Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 4866, 2023 03 24.
Article in English | MEDLINE | ID: mdl-36964177

ABSTRACT

Progress in sample preparation for scRNA-seq is reported based on RevGel-seq, a reversible-hydrogel technology optimized for samples of fresh cells. Complexes of one cell paired with one barcoded bead are stabilized by a chemical linker and dispersed in a hydrogel in the liquid state. Upon gelation on ice the complexes are immobilized and physically separated without requiring nanowells or droplets. Cell lysis is triggered by detergent diffusion, and RNA molecules are captured on the adjacent barcoded beads for further processing with reverse transcription and preparation for cDNA sequencing. As a proof of concept, analysis of PBMC using RevGel-seq achieves results similar to microfluidic-based technologies when using the same original sample and the same data analysis software. In addition, a clinically relevant application of RevGel-seq is presented for pancreatic islet cells. Furthermore, characterizations carried out on cardiomyocytes demonstrate that the hydrogel technology readily accommodates very large cells. Standard analyses are in the 10,000-input cell range with the current gelation device, in order to satisfy common requirements for single-cell research. A convenient stopping point after two hours has been established by freezing at the cell lysis step, with full preservation of gene expression profiles. Overall, our results show that RevGel-seq represents an accessible and efficient instrument-free alternative, enabling flexibility in terms of experimental design and timing of sample processing, while providing broad coverage of cell types.


Subject(s)
Sequence Analysis, RNA , Single-Cell Analysis , Sequence Analysis, RNA/methods , Hydrogels/chemistry , Single-Cell Analysis/methods , Humans , Animals , Mice , Gene Expression Profiling
2.
FEBS Lett ; 590(22): 4084-4104, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27645909

ABSTRACT

Cellular differentiation requires precisely regulated tissue-specific and developmental stage-specific gene expression patterns. Numerous studies have highlighted the predictive power of enhancers on lineage-specific gene expression programs and have started to unravel their mechanisms of action. We review here the dynamics of the enhancer landscape during hematopoietic differentiation and how enhancers function in the context of the 3D organization of the genome. We further discuss the involvement of aberrant enhancer activity in human diseases and emerging strategies aiming at controlling enhancer activity and chromosome topology for therapeutic purposes.


Subject(s)
Cell Differentiation/genetics , Enhancer Elements, Genetic , Genetic Diseases, Inborn/genetics , Hematopoietic Stem Cells , Cell Lineage/genetics , Chromosomes/genetics , Gene Expression Regulation, Developmental , Genetic Diseases, Inborn/pathology , Genome, Human , Humans , Organ Specificity/genetics
4.
J Exp Med ; 213(3): 433-49, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26880577

ABSTRACT

Understanding the molecular regulation of hematopoietic stem and progenitor cell (HSPC) engraftment is paramount to improving transplant outcomes. To discover novel regulators of HSPC repopulation, we transplanted >1,300 mice with shRNA-transduced HSPCs within 24 h of isolation and transduction to focus on detecting genes regulating repopulation. We identified 17 regulators of HSPC repopulation: Arhgef5, Armcx1, Cadps2, Crispld1, Emcn, Foxa3, Fstl1, Glis2, Gprasp2, Gpr56, Myct1, Nbea, P2ry14, Smarca2, Sox4, Stat4, and Zfp251. Knockdown of each of these genes yielded a loss of function, except in the cases of Armcx1 and Gprasp2, whose loss enhanced hematopoietic stem cell (HSC) repopulation. The discovery of multiple genes regulating vesicular trafficking, cell surface receptor turnover, and secretion of extracellular matrix components suggests active cross talk between HSCs and the niche and that HSCs may actively condition the niche to promote engraftment. We validated that Foxa3 is required for HSC repopulating activity, as Foxa3(-/-) HSC fails to repopulate ablated hosts efficiently, implicating for the first time Foxa genes as regulators of HSPCs. We further show that Foxa3 likely regulates the HSC response to hematologic stress. Each gene discovered here offers a window into the novel processes that regulate stable HSPC engraftment into an ablated host.


Subject(s)
Genetic Association Studies , Hematopoietic Stem Cells/cytology , Amino Acid Motifs , Animals , Cell Proliferation , Cytoprotection , Enhancer Elements, Genetic/genetics , Genetic Testing , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hepatocyte Nuclear Factor 3-gamma/metabolism , Mice, Inbred C57BL , Protein Binding , Reproducibility of Results , Signal Transduction , Stress, Physiological
5.
Nat Commun ; 6: 8893, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26593974

ABSTRACT

How transcription factors (TFs) cooperate within large protein complexes to allow rapid modulation of gene expression during development is still largely unknown. Here we show that the key haematopoietic LIM-domain-binding protein-1 (LDB1) TF complex contains several activator and repressor components that together maintain an erythroid-specific gene expression programme primed for rapid activation until differentiation is induced. A combination of proteomics, functional genomics and in vivo studies presented here identifies known and novel co-repressors, most notably the ETO2 and IRF2BP2 proteins, involved in maintaining this primed state. The ETO2-IRF2BP2 axis, interacting with the NCOR1/SMRT co-repressor complex, suppresses the expression of the vast majority of archetypical erythroid genes and pathways until its decommissioning at the onset of terminal erythroid differentiation. Our experiments demonstrate that multimeric regulatory complexes feature a dynamic interplay between activating and repressing components that determines lineage-specific gene expression and cellular differentiation.


Subject(s)
Carrier Proteins/metabolism , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Erythroid Cells/cytology , Erythropoiesis , Humans , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics
6.
Blood ; 123(4): 509-19, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24184684

ABSTRACT

Loss of hematopoietic stem cell (HSC) function and increased risk of developing hematopoietic malignancies are severe and concerning complications of anticancer radiotherapy and chemotherapy. We have previously shown that thrombopoietin (TPO), a critical HSC regulator, ensures HSC chromosomal integrity and function in response to γ-irradiation by regulating their DNA-damage response. TPO directly affects the double-strand break (DSB) repair machinery through increased DNA-protein kinase (DNA-PK) phosphorylation and nonhomologous end-joining (NHEJ) repair efficiency and fidelity. This effect is not shared by other HSC growth factors, suggesting that TPO triggers a specific signal in HSCs facilitating DNA-PK activation upon DNA damage. The discovery of these unique signaling pathways will provide a means of enhancing TPO-desirable effects on HSCs and improving the safety of anticancer DNA agents. We show here that TPO specifically triggers Erk and nuclear factor κB (NF-κB) pathways in mouse hematopoietic stem and progenitor cells (HSPCs). Both of these pathways are required for a TPO-mediated increase in DSB repair. They cooperate to induce and activate the early stress-response gene, Iex-1 (ier3), upon DNA damage. Iex-1 forms a complex with pERK and the catalytic subunit of DNA-PK, which is necessary and sufficient to promote TPO-increased DNA-PK activation and NHEJ DSB repair in both mouse and human HSPCs.


Subject(s)
DNA Repair , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic , Hematopoietic Stem Cells/cytology , Immediate-Early Proteins/metabolism , NF-kappa B/metabolism , Thrombopoietin/metabolism , Active Transport, Cell Nucleus , Animals , Antineoplastic Agents/chemistry , Catalytic Domain , DNA Breaks, Double-Stranded , DNA Damage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Signal Transduction , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...