showed rearrangement intermediates and expressed new Vbeta-chains on their surface. Investigation of the molecular pathway thus induced disclosed up-regulation of the RAG-2 transcript, but absence of the 'canonical' RAG-1 mRNA. A surprising finding was the demonstration of alternative splice forms of this mRNA, already expressed in untreated CD8+ T cells, encoding for the full-length RAG-1 protein, which was increased three-fold in the treated cells. All the V(D)J requirements were thus fulfilled when mature human CD8+ T cells were stimulated with anti-CD3 and IL7. Induction of TCR revision in vitro in mature T cells is an easily controllable system that could be employed in further studies to elucidate the molecular pathways involved in secondary V(D)J rearrangements in peripheral cells.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Models, Immunological , Receptors, Antigen, T-Cell/immunology , Antibodies/pharmacology , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/drug effects , Cell Line , Clone Cells , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Enterotoxins/pharmacology , Fluorescent Antibody Technique , Gene Rearrangement, T-Lymphocyte/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Interleukin-7/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology
15.
Cytometry A ; 73(6): 508-16, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18431795

ABSTRACT

Ataxia telangiectasia (A-T) is a progressive neurodegenerative disease with onset in early childhood, caused by mutations in the ATM (ataxia-telangiectasia mutated) gene. Diagnosis relies on laboratory tests showing high levels of serum alphafetoprotein, cell sensitivity to ionizing radiation (IR) and absence or reduced levels of ATM protein. Many tests, however, are not sufficiently sensitive or specific for A-T, have long turnaround times, or require large blood samples. This prompted us to develop a new flow cytometry method for the diagnosis of A-T based on the measurement of histone H2AX phosphorylation. We established normal ranges of histone H2AX phosphorylation after 2 Gy IR by testing T-cell lines, lymphoblastoid cell lines (LCLs) and/or peripheral blood mononuclear cells (PBMCs) or both from 20 genetically proven A-T and 46 control donors. To further evaluate the specificity and sensitivity of the test, we analyzed cells from 19 patients suspected of having A-T, and from one Friedreich Ataxia, one Ataxia with Oculomotor Apraxia type 2, and one Nijmegen Breakage Syndrome patients. Phosphorylated histone H2AX mean fluorescence intensity of irradiated A-T cells was significantly lower than that of healthy donors. The intrastaining, intraassay, and interassay imprecisions were

Subject(s)
Ataxia Telangiectasia/diagnosis , Flow Cytometry/methods , Histones/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Diagnosis, Differential , Histones/radiation effects , Humans , Immunoblotting , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/radiation effects , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Tumor Suppressor Proteins/genetics
16.
Cell Chem Biol ; 24(7): 833-844.e9, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28648376

ABSTRACT

Targeting of human cancer stem cells (CSCs) requires the identification of vulnerabilities unique to CSCs versus healthy resident stem cells (SCs). Unfortunately, dysregulated pathways that support transformed CSCs, such as Wnt/ß-catenin signaling, are also critical regulators of healthy SCs. Using the ICG-001 and CWP family of small molecules, we reveal Sam68 as a previously unappreciated modulator of Wnt/ß-catenin signaling within CSCs. Disruption of CBP-ß-catenin interaction via ICG-001/CWP induces the formation of a Sam68-CBP complex in CSCs that alters Wnt signaling toward apoptosis and differentiation induction. Our study identifies Sam68 as a regulator of human CSC vulnerability.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA-Binding Proteins/metabolism , Neoplastic Stem Cells/metabolism , Peptide Fragments/metabolism , RNA-Binding Proteins/metabolism , Sialoglycoproteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Animals , Apoptosis/drug effects , Azabicyclo Compounds/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Middle Aged , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/transplantation , Organophosphates/pharmacology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidinones/pharmacology , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Sialoglycoproteins/antagonists & inhibitors , Sialoglycoproteins/genetics , Sumoylation/drug effects , Transcriptome/drug effects , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
17.
Stem Cell Res ; 15(1): 240-2, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26141785

ABSTRACT

The combination of OCT4 expression and short-term exposure to reprogramming media induces a state of transcriptional plasticity in human fibroblasts, capable of responding to changes in the extracellular environment. Here we provide characterization of iPSCs established through continued culture of OCT4-induced plastic human fibroblasts in pluripotent-supportive reprogramming media. Human iPSC(OCT4) are morphologically indistinguishable from conventionally derived iPSCs and express core proteins involved in maintenance of pluripotency. iPSC(OCT4) display bona fide functional pluripotency as measured by in vivo teratoma formation consisting of the three germ layers.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/pharmacology , Adult , Animals , Cellular Reprogramming/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Mice, Inbred NOD , Mice, SCID
18.
Stem Cell Res ; 15(1): 221-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26117529

ABSTRACT

The combination of OCT4 expression and short-term exposure to reprogramming media induces a state of transcriptional plasticity in human fibroblasts, capable of responding to changes in the extracellular environment that facilitate direct cell fate conversion toward lineage specific progenitors. Here we reveal that continued exposure of OCT4-induced plastic human fibroblasts to reprogramming media (RM) is sufficient to induce pluripotency. OCT4-derived induced pluripotent stem cell (iPSC(OCT4)) colonies emerged after prolonged culture in RM, and formed independently of lineage specific progenitors. Human iPSC(OCT4) are morphologically indistinguishable from conventionally derived iPSCs and express core proteins involved in maintenance of pluripotency. iPSC(OCT4) display in vivo functional pluripotency as measured by teratoma formation consisting of the three germ layers, and are capable of targeted in vitro differentiation. Our study indicates that acquisition of pluripotency is one of multiple cell fate choices that can be facilitated through environmental stimulation of OCT4-induced plasticity, and suggests the role of other reprogramming factors to induce pluripotency can be substituted by prolonged culture of plastic fibroblasts.


Subject(s)
Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/pharmacology , Adult , Animals , Cell Lineage/drug effects , Fibroblasts/drug effects , Humans , Immunophenotyping , Mice, Inbred NOD , Mice, SCID , Models, Biological
19.
PLoS One ; 7(1): e30234, 2012.
Article in English | MEDLINE | ID: mdl-22291922

ABSTRACT

The ability to reprogram somatic cells to induced pluripotent stem cells (iPSCs), exhibiting properties similar to those of embryonic stem cells (ESCs), has attracted much attention, with many studies focused on improving efficiency of derivation and unraveling the mechanisms of reprogramming. Despite this widespread interest, our knowledge of the molecular signaling pathways that are active in iPSCs and that play a role in controlling their fate have not been studied in detail. To address this shortfall, we have characterized the influence of different signals on the behavior of a model mouse iPSC line. We demonstrate significant responses of this iPSC line to the presence of serum, which leads to profoundly enhanced proliferation and, depending on the medium used, a reduction in the capacity of the iPSCs to self-renew. Surprisingly, this iPSC line was less sensitive to withdrawal of LIF compared to ESCs, exemplified by maintenance of expression of a Nanog-GFP reporter and enhanced self-renewal in the absence of LIF. While inhibition of phosphoinositide-3 kinase (PI3K) signaling decreased iPSC self-renewal, inhibition of Gsk-3 promoted it, even in the absence of LIF. High passages of this iPSC line displayed altered characteristics, including genetic instability and a reduced ability to self-renew. However, this second feature could be restored upon inhibition of Gsk-3. Collectively, our data suggest modulation of Gsk-3 activity plays a key role in the control of iPSC fate. We propose that more careful consideration should be given to characterization of the molecular pathways that control the fate of different iPSC lines, since perturbations from those observed in naïve pluripotent ESCs could render iPSCs and their derivatives susceptible to aberrant and potentially undesirable behaviors.


Subject(s)
Cell Proliferation , Induced Pluripotent Stem Cells/physiology , Signal Transduction/physiology , Animals , Cell Culture Techniques , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Cellular Reprogramming/drug effects , Cellular Reprogramming/physiology , Culture Media/pharmacology , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3/physiology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Leukemia Inhibitory Factor/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors
20.
Mol Immunol ; 48(12-13): 1369-76, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21481940

ABSTRACT

Induction of the TCR signaling pathway terminates the expression of RAG genes, and a link between this pathway and their transcriptional control is evident from the recent demonstration of their re-expression if the TCR is subsequently lost or down-regulated. Since unstimulated T cells display a steady-state level of "tonic" TCR signaling, i.e. in the absence of any antigenic stimulus, it was uncertain whether this control was exerted through ligand-dependent or ligand-independent TCR signaling. Here we demonstrate for the first time that exogenous TCR α and ß chains transferred into the human immature RAG(+) T cell line Sup-T1 by lentiviral transduction inhibit RAG expression through tonic signaling, and that this inhibition could itself be reverted by pharmacological tonic pathway inhibitors. We also suggest that mature T cells already expressing an endogenous TCR on their surface maintain some levels of plasticity at the RAG locus when their basal TCR signaling is interfered with. Lastly, we show that the TCR constructs employed in TCR gene therapy do not possess the same basal signaling transduction capability, a feature that may have therapeutic implications.


Subject(s)
DNA-Binding Proteins/genetics , Genes, RAG-1 , Nuclear Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Benzamides , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Child , DNA-Binding Proteins/metabolism , Dimethyl Sulfoxide/pharmacology , Down-Regulation , Flow Cytometry , Fluorescent Antibody Technique , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Imatinib Mesylate , Immunoblotting , Jurkat Cells , Ligands , Nuclear Proteins/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Reverse Transcriptase Polymerase Chain Reaction , Tacrolimus/pharmacology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL