Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Nature ; 630(8016): 412-420, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839950

ABSTRACT

The processes that govern human haematopoietic stem cell (HSC) self-renewal and engraftment are poorly understood and challenging to recapitulate in culture to reliably expand functional HSCs1-3. Here we identify MYC target 1 (MYCT1; also known as MTLC) as a crucial human HSC regulator that moderates endocytosis and environmental sensing in HSCs. MYCT1 is selectively expressed in undifferentiated human haematopoietic stem and progenitor cells (HSPCs) and endothelial cells but becomes markedly downregulated during HSC culture. Lentivirus-mediated knockdown of MYCT1 prevented human fetal liver and cord blood (CB) HSPC expansion and engraftment. By contrast, restoring MYCT1 expression improved the expansion and engraftment of cultured CB HSPCs. Single-cell RNA sequencing of human CB HSPCs in which MYCT1 was knocked down or overexpressed revealed that MYCT1 governs important regulatory programmes and cellular properties essential for HSC stemness, such as ETS factor expression and low mitochondrial activity. MYCT1 is localized in the endosomal membrane in HSPCs and interacts with vesicle trafficking regulators and signalling machinery. MYCT1 loss in HSPCs led to excessive endocytosis and hyperactive signalling responses, whereas restoring MYCT1 expression balanced culture-induced endocytosis and dysregulated signalling. Moreover, sorting cultured CB HSPCs on the basis of lowest endocytosis rate identified HSPCs with preserved MYCT1 expression and MYCT1-regulated HSC stemness programmes. Our work identifies MYCT1-moderated endocytosis and environmental sensing as essential regulatory mechanisms required to preserve human HSC stemness. Our data also pinpoint silencing of MYCT1 as a cell-culture-induced vulnerability that compromises human HSC expansion.


Subject(s)
Cell Self Renewal , Hematopoietic Stem Cells , Nuclear Proteins , Animals , Female , Humans , Male , Mice , Cells, Cultured , Endocytosis , Endosomes/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fetal Blood/cytology , Gene Knockdown Techniques , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Liver/cytology , Liver/metabolism , Liver/embryology , Mitochondria/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Single-Cell Gene Expression Analysis
2.
Nature ; 604(7906): 534-540, 2022 04.
Article in English | MEDLINE | ID: mdl-35418685

ABSTRACT

The ontogeny of human haematopoietic stem cells (HSCs) is poorly defined owing to the inability to identify HSCs as they emerge and mature at different haematopoietic sites1. Here we created a single-cell transcriptome map of human haematopoietic tissues from the first trimester to birth and found that the HSC signature RUNX1+HOXA9+MLLT3+MECOM+HLF+SPINK2+ distinguishes HSCs from progenitors throughout gestation. In addition to the aorta-gonad-mesonephros region, nascent HSCs populated the placenta and yolk sac before colonizing the liver at 6 weeks. A comparison of HSCs at different maturation stages revealed the establishment of HSC transcription factor machinery after the emergence of HSCs, whereas their surface phenotype evolved throughout development. The HSC transition to the liver marked a molecular shift evidenced by suppression of surface antigens reflecting nascent HSC identity, and acquisition of the HSC maturity markers CD133 (encoded by PROM1) and HLA-DR. HSC origin was tracked to ALDH1A1+KCNK17+ haemogenic endothelial cells, which arose from an IL33+ALDH1A1+ arterial endothelial subset termed pre-haemogenic endothelial cells. Using spatial transcriptomics and immunofluorescence, we visualized this process in ventrally located intra-aortic haematopoietic clusters. The in vivo map of human HSC ontogeny validated the generation of aorta-gonad-mesonephros-like definitive haematopoietic stem and progenitor cells from human pluripotent stem cells, and serves as a guide to improve their maturation to functional HSCs.


Subject(s)
Endothelial Cells , Hematopoietic Stem Cells , Cell Differentiation , Endothelium , Female , Hematopoiesis , Humans , Mesonephros , Pregnancy
3.
Blood ; 142(6): 519-532, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37339578

ABSTRACT

Developmental hematopoiesis consists of multiple, partially overlapping hematopoietic waves that generate the differentiated blood cells required for embryonic development while establishing a pool of undifferentiated hematopoietic stem cells (HSCs) for postnatal life. This multilayered design in which active hematopoiesis migrates through diverse extra and intraembryonic tissues has made it difficult to define a roadmap for generating HSCs vs non-self-renewing progenitors, especially in humans. Recent single-cell studies have helped in identifying the rare human HSCs at stages when functional assays are unsuitable for distinguishing them from progenitors. This approach has made it possible to track the origin of human HSCs to the unique type of arterial endothelium in the aorta-gonad-mesonephros region and document novel benchmarks for HSC migration and maturation in the conceptus. These studies have delivered new insights into the intricate process of HSC generation and provided tools to inform the in vitro efforts to replicate the physiological developmental journey from pluripotent stem cells via distinct mesodermal and endothelial intermediates to HSCs.


Subject(s)
Embryo, Mammalian , Hematopoietic Stem Cells , Female , Pregnancy , Humans , Hematopoiesis/physiology , Mesonephros
4.
Nature ; 576(7786): 281-286, 2019 12.
Article in English | MEDLINE | ID: mdl-31776511

ABSTRACT

Limited knowledge of the mechanisms that govern the self-renewal of human haematopoietic stem cells (HSCs), and why this fails in culture, have impeded the expansion of HSCs for transplantation1. Here we identify MLLT3 (also known as AF9) as a crucial regulator of HSCs that is highly enriched in human fetal, neonatal and adult HSCs, but downregulated in culture. Depletion of MLLT3 prevented the maintenance of transplantable human haematopoietic stem or progenitor cells (HSPCs) in culture, whereas stabilizing MLLT3 expression in culture enabled more than 12-fold expansion of transplantable HSCs that provided balanced multilineage reconstitution in primary and secondary mouse recipients. Similar to endogenous MLLT3, overexpressed MLLT3 localized to active promoters in HSPCs, sustained levels of H3K79me2 and protected the HSC transcriptional program in culture. MLLT3 thus acts as HSC maintenance factor that links histone reader and modifying activities to modulate HSC gene expression, and may provide a promising approach to expand HSCs for transplantation.


Subject(s)
Cell Self Renewal , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Nuclear Proteins/metabolism , Animals , Cells, Cultured , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Humans , Mice , Nuclear Proteins/genetics , Protein Binding
5.
EMBO J ; 33(6): 534-5, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24562387

ABSTRACT

Stem cells ensure the maintenance of tissue homeostasis throughout life by tightly regulating their self-renewal and differentiation. In a recent study published in Nature, Nakada et al, 2014 unveil an unexpected endocrine mechanism that regulates hematopoietic stem cell (HSC) self-renewal.


Subject(s)
Estrogens/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Female , Male , Pregnancy
6.
PLoS Pathog ; 11(6): e1004955, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26067822

ABSTRACT

Highly active antiretroviral therapy (HAART) suppresses human immunodeficiency virus (HIV) replication to undetectable levels but cannot fully eradicate the virus because a small reservoir of CD4+ T cells remains latently infected. Since HIV efficiently infects only activated CD4+ T cells and since latent HIV primarily resides in resting CD4+ T cells, it is generally assumed that latency is established when a productively infected cell recycles to a resting state, trapping the virus in a latent state. In this study, we use a dual reporter virus--HIV Duo-Fluo I, which identifies latently infected cells immediately after infection--to investigate how T cell activation affects the establishment of HIV latency. We show that HIV latency can arise from the direct infection of both resting and activated CD4+ T cells. Importantly, returning productively infected cells to a resting state is not associated with a significant silencing of the integrated HIV. We further show that resting CD4+ T cells from human lymphoid tissue (tonsil, spleen) show increased latency after infection when compared to peripheral blood. Our findings raise significant questions regarding the most commonly accepted model for the establishment of latent HIV and suggest that infection of both resting and activated primary CD4+ T cells produce latency.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV-1/physiology , Lymphocyte Activation/immunology , Virus Latency/physiology , Cell Separation , Cells, Cultured , Flow Cytometry , Humans , Virus Replication/immunology
7.
Proc Natl Acad Sci U S A ; 109(28): 11318-23, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733740

ABSTRACT

PIK3R2 encodes a ubiquitous regulatory subunit (p85ß) of PI3K, an enzyme that generates 3-polyphosphoinositides at the plasma membrane. PI3K activation triggers cell survival and migration. We found that p85ß expression is elevated in breast and colon carcinomas and that its increased expression correlates with PI3K pathway activation and tumor progression. p85ß expression induced moderate PIP(3) generation at the cell membrane and enhanced cell invasion. In accordance, genetic alteration of pik3r2 expression levels modulated tumor progression in vivo. Increased p85ß expression thus represents a cellular strategy in cancer progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Phosphatidylinositol 3-Kinases/physiology , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Colonic Neoplasms/metabolism , Disease Progression , Female , Humans , Mice , Mice, SCID , Mice, Transgenic , NIH 3T3 Cells , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary , Signal Transduction
8.
Diabetologia ; 57(6): 1154-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24570141

ABSTRACT

AIMS/HYPOTHESIS: The association between low birthweight (LBW) and risk of developing type 2 diabetes may involve epigenetic mechanisms, with skeletal muscle being a prime target tissue. Differential DNA methylation patterns have been observed in single genes in muscle tissue from type 2 diabetic and LBW individuals, and we recently showed multiple DNA methylation changes during short-term high-fat overfeeding in muscle of healthy people. In a randomised crossover study, we analysed genome-wide DNA promoter methylation in skeletal muscle of 17 young LBW men and 23 matched normal birthweight (NBW) men after a control and a 5 day high-fat overfeeding diet. METHODS: DNA methylation was measured using Illumina's Infinium BeadArray covering 27,578 CpG sites representing 14,475 different genes. RESULTS: After correction for multiple comparisons, DNA methylation levels were found to be similar in the LBW and NBW groups during the control diet. Whereas widespread DNA methylation changes were observed in the NBW group in response to high-fat overfeeding, only a few methylation changes were seen in the LBW group (χ(2), p < 0.001). CONCLUSIONS/INTERPRETATION: Our results indicate lower DNA methylation plasticity in skeletal muscle from LBW vs NBW men, potentially contributing to understanding the link between LBW and increased risk of type 2 diabetes.


Subject(s)
DNA Methylation/genetics , Diet, High-Fat/adverse effects , Infant, Low Birth Weight/physiology , Adult , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic/genetics , Humans , Insulin Resistance/genetics , Insulin Resistance/physiology , Male , Muscle, Skeletal/metabolism , Young Adult , DNA Methyltransferase 3B
9.
Nucleic Acids Res ; 40(1): 116-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21911366

ABSTRACT

Global mechanisms defining the gene expression programs specific for hematopoiesis are still not fully understood. Here, we show that promoter DNA demethylation is associated with the activation of hematopoietic-specific genes. Using genome-wide promoter methylation arrays, we identified 694 hematopoietic-specific genes repressed by promoter DNA methylation in human embryonic stem cells and whose loss of methylation in hematopoietic can be associated with gene expression. The association between promoter methylation and gene expression was studied for many hematopoietic-specific genes including CD45, CD34, CD28, CD19, the T cell receptor (TCR), the MHC class II gene HLA-DR, perforin 1 and the phosphoinositide 3-kinase (PI3K) and results indicated that DNA demethylation was not always sufficient for gene activation. Promoter demethylation occurred either early during embryonic development or later on during hematopoietic differentiation. Analysis of the genome-wide promoter methylation status of induced pluripotent stem cells (iPSCs) generated from somatic CD34(+) HSPCs and differentiated derivatives from CD34(+) HSPCs confirmed the role of DNA methylation in regulating the expression of genes of the hemato-immune system, and indicated that promoter methylation of these genes may be associated to stemness. Together, these data suggest that promoter DNA demethylation might play a role in the tissue/cell-specific genome-wide gene regulation within the hematopoietic compartment.


Subject(s)
DNA Methylation , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Promoter Regions, Genetic , Animals , Cell Dedifferentiation , Cell Line , Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Humans , Infant, Newborn , Mice , Oligonucleotide Array Sequence Analysis
10.
Nat Commun ; 15(1): 1579, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383676

ABSTRACT

Oncogene-induced replication stress is a crucial driver of genomic instability and one of the key events contributing to the onset and evolution of cancer. Despite its critical role in cancer, the mechanisms that generate oncogene-induced replication stress remain not fully understood. Here, we report that an oncogenic c-Myc-dependent increase in cohesins on DNA contributes to the induction of replication stress. Accumulation of cohesins on chromatin is not sufficient to cause replication stress, but also requires cohesins to accumulate at specific sites in a CTCF-dependent manner. We propose that the increased accumulation of cohesins at CTCF site interferes with the progression of replication forks, contributing to oncogene-induced replication stress. This is different from, and independent of, previously suggested mechanisms of oncogene-induced replication stress. This, together with the reported protective role of cohesins in preventing replication stress-induced DNA damage, supports a double-edge involvement of cohesins in causing and tolerating oncogene-induced replication stress.


Subject(s)
Cohesins , Neoplasms , Humans , Chromatin , Cell Cycle Proteins/metabolism , DNA Replication , DNA
11.
Proc Natl Acad Sci U S A ; 107(31): 13736-41, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20631301

ABSTRACT

The longevity-promoting NAD+-dependent class III histone deacetylase Sirtuin 1 (SIRT1) is involved in stem cell function by controlling cell fate decision and/or by regulating the p53-dependent expression of NANOG. We show that SIRT1 is down-regulated precisely during human embryonic stem cell differentiation at both mRNA and protein levels and that the decrease in Sirt1 mRNA is mediated by a molecular pathway that involves the RNA-binding protein HuR and the arginine methyltransferase coactivator-associated arginine methyltransferase 1 (CARM1). SIRT1 down-regulation leads to reactivation of key developmental genes such as the neuroretinal morphogenesis effectors DLL4, TBX3, and PAX6, which are epigenetically repressed by this histone deacetylase in pluripotent human embryonic stem cells. Our results indicate that SIRT1 is regulated during stem cell differentiation in the context of a yet-unknown epigenetic pathway that controls specific developmental genes in embryonic stem cells.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Sirtuin 1/metabolism , Animals , CARD Signaling Adaptor Proteins/metabolism , Cell Line , Guanylate Cyclase/metabolism , Humans , Mice , Mice, Knockout , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA Stability , Sirtuin 1/deficiency , Sirtuin 1/genetics
12.
Adv Exp Med Biol ; 741: 231-53, 2012.
Article in English | MEDLINE | ID: mdl-22457114

ABSTRACT

Understanding the molecular mechanisms involved in the control of cell differentiation during embryonic development is currently one of the main objectives of developmental biology. This knowledge will provide a basis for the development of new strategies in the field of regenerative medicine, one of the most promising weapons to fight many human diseases. Cell differentiation during embryonic development is controlled primarily by epigenetic factors, that is, mechanisms involved in the regulation of chromatin structure and gene expression. Here we describe the best known epigenetic modifications, and pathways, mainly focused on DNA methylation and histone modifications, and try to depict the state of art in our knowledge about epigenetic regulation of embryonic stem cell maintenance and differentiation.


Subject(s)
Embryonic Stem Cells/physiology , Epigenesis, Genetic , Animals , Cell Differentiation/physiology , Chromatin/chemistry , Chromatin/metabolism , DNA Methylation , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Embryonic Stem Cells/cytology , Histones/metabolism , Humans , Protein Processing, Post-Translational
13.
Adv Exp Med Biol ; 738: 236-55, 2012.
Article in English | MEDLINE | ID: mdl-22399383

ABSTRACT

Epigenetics is a new and expanding science that studies the chromatin-based regulation of gene expression. It is achieving considerable importance, especially with regard to developmental mechanisms that drive cell and organ differentiation, as well as in all those biological processes that involve response and adaptation to environmental stimuli. One of the most interesting biological questions concerning animals, especially human beings, is the ability to distinguish self from nonself. This ability has developed throughout evolution, both as the main function of the immune system, which defends against attack by foreign organisms and at the level of consciousness of oneself as an individual, one of the highest functions of the brain that enables social life. Here we will attempt to dissect the epigenetic mechanisms involved in establishing these higher functions and describe some alterations of the epigenetic machinery responsible for the impairment of correct self-recognition and self-identity.


Subject(s)
Adaptation, Physiological/immunology , Adaptive Immunity/physiology , Epigenesis, Genetic/immunology , Evolution, Molecular , Self Tolerance/physiology , Adaptation, Physiological/genetics , Animals , Humans
14.
Sci Rep ; 12(1): 14804, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36045139

ABSTRACT

Post-translational modifications, such as lysine acetylation, regulate the activity of diverse proteins across many cellular compartments. Protein deacetylation in mitochondria is catalyzed by the enzymatic activity of the NAD+-dependent deacetylase sirtuin 3 (SIRT3), however it remains unclear whether corresponding mitochondrial acetyltransferases exist. We used a bioinformatics approach to search for mitochondrial proteins with an acetyltransferase catalytic domain, and identified a novel splice variant of ELP3 (mt-ELP3) of the elongator complex, which localizes to the mitochondrial matrix in mammalian cells. Unexpectedly, mt-ELP3 does not mediate mitochondrial protein acetylation but instead induces a post-transcriptional modification of mitochondrial-transfer RNAs (mt-tRNAs). Overexpression of mt-ELP3 leads to the protection of mt-tRNAs against the tRNA-specific RNase angiogenin, increases mitochondrial translation, and furthermore increases expression of OXPHOS complexes. This study thus identifies mt-ELP3 as a non-canonical mt-tRNA modifying enzyme.


Subject(s)
Histone Acetyltransferases , RNA Processing, Post-Transcriptional , Animals , Histone Acetyltransferases/metabolism , Mammals/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Biosynthesis , RNA, Mitochondrial/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism
15.
Blood ; 113(11): 2488-97, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-19075189

ABSTRACT

Lymphomas are assumed to originate at different stages of lymphocyte development through chromosomal aberrations. Thus, different lymphomas resemble lymphocytes at distinct differentiation stages and show characteristic morphologic, genetic, and transcriptional features. Here, we have performed a microarray-based DNA methylation profiling of 83 mature aggressive B-cell non-Hodgkin lymphomas (maB-NHLs) characterized for their morphologic, genetic, and transcriptional features, including molecular Burkitt lymphomas and diffuse large B-cell lymphomas. Hierarchic clustering indicated that methylation patterns in maB-NHLs were not strictly associated with morphologic, genetic, or transcriptional features. By supervised analyses, we identified 56 genes de novo methylated in all lymphoma subtypes studied and 22 methylated in a lymphoma subtype-specific manner. Remarkably, the group of genes de novo methylated in all lymphoma subtypes was significantly enriched for polycomb targets in embryonic stem cells. De novo methylated genes in all maB-NHLs studied were expressed at low levels in lymphomas and normal hematopoietic tissues but not in nonhematopoietic tissues. These findings, especially the enrichment for polycomb targets in stem cells, indicate that maB-NHLs with different morphologic, genetic, and transcriptional background share a similar stem cell-like epigenetic pattern. This suggests that maB-NHLs originate from cells with stem cell features or that stemness was acquired during lymphomagenesis by epigenetic remodeling.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic/physiology , Gene Expression Profiling , Genomics/methods , Lymphoma, B-Cell/genetics , Oligonucleotide Array Sequence Analysis , Cell Transformation, Neoplastic/pathology , DNA Methylation/physiology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Humans , Lymphoma, B-Cell/pathology , Male , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis/methods , Transcription, Genetic/physiology , Tumor Cells, Cultured
16.
Mol Cancer ; 9: 170, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20591152

ABSTRACT

BACKGROUND: Wnt factors control cell differentiation through semi-independent molecular cascades known as the beta-catenin-dependent (canonical) and -independent (non-canonical) Wnt signalling pathways. Genetic and epigenetic alteration of components of the canonical Wnt signalling pathway is one of the primary mechanisms underlying colon cancer. Despite increasing evidence of the role of the non-canonical pathways in tumourigenesis, however, the underlying molecular mechanisms are poorly understood. RESULTS: Here we report that the receptor tyrosine kinase-like orphan receptor 2 (ROR2), a transmembrane receptor for Wnt factors that activates non-canonical pathways, is frequently repressed by aberrant promoter hypermethylation in human colon cancer cell lines and primary tumours. By restoring ROR2 activity in colon cancer cells harbouring ROR2 promoter hypermethylation, we show that the role of ROR2 in colon cancer cells is mediated, at least in part, by canonical Wnt and that its epigenetic-dependent loss can be pro-tumourigenic. CONCLUSIONS: Our data show the importance of epigenetic alterations of ROR2 in colon cancer, highlighting the close interconnection between canonical and non-canonical Wnt signalling pathways in this type of tumour.


Subject(s)
Colonic Neoplasms/genetics , Epigenesis, Genetic , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Wnt Proteins/physiology , Colonic Neoplasms/pathology , DNA Methylation , Humans , Promoter Regions, Genetic
17.
BMC Mol Biol ; 9: 81, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18817541

ABSTRACT

BACKGROUND: Regulation of the expression of particular genes can rely on mechanisms that are different from classical transcriptional and translational control. The LY6G5B and LY6G6D genes encode LY-6 domain proteins, whose expression seems to be regulated in an original fashion, consisting of an intron retention event which generates, through an early premature stop codon, a non-coding transcript, preventing expression in most cell lines and tissues. RESULTS: The MHC LY-6 non-coding transcripts have shown to be stable and very abundant in the cell, and not subject to Nonsense Mediated Decay (NMD). This retention event appears not to be solely dependent on intron features, because in the case of LY6G5B, when the intron is inserted in the artificial context of a luciferase expression plasmid, it is fully spliced but strongly stabilises the resulting luciferase transcript. In addition, by quantitative PCR we found that the retained and spliced forms are differentially expressed in tissues indicating an active regulation of the non-coding transcript. EST database analysis revealed that these genes have an alternative expression pathway with the formation of Transcription Induced Chimeras (TIC). This data was confirmed by RT-PCR, revealing the presence of different transcripts that would encode the chimeric proteins CSNKbeta-LY6G5B and G6F-LY6G6D, in which the LY-6 domain would join to a kinase domain and an Ig-like domain, respectively. CONCLUSION: In conclusion, the LY6G5B and LY6G6D intron-retained transcripts are not subjected to NMD and are more abundant than the properly spliced forms. In addition, these genes form chimeric transcripts with their neighbouring same orientation 5' genes. Of interest is the fact that the 5' genes (CSNKbeta or G6F) undergo differential splicing only in the context of the chimera (CSNKbeta-LY6G5B or G6F-LY6G6C) and not on their own.


Subject(s)
Antigens, Ly/genetics , Introns , Mutant Chimeric Proteins/genetics , Alternative Splicing , Antigens, Ly/biosynthesis , Cell Line , Codon, Nonsense , Gene Expression Regulation , Humans , Mutant Chimeric Proteins/biosynthesis , Organ Specificity , RNA Stability
18.
Exp Hematol ; 65: 1-16, 2018 09.
Article in English | MEDLINE | ID: mdl-29981365

ABSTRACT

Hematopoietic stem cells (HSCs) are multipotent cells responsible for the maintenance of the hematopoietic system throughout life. Dysregulation of the balance in HSC self-renewal, death, and differentiation can have serious consequences such as myelodysplastic syndromes or leukemia. All-trans retinoic acid (ATRA), the biologically active metabolite of vitamin A/RA, has been shown to have pleiotropic effects on hematopoietic cells, enhancing HSC self-renewal while also increasing differentiation of more mature progenitors. Furthermore, ATRA has been shown to have key roles in regulating the specification and formation of hematopoietic cells from pluripotent stem cells including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Here, we summarize the known roles of vitamin A and RA receptors in the regulation of hematopoiesis from HSCs, ES, and iPSCs.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Retinoids/metabolism , Hematopoiesis , Hematopoietic Stem Cells/physiology , Humans , Models, Biological , Receptors, Retinoic Acid/physiology , Signal Transduction
19.
Dev Cell ; 36(5): 479-80, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26954540

ABSTRACT

Reporting in Developmental Cell, Pereira et al. (2016) use in vitro lineage reprogramming insights to inform understanding of hematopoietic stem cell (HSC) development in vivo. They find Prom1(+)Sca1(+)CD34(+)CD45(-) hemogenic precursors, akin to fibroblast-derived hemato-vascular precursors, in mouse placenta and embryo. The cells mature into transplantable HSCs in culture.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Cellular Reprogramming , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Mouse Embryonic Stem Cells/cytology , Animals , Female , Pregnancy
20.
Nat Cell Biol ; 18(6): 595-606, 2016 06.
Article in English | MEDLINE | ID: mdl-27183470

ABSTRACT

Pluripotent stem cells (PSCs) may provide a potential source of haematopoietic stem/progenitor cells (HSPCs) for transplantation; however, unknown molecular barriers prevent the self-renewal of PSC-HSPCs. Using two-step differentiation, human embryonic stem cells (hESCs) differentiated in vitro into multipotent haematopoietic cells that had the CD34(+)CD38(-/lo)CD90(+)CD45(+)GPI-80(+) fetal liver (FL) HSPC immunophenotype, but exhibited poor expansion potential and engraftment ability. Transcriptome analysis of immunophenotypic hESC-HSPCs revealed that, despite their molecular resemblance to FL-HSPCs, medial HOXA genes remained suppressed. Knockdown of HOXA7 disrupted FL-HSPC function and caused transcriptome dysregulation that resembled hESC-derived progenitors. Overexpression of medial HOXA genes prolonged FL-HSPC maintenance but was insufficient to confer self-renewal to hESC-HSPCs. Stimulation of retinoic acid signalling during endothelial-to-haematopoietic transition induced the HOXA cluster and other HSC/definitive haemogenic endothelium genes, and prolonged HSPC maintenance in culture. Thus, medial HOXA gene expression induced by retinoic acid signalling marks the establishment of the definitive HSPC fate and controls HSPC identity and function.


Subject(s)
Cell Differentiation/physiology , Cell Lineage , Genes, Homeobox/genetics , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Multipotent Stem Cells/cytology , Antigens, CD34/metabolism , Cells, Cultured , Gene Expression Profiling/methods , Homeodomain Proteins/genetics , Humans , Leukocyte Common Antigens/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL