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










Publication year range
1.
Int J Mol Sci ; 20(7)2019 Apr 08.
Article in English | MEDLINE | ID: mdl-30965622

ABSTRACT

Although human pluripotent stem cells (hPSCs) can theoretically differentiate into any cell type, their ability to produce hematopoietic cells is highly variable from one cell line to another. The underlying mechanisms of this heterogeneity are not clearly understood. Here, using a whole miRNome analysis approach in hPSCs, we discovered that their hematopoietic competency was associated with the expression of several miRNAs and conversely correlated to that of miR-206 specifically. Lentiviral-based miR-206 ectopic expression in H1 hematopoietic competent embryonic stem (ES) cells markedly impaired their differentiation toward the blood lineage. Integrative bioinformatics identified a potential miR-206 target gene network which included hematopoietic master regulators RUNX1 and TAL1. This work sheds light on the critical role of miR-206 in the generation of blood cells off hPSCs. Our results pave the way for future genetic manipulation of hPSCs aimed at increasing their blood regenerative potential and designing better protocols for the generation of bona fide hPSC-derived hematopoietic stem cells.


Subject(s)
MicroRNAs/metabolism , Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Cell Line , Cell Lineage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Pluripotent Stem Cells/metabolism
2.
Stem Cell Res ; 26: 8-16, 2018 01.
Article in English | MEDLINE | ID: mdl-29197744

ABSTRACT

MEN2A is a hereditary cancer-predisposing syndrome that affects patients with germline RET mutations. The effects of this oncogenic tyrosine kinase in the context of primitive stem cells are not known. In order to study these events, we generated a MEN2A induced Pluripotent Stem Cell (iPSC) line from a patient with RET mutation and an isogenic counterpart by CRISPR-Cas9 correction of the mutation. Whole exome sequencing of iPSC before and after CRISPR-Cas9 genome edition revealed no major exonic off target effect of the CRISPR correction. However, an integrative differential gene expression analysis of iPSC with oncogenic RETC634Y and its gene-corrected iPSC with RETY634C as well as RETwt iPSCs revealed activation of the Early Growth Response 1 (EGR1) transcriptional program in RET-mutated iPSC, a pathway shown to be involved in RET-induced oncogenesis. These data constitute the first proof of concept of the feasibility of the use of an iPSC and its genome-corrected counterpart to unravel the molecular mechanisms underlying the development of the hereditary MEN2A cancer predisposing syndrome.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Early Growth Response Protein 1/genetics , Induced Pluripotent Stem Cells/pathology , Multiple Endocrine Neoplasia Type 2a/genetics , Mutation , Proto-Oncogene Proteins c-ret/genetics , Transcriptome , Adult , Genome, Human , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Multiple Endocrine Neoplasia Type 2a/pathology , Multiple Endocrine Neoplasia Type 2a/prevention & control , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Tumor Cells, Cultured
3.
Sci Rep ; 7(1): 15567, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29138439

ABSTRACT

Intracellular pathogenic microorganisms and toxins exploit host cell mechanisms to enter, exert their deleterious effects as well as hijack host nutrition for their development. A potential approach to treat multiple pathogen infections and that should not induce drug resistance is the use of small molecules that target host components. We identified the compound 1-adamantyl (5-bromo-2-methoxybenzyl) amine (ABMA) from a cell-based high throughput screening for its capacity to protect human cells and mice against ricin toxin without toxicity. This compound efficiently protects cells against various toxins and pathogens including viruses, intracellular bacteria and parasite. ABMA provokes Rab7-positive late endosomal compartment accumulation in mammalian cells without affecting other organelles (early endosomes, lysosomes, the Golgi apparatus, the endoplasmic reticulum or the nucleus). As the mechanism of action of ABMA is restricted to host-endosomal compartments, it reduces cell infection by pathogens that depend on this pathway to invade cells. ABMA may represent a novel class of broad-spectrum compounds with therapeutic potential against diverse severe infectious diseases.


Subject(s)
Adamantane/analogs & derivatives , Benzyl Compounds/pharmacology , Endosomes/drug effects , Ricin/antagonists & inhibitors , Toxins, Biological/antagonists & inhibitors , Adamantane/chemistry , Adamantane/pharmacology , Animals , Benzyl Compounds/chemistry , Benzylamines , Cell Compartmentation/drug effects , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , HeLa Cells , Humans , Lysosomes/drug effects , Mice , Ricin/drug effects , Ricin/toxicity , Toxins, Biological/chemistry , Toxins, Biological/toxicity
4.
Exp Hematol ; 53: 48-58, 2017 09.
Article in English | MEDLINE | ID: mdl-28602946

ABSTRACT

We report here the first use of whole-genome sequencing (WGS) to examine the initial clonal dynamics in an unusual patient with chronic myeloid leukemia (CML), who presented in chronic phase (CP) with doubly marked BCR-ABL1+/JAK2V617F-mutant cells and, over a 9-year period, progressed into an accelerated phase (AP) and then terminal blast phase (BP). WGS revealed that the diagnostic cells also contained mutations in ASXL1, SEC23B, MAD1L1, and RREB1 as well as 12,000 additional uncommon DNA variants. WGS of endothelial cells generated from circulating precursors revealed many of these were shared with the CML clone. Surprisingly, WGS of induced pluripotent stem cells (iPSCs) derived from the AP cells revealed only six additional coding somatic mutations, despite retention by the hematopoietic progeny of the parental AP cell levels of BCR-ABL1 expression and sensitivity to imatinib and pimozide. Limited analysis of BP cells revealed independent subclonal progression to homozygosity of the MAD1L1 and RREB1 variants. MAD1L1 and SEC23B mutations were also identified in 2 of 101 cases of myeloproliferative neoplasms, but not in 42 healthy subjects. These findings challenge historic concepts of clonal evolution in CML.


Subject(s)
Janus Kinase 2/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mutation , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Genome-Wide Association Study , Humans , Induced Pluripotent Stem Cells/physiology , Male , Middle Aged , Nuclear Proteins/genetics , Transcription Factors/genetics
5.
PLoS One ; 11(3): e0149291, 2016.
Article in English | MEDLINE | ID: mdl-26938212

ABSTRACT

Hematopoiesis generated from human embryonic stem cells (ES) and induced pluripotent stem cells (iPS) are unprecedented resources for cell therapy. We compared hematopoietic differentiation potentials from ES and iPS cell lines originated from various donors and derived them using integrative and non-integrative vectors. Significant differences in differentiation toward hematopoietic lineage were observed among ES and iPS. The ability of engraftment of iPS or ES-derived cells in NOG mice varied among the lines with low levels of chimerism. iPS generated from ES cell-derived mesenchymal stem cells (MSC) reproduce a similar hematopoietic outcome compared to their parental ES cell line. We were not able to identify any specific hematopoietic transcription factors that allow to distinguish between good versus poor hematopoiesis in undifferentiated ES or iPS cell lines. There is a relatively unpredictable variation in hematopoietic differentiation between ES and iPS cell lines that could not be predicted based on phenotype or gene expression of the undifferentiated cells. These results demonstrate the influence of genetic background in variation of hematopoietic potential rather than the reprogramming process.


Subject(s)
Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Genetic Heterogeneity , Graft Survival , Hematopoiesis/genetics , Induced Pluripotent Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cell Lineage/genetics , Chimerism , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Gene Expression , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Lentivirus/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Retroviridae/genetics , Tissue Donors , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous
6.
Mol Cancer Ther ; 14(1): 213-24, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25492619

ABSTRACT

Midkine (MDK) is a member of a new family of neurotrophic factors considered as rate-limiting growth and angiogenic factors implicated in the onset, invasion, and metastatic process of neuronal tumors, including neuroblastoma. We showed that all neuroblastoma cell lines highly expressed MDK, indicating that it is a critical player in tumor development, which may henceforth represent an attractive therapeutic target. We showed that the knockdown of MDK expression by siRNA led to a marked and significant decrease in neuroblastoma (IGR-N91 and SH-SY5Y) cell proliferation in vitro. Using a new strategy, we then evaluated the antitumor effect of a truncated MDK protein, lacking the C-terminal 81-121 portion of the molecule (MDKΔ81-121), which may act as a dominant-negative effector for its mitogenic, angiogenic, and tumorigenic activities by heterodimerizing with the wild-type protein. In vitro studies showed that MDKΔ81-121 selectively inhibited MDK-dependent tumor cells and was able to strongly reduce endothelial cell proliferation and migration and to induce ER stress-mediated apoptosis. We then investigated the effects of MDKΔ81-121 in vivo using electrotransfer of a plasmid encoding a secretable form of MDKΔ81-121 into tibialis cranialis muscles of nude mice. We showed that MDKΔ81-121 dramatically inhibited (up to 91%) tumor development and growth. This inhibition was correlated with the detection of the MDKΔ81-121 molecule in plasma and the suppression of intratumor neovascularization. Our findings demonstrate that MDK inhibition is a tractable therapeutic target for this lethal pediatric malignancy.


Subject(s)
Cytokines/genetics , Cytokines/pharmacology , Neovascularization, Pathologic/drug therapy , Neuroblastoma/therapy , Animals , COS Cells , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Chlorocebus aethiops , Coculture Techniques , Cytokines/metabolism , Gene Knockdown Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Midkine , Neuroblastoma/blood supply , Plasmids/genetics
7.
Biores Open Access ; 3(5): 206-16, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25371857

ABSTRACT

The fine analysis of cell components during the generation of pluripotent cells and their comparison to bone fide human embryonic stem cells (hESCs) are valuable tools to understand their biological behavior. In this report, human mesenchymal cells (hMSCs) generated from the human ES cell line H9, were reprogrammed back to induced pluripotent state using Oct-4, Sox2, Nanog, and Lin28 transgenes. Human induced pluripotent stem cells (hIPSCs) were analyzed using electron microscopy and compared with regard to the original hESCs and the hMSCs from which they were derived. This analysis shows that hIPSCs and the original hESCs are morphologically undistinguishable but differ from the hMSCs with respect to the presence of several morphological features of undifferentiated cells at both the cytoplasmic (ribosomes, lipid droplets, glycogen, scarce reticulum) and nuclear levels (features of nuclear plasticity, presence of euchromatin, reticulated nucleoli). We show that hIPSC colonies generated this way presented epithelial aspects with specialized junctions highlighting morphological criteria of the mesenchymal-epithelial transition in cells engaged in a successful reprogramming process. Electron microscopic analysis revealed also specific morphological aspects of partially reprogrammed cells. These results highlight the valuable use of electron microscopy for a better knowledge of the morphological aspects of IPSC and cellular reprogramming.

8.
Stem Cells Dev ; 23(24): 2983-95, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-24955741

ABSTRACT

During human embryonic stem cell (ESC) hematopoietic differentiation, the description of the initial steps of lymphopoiesis remains elusive. Using a two-step culture procedure, we identified two original populations of ESC-derived hematopoietic progenitor cells (HPCs) with CD34(+)CD45RA(+)CD7(-) and CD34(+)CD45RA(+)CD7(+) phenotypes. Bulk cultures and limiting dilution assays, culture with MS5 cells in the presence of Notch ligand Delta-like-1 (DL-1), and ex vivo colonization tests using fetal thymic organ cultures showed that although CD34(+)CD45RA(+)CD7(-) HPCs could generate cells of the three lymphoid lineages, their potential was skewed toward the B cell lineages. In contrast, CD34(+)CD45RA(+)CD7(+) HPCs predominantly exhibited a T/natural killer (NK) cell differentiation potential. Furthermore these cells could differentiate equivalently into cells of the granulo-macrophagic lineage and dendritic cells and lacked erythroid potential. Expression profiling of 18 markers by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed that CD34(+)CD45RA(+)CD7(-) and CD34(+)CD45RA(+)CD7(+) HPCs express genes of the lymphoid specification and that CD34(+)CD45RA(+)CD7(-) cells express B-cell-associated genes, while CD34(+)CD45RA(+)CD7(+) HPCs display a T-cell molecular profile. Altogether, these findings indicate that CD34(+)CD45RA(+)CD7(-) and CD34(+)CD45RA(+)CD7(+) HPCs correspond to candidate multipotent early lymphoid progenitors polarized toward either the B or T/NK lineage, respectively. This work should improve our understanding of the early steps of lymphopoiesis from pluripotent stem cells and pave the way for the production of lymphocytes for cell-based immunotherapy and lymphoid development studies.


Subject(s)
Embryonic Stem Cells/cytology , Hematopoiesis , Lymphoid Progenitor Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Calcium-Binding Proteins , Cell Line , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lymphoid Progenitor Cells/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred NOD , Pluripotent Stem Cells/metabolism
9.
Am J Pathol ; 180(5): 2084-96, 2012 May.
Article in English | MEDLINE | ID: mdl-22425713

ABSTRACT

Because many of the genes used to produce induced pluripotent stem cells (iPSCs) from somatic cells are either outright established oncogenes, such as c-myc and Klf4, or potentially related to tumorigenesis in various cancers, both the safety and the risks of tumorigenesis linked to iPSC generation require evaluation. In this work, we generated, by lentivirus-mediated gene transfer of Oct4, Sox2, Nanog, and Lin28, two types of iPSCs from human mesenchymal stem cells and human amniotic fluid-derived cells: fully reprogrammed iPSCs with silencing of the four transgenes and partially reprogrammed iPSCs that still express one or several transgenes. We assessed the behavior of these cells during both their differentiation and proliferation using in vivo teratoma assays in nonobese diabetic mice with severe combined immunodeficiency. In contrast to fully reprogrammed iPSCs, 43% of partially reprogrammed iPSC cases (6 of 14 teratomas) generated major dysplasia and malignant tumors, with yolk sac tumors and embryonal carcinomas positive for α-fetoprotein, cytokeratin AE1/AE3, and CD30. This correlated with the expression of one or several transgenes used for the reprogramming, down-regulation of CDK 1A mRNA (p21/CDKN1A), and up-regulation of antiapoptotic Bcl-2 mRNA. Therefore, the oncogenicity of therapeutically valuable patient-specific iPSC-derived cells should be scrupulously evaluated before they are used for any clinical applications.


Subject(s)
Cell Transformation, Neoplastic/pathology , Induced Pluripotent Stem Cells/pathology , Ki-1 Antigen/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Animals , Cell Differentiation/physiology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cellular Reprogramming/physiology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Karyotype , Kruppel-Like Factor 4 , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Teratoma/metabolism , Teratoma/pathology , Transgenes/genetics
10.
Int J Oncol ; 38(1): 179-88, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21109939

ABSTRACT

We previously demonstrated, using the glioblastoma cell line U87MG as an experimental model, that the adenoviral mediated overexpression of the truncated protein HARPΔ111-136 inhibits the proliferation of these cells in vitro as well as tumor growth and angiogenesis in vivo. This study focused on identifying the underlying mechanisms for the observed antitumoral effect. The present study demonstrated that HARPΔ111-136 induced the ATF4/ATF3/CHOP cascade resulting in a strong expression of the proapoptotic protein CHOP, leading to tumor cell apoptosis as demonstrated by PARP cleavage and FACS analysis. siRNA-mediated CHOP gene silencing abolished Ad-HARPΔ111-136 induced apoptosis. Moreover, Ad-HARPΔ111-136 increased the expression of the death receptor DR5 and enhanced U87MG cells sensitivity in vitro to TRAIL a DR5 ligand with subsequent activation of caspase 8. Infection of U87MG cells with Ad-HARPΔ111-136 also enhanced radiation-induced apoptosis. In vivo, the combination of Ad-HARPΔ111-136 and radiation therapy resulted in a striking inhibition (92%) of the growth of U87MG xenografts, resulting from the potent effect on tumor angiogenesis and tumor cell apoptosis as determined by TUNEL analysis. Taken together, our results indicated that the inhibitor HARPΔ111-136 sensitized U87MG cells to apoptosis.


Subject(s)
Apoptosis/radiation effects , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , DNA Helicases/biosynthesis , Glioblastoma/pathology , Glioblastoma/radiotherapy , Peptide Fragments/biosynthesis , Animals , Apoptosis/physiology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , DNA Helicases/genetics , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Immunohistochemistry , Mice , Mice, Nude , Peptide Fragments/genetics , Transcription Factor CHOP , Transfection , Xenograft Model Antitumor Assays
11.
Cell ; 141(2): 231-42, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20403321

ABSTRACT

Bacterial Shiga-like toxins are virulence factors that constitute a significant public health threat worldwide, and the plant toxin ricin is a potential bioterror weapon. To gain access to their cytosolic target, ribosomal RNA, these toxins follow the retrograde transport route from the plasma membrane to the endoplasmic reticulum, via endosomes and the Golgi apparatus. Here, we used high-throughput screening to identify small molecule inhibitors that protect cells from ricin and Shiga-like toxins. We identified two compounds that selectively block retrograde toxin trafficking at the early endosome-TGN interface, without affecting compartment morphology, endogenous retrograde cargos, or other trafficking steps, demonstrating an unexpected degree of selectivity and lack of toxicity. In mice, one compound clearly protects from lethal nasal exposure to ricin. Our work discovers the first small molecule that shows efficacy against ricin in animal experiments and identifies the retrograde route as a potential therapeutic target.


Subject(s)
Benzamides/pharmacology , Benzodiazepinones/pharmacology , Cytoprotection , Protein Transport , Ricin/antagonists & inhibitors , Thiophenes/pharmacology , Administration, Intranasal , Animals , Benzamides/chemistry , Benzodiazepinones/chemistry , Cell Line, Tumor , Cell Membrane/metabolism , Endocytosis , Endosomes/metabolism , Golgi Apparatus/metabolism , HeLa Cells , High-Throughput Screening Assays , Humans , Mice , Qa-SNARE Proteins/metabolism , Ricin/administration & dosage , Ricin/toxicity , Shiga Toxins/antagonists & inhibitors , Shiga Toxins/toxicity , Thiophenes/chemistry , trans-Golgi Network/metabolism
12.
Int J Cancer ; 127(5): 1038-51, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20013808

ABSTRACT

Glioblastoma is the most common primary brain tumor in human adults. Since existing treatments are not effective enough, novel therapeutic targets must be sought. The heparin-binding growth factor, heparin affin regulatory peptide (HARP), also known as pleiotrophin (PTN), could potentially represent such a target. We have previously shown that a mutant protein, HARPDelta111-136, which lacks HARP's C-terminal 26 amino acids, acts as a dominant negative HARP effector by heterodimerizing with the wild-type growth factor. The aim of our study was to evaluate the potential inhibitory activity of HARPDelta111-136 on the U87 MG human glioblastoma cell line. By overexpressing the truncated form of HARP in stably established clones of U87 MG cells, we observed an inhibition of proliferation under both anchorage-dependent and anchorage-independent conditions. We confirmed these results in an in vivo subcutaneous tumor xenograft model. In addition, we found that HARPDelta111-136 inhibited cell proliferation in a paracrine manner. Analysis of key cellular pathways revealed a decrease of cell adhesion in U87 MG cells that overexpressed the mutant protein, which could explain this inhibitory effect. A replication-defective adenovirus model that encoded HARPDelta111-136 supported a putative antiproliferative role for the truncated protein in vitro and in vivo. Interestingly, HARPDelta111-136 was also able to abolish angiogenic activity in HUVEC proliferation and in a Matrigel plug assay. These results demonstrate that considering its antiproliferative and angiostatic effects, HARPDelta111-136 could be of great interest when used in conjunction with standard treatments.


Subject(s)
Brain Neoplasms/pathology , Carrier Proteins/genetics , Cytokines/genetics , Glioblastoma/pathology , Mutation/genetics , Proto-Oncogene Proteins/genetics , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , CHO Cells , Cell Adhesion , Cell Movement , Cell Proliferation , Collagen/metabolism , Cricetinae , Cricetulus , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Immunoenzyme Techniques , Laminin/metabolism , Mice , Mice, Nude , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Proteoglycans/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...