Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Exp Hematol ; 127: 40-51, 2023 11.
Article in English | MEDLINE | ID: mdl-37666355

ABSTRACT

Hematopoietic stem cells (HSCs) enable hematopoietic stem cell transplantation (HCT) through their ability to replenish the entire blood system. Proliferation of HSCs is linked to decreased reconstitution potential, and a precise regulation of actively dividing HSCs is thus essential to ensure long-term functionality. This regulation becomes important in the transplantation setting where HSCs undergo proliferation followed by a gradual transition to quiescence and homeostasis. Although mouse HSCs have been well studied under homeostatic conditions, the mechanisms regulating HSC activation under stress remain unclear. Here, we analyzed the different phases of regeneration after transplantation. We isolated bone marrow from mice at 8 time points after transplantation and examined the reconstitution dynamics and transcriptional profiles of stem and progenitor populations. We found that regenerating HSCs initially produced rapidly expanding progenitors and displayed distinct changes in fatty acid metabolism and glycolysis. Moreover, we observed molecular changes in cell cycle, MYC and mTOR signaling in both HSCs, and progenitor subsets. We used a decay rate model to fit the temporal transcription profiles of regenerating HSCs and identified genes with progressively decreased or increased expression after transplantation. These genes overlapped to a large extent with published gene sets associated with key aspects of HSC function, demonstrating the potential of this data set as a resource for identification of novel HSC regulators. Taken together, our study provides a detailed functional and molecular characterization of HSCs at different phases of regeneration and identifies a gene set associated with the transition from proliferation to quiescence.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Mice , Animals , Hematopoietic Stem Cells/metabolism , Bone Marrow , Cell Cycle/genetics , Signal Transduction
2.
Blood Adv ; 7(24): 7407-7417, 2023 12 26.
Article in English | MEDLINE | ID: mdl-37487020

ABSTRACT

Culture conditions in which hematopoietic stem cells (HSCs) can be expanded for clinical benefit are highly sought after. To elucidate regulatory mechanisms governing the maintenance and propagation of human HSCs ex vivo, we screened libraries of annotated small molecules in human cord blood cells using an optimized assay for detection of functional HSCs during culture. We found that the antifungal agent ciclopirox ethanolamine (CPX) selectively supported immature CD34+CD90+ cells during culture and enhanced their long-term in vivo repopulation capacity. Purified HSCs treated with CPX showed a reduced cell division rate and an enrichment of HSC-specific gene expression patterns. Mechanistically, we found that the HSC stimulating effect of CPX was directly mediated by chelation of the intracellular iron pool, which in turn affected iron-dependent proteins and enzymes mediating cellular metabolism and respiration. Our findings unveil a significant impact of iron homeostasis in regulation of human HSCs, with important implications for both basic HSC biology and clinical hematology.


Subject(s)
Hematopoietic Stem Cells , Iron , Humans , Ciclopirox/pharmacology , Ciclopirox/metabolism , Iron/metabolism , Hematopoietic Stem Cells/metabolism , Antigens, CD34/metabolism , Ethanolamines/metabolism , Ethanolamines/pharmacology
3.
J Biol Chem ; 299(5): 104662, 2023 05.
Article in English | MEDLINE | ID: mdl-36997086

ABSTRACT

To chemically modulate the ubiquitin-proteasome system for the degradation of specific target proteins is currently emerging as an alternative therapeutic modality. Earlier, we discovered such properties of the stem cell-supporting small molecule UM171 and identified that members of the CoREST complex (RCOR1 and LSD1) are targeted for degradation. UM171 supports the in vitro propagation of hematopoietic stem cells by transiently perturbing the differentiation-promoting effects of CoREST. Here, we employed global proteomics to map the UM171-targeted proteome and identified the additional target proteins, namely RCOR3, RREB1, ZNF217, and MIER2. Further, we discovered that critical elements recognized by Cul3KBTBD4 ligase in the presence of UM171 are located within the EGL-27 and MTA1 homology 2 (ELM2) domain of the substrate proteins. Subsequent experiments identified conserved amino acid sites in the N-terminus of the ELM2 domain that are essential for UM171-mediated degradation. Overall, our findings provide a detailed account on the ELM2 degrome targeted by UM171 and identify critical sites required for UM171-mediated degradation of specific substrates. Given the target profile, our results are highly relevant in a clinical context and point towards new therapeutic applications for UM171.


Subject(s)
Carrier Proteins , Cullin Proteins , Hematopoietic Stem Cells , Protein Domains , Protein Kinases , Proteolysis , Cell Differentiation/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Substrate Specificity , Ubiquitin/metabolism , Cullin Proteins/metabolism , Carrier Proteins/metabolism , Protein Kinases/chemistry
4.
Sci Rep ; 12(1): 18169, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36307542

ABSTRACT

The CRISPR/Cas9 system offers enormous versatility for functional genomics but many applications have proven to be challenging in primary human cells compared to cell lines or mouse cells. Here, to establish a paradigm for multiplexed gene editing in primary human cord blood-derived hematopoietic stem and progenitor cells (HSPCs), we used co-delivery of lentiviral sgRNA vectors expressing either Enhanced Green Fluorescent Protein (EGFP) or Kusabira Orange (KuO), together with Cas9 mRNA, to simultaneously edit two genetic loci. The fluorescent markers allow for tracking of either single- or double-edited cells, and we could achieve robust double knockout of the cell surface molecules CD45 and CD44 with an efficiency of ~ 70%. As a functional proof of concept, we demonstrate that this system can be used to model gene dependencies for cell survival, by simultaneously targeting the cohesin genes STAG1 and STAG2. Moreover, we show combinatorial effects with potential synergy for HSPC expansion by targeting the Aryl Hydrocarbon Receptor (AHR) in conjunction with members of the CoREST complex. Taken together, our traceable multiplexed CRISPR/Cas9 system enables studies of genetic dependencies and cooperation in primary HSPCs, and has important implications for modelling polygenic diseases, as well as investigation of the underlying mechanisms of gene interactions.


Subject(s)
Gene Editing , Hematopoietic Stem Cells , Humans , Mice , Animals , Hematopoietic Stem Cells/metabolism , Gene Targeting , Cell Line , CRISPR-Cas Systems
5.
Exp Hematol ; 115: 20-29, 2022 11.
Article in English | MEDLINE | ID: mdl-36041657

ABSTRACT

The molecular mechanisms regulating key fate decisions of hematopoietic stem cells (HSCs) remain incompletely understood. Here, we targeted global shRNA libraries to primary human hematopoietic stem and progenitor cells (HSPCs) to screen for modifiers of self-renewal and differentiation, and identified metastasis-associated 1 (MTA1) as a negative regulator of human HSPC propagation in vitro. Knockdown of MTA1 by independent shRNAs in primary human cord blood (CB) HSPCs led to a cell expansion during culture and a relative accumulation of immature CD34+CD90+ cells with perturbed in vitro differentiation potential. Transplantation experiments in immunodeficient mice revealed a significant reduction in human chimerism in both blood and bone marrow from HSPCs with knockdown of MTA1, possibly caused by reduced maturation of blood cells. We further found that MTA1 associates with the nucleosome remodeling deacetylase (NuRD) complex in human HSPCs, and on knockdown of MTA1, we observed an increase in H3K27Ac marks coupled with a downregulation of genes linked to differentiation toward the erythroid lineage. Together, our findings identify MTA1 as a novel regulator of human HSPCs in vitro and in vivo with critical functions for differentiation commitment.


Subject(s)
Fetal Blood , Hematopoietic Stem Cells , Humans , Mice , Animals , RNA Interference , Antigens, CD34 , Cell Differentiation/genetics , Repressor Proteins/genetics , Trans-Activators/genetics
7.
Sci Rep ; 10(1): 22393, 2020 12 28.
Article in English | MEDLINE | ID: mdl-33372184

ABSTRACT

The CRISPR/Cas9 system is a versatile tool for functional genomics and forward genetic screens in mammalian cells. However, it has been challenging to deliver the CRISPR components to sensitive cell types, such as primary human hematopoietic stem and progenitor cells (HSPCs), partly due to lentiviral transduction of Cas9 being extremely inefficient in these cells. Here, to overcome these hurdles, we developed a combinatorial system using stable lentiviral delivery of single guide RNA (sgRNA) followed by transient transfection of Cas9 mRNA by electroporation in human cord blood-derived CD34+ HSPCs. We further applied an optimized sgRNA structure, that significantly improved editing efficiency in this context, and we obtained knockout levels reaching 90% for the cell surface proteins CD45 and CD44 in sgRNA transduced HSPCs. Our combinatorial CRISPR/Cas9 delivery approach had no negative influence on CD34 expression or colony forming capacity in vitro compared to non-treated HSPCs. Furthermore, gene edited HSPCs showed intact in vivo reconstitution capacity following transplantation to immunodeficient mice. Taken together, we developed a paradigm for combinatorial CRISPR/Cas9 delivery that enables efficient and traceable gene editing in primary human HSPCs, and is compatible with high functionality both in vitro and in vivo.


Subject(s)
CRISPR-Cas Systems , Cell Tracking , Gene Editing , Gene Transfer Techniques , Hematopoietic Stem Cells/metabolism , Lentivirus , RNA/genetics , Animals , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Heterografts , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , K562 Cells , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred NOD , Mice, SCID
8.
Proc Natl Acad Sci U S A ; 117(35): 21267-21273, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817519

ABSTRACT

Introduction of exogenous genetic material into primary stem cells is essential for studying biological function and for clinical applications. Traditional delivery methods for nucleic acids, such as electroporation, have advanced the field, but have negative effects on stem cell function and viability. We introduce nanostraw-assisted transfection as an alternative method for RNA delivery to human hematopoietic stem and progenitor cells (HSPCs). Nanostraws are hollow alumina nanotubes that can be used to deliver biomolecules to living cells. We use nanostraws to target human primary HSPCs and show efficient delivery of mRNA, short interfering RNAs (siRNAs), DNA oligonucleotides, and dextrans of sizes ranging from 6 kDa to 2,000 kDa. Nanostraw-treated cells were fully functional and viable, with no impairment in their proliferative or colony-forming capacity, and showed similar long-term engraftment potential in vivo as untreated cells. Additionally, we found that gene expression of the cells was not perturbed by nanostraw treatment, while conventional electroporation changed the expression of more than 2,000 genes. Our results show that nanostraw-mediated transfection is a gentle alternative to established gene delivery methods, and uniquely suited for nonperturbative treatment of sensitive primary stem cells.


Subject(s)
Gene Transfer Techniques , Hematopoietic Stem Cells , Nanostructures , Animals , Hematopoietic Stem Cell Transplantation , Humans , Mice , Microinjections
9.
Blood ; 136(19): 2151-2161, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32582923

ABSTRACT

Culture conditions in which hematopoietic stem cells (HSCs) can be expanded for clinical benefit are highly sought after. Here, we report that inhibition of the epigenetic regulator lysine-specific histone demethylase 1A (LSD1) induces a rapid expansion of human cord blood-derived CD34+ cells and promotes in vitro propagation of long-term repopulating HSCs by preventing differentiation. The phenotype and molecular characteristics of cells treated with LSD1 inhibitors were highly similar to cells treated with UM171, an agent promoting expansion of HSCs through undefined mechanisms and currently being tested in clinical trials. Strikingly, we found that LSD1, as well as other members of the LSD1-containing chromatin remodeling complex CoREST, is rapidly polyubiquitinated and degraded upon UM171 treatment. CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 depletion of the CoREST core member, RCOR1, resulted in expansion of CD34+ cells similar to LSD1 inhibition and UM171. Taken together, LSD1 and CoREST restrict HSC expansion and are principal targets of UM171, forming a mechanistic basis for the HSC-promoting activity of UM171.


Subject(s)
Cell Differentiation , Co-Repressor Proteins/metabolism , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Histone Demethylases/antagonists & inhibitors , Indoles/pharmacology , Nerve Tissue Proteins/metabolism , Pyrimidines/pharmacology , Antigens, CD34/metabolism , Cell Proliferation , Co-Repressor Proteins/genetics , Fetal Blood/drug effects , Fetal Blood/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Nerve Tissue Proteins/genetics
10.
Blood Adv ; 3(4): 681-691, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808686

ABSTRACT

Identification of determinants of fate choices in hematopoietic stem cells (HSCs) is essential to improve the clinical use of HSCs and to enhance our understanding of the biology of normal and malignant hematopoiesis. Here, we show that high-mobility group AT hook 2 (HMGA2), a nonhistone chromosomal-binding protein, is highly and preferentially expressed in HSCs and in the most immature progenitor cell subset of fetal, neonatal, and adult human hematopoiesis. Knockdown of HMGA2 by short hairpin RNA impaired the long-term hematopoietic reconstitution of cord blood (CB)-derived CB CD34+ cells. Conversely, overexpression of HMGA2 in CB CD34+ cells led to overall enhanced reconstitution in serial transplantation assays accompanied by a skewing toward the myeloerythroid lineages. RNA-sequencing analysis showed that enforced HMGA2 expression in CD34+ cells induced gene-expression signatures associated with differentiation toward megakaryocyte-erythroid and myeloid lineages, as well as signatures associated with growth and survival, which at the protein level were coupled with strong activation of AKT. Taken together, our findings demonstrate a key role of HMGA2 in regulation of both proliferation and differentiation of human HSPCs.


Subject(s)
HMGA2 Protein/genetics , Hematopoiesis , Hematopoietic Stem Cells/cytology , Animals , Cell Proliferation , Cells, Cultured , Erythroid Cells/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Mice, SCID , Myeloid Cells/cytology , Up-Regulation
12.
Haematologica ; 103(9): 1444-1450, 2018 09.
Article in English | MEDLINE | ID: mdl-29880606

ABSTRACT

Despite extensive studies, defining culture conditions in which hematopoietic stem cells can be expanded ex vivo has been challenging. Here we show that chemical inhibition of the NF-κB signaling pathway leads to a significant improvement of hematopoietic stem cell function from ex vivo cultured human umbilical cord blood derived CD34+ cells. We found a distinct peak of activation of the NF-κB pathway shortly after cells were put in culture, and consequently inhibition of the pathway was both necessary and sufficient during the first 24 hours of culture where it reduced the levels of several pro-inflammatory cytokines. Taken together, NF-κB pathway inhibition facilitates propagation of hematopoietic stem cells in culture and may complement other strategies for hematopoietic stem cell expansion by relieving stress signals that are induced as an immediate response to culture initiation.


Subject(s)
Hematopoietic Stem Cells/metabolism , NF-kappa B/metabolism , Signal Transduction , Animals , Antigens, CD34/metabolism , Biomarkers , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Inflammation Mediators/metabolism , Mice , Phenotype , Signal Transduction/drug effects , Thy-1 Antigens/metabolism
13.
Cell Rep ; 21(12): 3514-3523, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29262330

ABSTRACT

The transcription factor hepatic leukemia factor (HLF) is strongly expressed in hematopoietic stem cells (HSCs) and is thought to influence both HSC self-renewal and leukemogenesis. However, the physiological role of HLF in hematopoiesis and HSC function is unclear. Here, we report that mice lacking Hlf are viable with essentially normal hematopoietic parameters, including an intact HSC pool during steady-state hematopoiesis. In contrast, when challenged through transplantation, Hlf-deficient HSCs showed an impaired ability to reconstitute hematopoiesis and became gradually exhausted upon serial transplantation. Transcriptional profiling of Hlf-deficient HSCs revealed changes associated with enhanced cellular activation, and cell-cycle analysis demonstrated a significant reduction of quiescent HSCs. Accordingly, toxic insults targeting dividing cells completely eradicated the HSC pool in Hlf-deficient mice. In summary, our findings point to HLF as a critical regulator of HSC quiescence and as an essential factor for maintaining the HSC pool during regeneration.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Proliferation , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Animals , Apoptosis , Basic-Leucine Zipper Transcription Factors/genetics , Cells, Cultured , DNA Damage , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...