Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 31(3): 378-397.e12, 2024 03 07.
Article in English | MEDLINE | ID: mdl-38402617

ABSTRACT

Mechanisms governing the maintenance of blood-producing hematopoietic stem and multipotent progenitor cells (HSPCs) are incompletely understood, particularly those regulating fate, ensuring long-term maintenance, and preventing aging-associated stem cell dysfunction. We uncovered a role for transitory free cytoplasmic iron as a rheostat for adult stem cell fate control. We found that HSPCs harbor comparatively small amounts of free iron and show the activation of a conserved molecular response to limited iron-particularly during mitosis. To study the functional and molecular consequences of iron restriction, we developed models allowing for transient iron bioavailability limitation and combined single-molecule RNA quantification, metabolomics, and single-cell transcriptomic analyses with functional studies. Our data reveal that the activation of the limited iron response triggers coordinated metabolic and epigenetic events, establishing stemness-conferring gene regulation. Notably, we find that aging-associated cytoplasmic iron loading reversibly attenuates iron-dependent cell fate control, explicating intervention strategies for dysfunctional aged stem cells.


Subject(s)
Hematopoiesis , Iron , Hematopoiesis/genetics , Iron/metabolism , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Gene Expression Regulation , Cell Differentiation
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.
Cell ; 186(4): 685-687, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36803600

ABSTRACT

Curtailed protein translation ensures stemness and multipotency in embryonic and adult tissue-specific stem cells. In this issue of Cell, a study led by Zhao and colleagues uncovered increased susceptibility of hematopoietic stem cells (HSC) to iron-dependent programmed necrotic cell death (ferroptosis) as a consequence of low protein synthesis.


Subject(s)
Hematopoietic Stem Cells , Protein Biosynthesis , Cell Proliferation , Ferroptosis
4.
Nature ; 591(7848): 117-123, 2021 03.
Article in English | MEDLINE | ID: mdl-33442062

ABSTRACT

The activation of mostly quiescent haematopoietic stem cells (HSCs) is a prerequisite for life-long production of blood cells1. This process requires major molecular adaptations to allow HSCs to meet the regulatory and metabolic requirements for cell division2-4. The mechanisms that govern cellular reprograming upon stem-cell activation, and the subsequent return of stem cells to quiescence, have not been fully characterized. Here we show that chaperone-mediated autophagy (CMA)5, a selective form of lysosomal protein degradation, is involved in sustaining HSC function in adult mice. CMA is required for protein quality control in stem cells and for the upregulation of fatty acid metabolism upon HSC activation. We find that CMA activity in HSCs decreases with age and show that genetic or pharmacological activation of CMA can restore the functionality of old mouse and human HSCs. Together, our findings provide mechanistic insights into a role for CMA in sustaining quality control, appropriate energetics and overall long-term HSC function. Our work suggests that CMA may be a promising therapeutic target for enhancing HSC function in conditions such as ageing or stem-cell transplantation.


Subject(s)
Chaperone-Mediated Autophagy/physiology , Hematopoietic Stem Cells/physiology , Adult , Aged , Aging , Animals , Cell Self Renewal , Cells, Cultured , Chaperone-Mediated Autophagy/drug effects , Chaperone-Mediated Autophagy/genetics , Energy Metabolism , Female , Glycolysis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Linoleic Acid/metabolism , Male , Mice , Middle Aged , Multiple Myeloma/pathology , Rejuvenation , Young Adult
5.
Nat Med ; 25(1): 103-110, 2019 01.
Article in English | MEDLINE | ID: mdl-30510255

ABSTRACT

Myelodysplastic syndromes (MDS) frequently progress to acute myeloid leukemia (AML); however, the cells leading to malignant transformation have not been directly elucidated. As progression of MDS to AML in humans provides a biological system to determine the cellular origins and mechanisms of neoplastic transformation, we studied highly fractionated stem cell populations in longitudinal samples of patients with MDS who progressed to AML. Targeted deep sequencing combined with single-cell sequencing of sorted cell populations revealed that stem cells at the MDS stage, including immunophenotypically and functionally defined pre-MDS stem cells (pre-MDS-SC), had a significantly higher subclonal complexity compared to blast cells and contained a large number of aging-related variants. Single-cell targeted resequencing of highly fractionated stem cells revealed a pattern of nonlinear, parallel clonal evolution, with distinct subclones within pre-MDS-SC and MDS-SC contributing to generation of MDS blasts or progression to AML, respectively. Furthermore, phenotypically aberrant stem cell clones expanded during transformation and stem cell subclones that were not detectable in MDS blasts became dominant upon AML progression. These results reveal a crucial role of diverse stem cell compartments during MDS progression to AML and have implications for current bulk cell-focused precision oncology approaches, both in MDS and possibly other cancers that evolve from premalignant conditions, that may miss pre-existing rare aberrant stem cells that drive disease progression and leukemic transformation.


Subject(s)
Disease Progression , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/pathology , Stem Cells/metabolism , Clone Cells , Humans , Leukemia, Myeloid, Acute/genetics , Models, Biological , Mutation/genetics , Myelodysplastic Syndromes/genetics
6.
Nat Med ; 25(3): 529, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30568307

ABSTRACT

In the version of this article originally published, Ulrich Steidl's name was listed as "and Ulrich Steidl." His name has been updated to "Ulrich Steidl." The error has been fixed in the print, PDF and HTML versions of this article.

7.
Sci Transl Med ; 10(458)2018 09 12.
Article in English | MEDLINE | ID: mdl-30209246

ABSTRACT

Eltrombopag (EP), a small-molecule thrombopoietin receptor (TPO-R) agonist and potent intracellular iron chelator, has shown remarkable efficacy in stimulating sustained multilineage hematopoiesis in patients with bone marrow failure syndromes, suggesting an effect at the most immature hematopoietic stem and multipotent progenitor level. Although the functional and molecular effects of EP on megakaryopoiesis have been studied in the past, mechanistic insights into its effects on the earliest stages of hematopoiesis have been limited. We investigated the effects of EP treatment on hematopoietic stem cell (HSC) function using purified primary HSCs in separation-of-function mouse models, including a TPO-R-deficient strain, and stem cells isolated from patients undergoing TPO-R agonist treatment. Our mechanistic studies showed a stimulatory effect on stem cell self-renewal independently of TPO-R. Human and mouse HSCs responded to acute EP treatment with metabolic and gene expression alterations consistent with a reduction of intracellular labile iron pools that are essential for stem cell maintenance. Iron preloading prevented the stem cell stimulatory effects of EP. Moreover, comparative analysis of stem cells in the bone marrow of patients receiving EP showed a marked increase in the number of functional stem cells compared to patients undergoing therapy with romiplostim, another TPO-R agonist lacking an iron-chelating ability. Together, our study demonstrates that EP stimulates hematopoiesis at the stem cell level through iron chelation-mediated molecular reprogramming and indicates that labile iron pool-regulated pathways can modulate HSC function.


Subject(s)
Benzoates/pharmacology , Hematopoietic Stem Cells/metabolism , Hydrazines/pharmacology , Pyrazoles/pharmacology , Receptors, Thrombopoietin/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Cell Self Renewal/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Homeostasis/drug effects , Iron/metabolism , Iron Chelating Agents/pharmacology , Mice , Signal Transduction/drug effects
8.
Nat Chem Biol ; 11(11): 878-86, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26436839

ABSTRACT

Neomorphic mutations in isocitrate dehydrogenase 1 (IDH1) are driver mutations in acute myeloid leukemia (AML) and other cancers. We report the development of new allosteric inhibitors of mutant IDH1. Crystallographic and biochemical results demonstrated that compounds of this chemical series bind to an allosteric site and lock the enzyme in a catalytically inactive conformation, thereby enabling inhibition of different clinically relevant IDH1 mutants. Treatment of IDH1 mutant primary AML cells uniformly led to a decrease in intracellular 2-HG, abrogation of the myeloid differentiation block and induction of granulocytic differentiation at the level of leukemic blasts and more immature stem-like cells, in vitro and in vivo. Molecularly, treatment with the inhibitors led to a reversal of the DNA cytosine hypermethylation patterns caused by mutant IDH1 in the cells of individuals with AML. Our study provides proof of concept for the molecular and biological activity of novel allosteric inhibitors for targeting different mutant forms of IDH1 in leukemia.


Subject(s)
Dihydropyridines/pharmacology , Enzyme Inhibitors/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Pyrazoles/pharmacology , Allosteric Regulation , Allosteric Site , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , CpG Islands , Crystallography, X-Ray , Cytosine/chemistry , Cytosine/metabolism , DNA Methylation/drug effects , Dihydropyridines/chemistry , Dihydropyridines/pharmacokinetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Granulocytes/drug effects , Granulocytes/enzymology , Granulocytes/pathology , Humans , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Kinetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Models, Molecular , Mutation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Primary Cell Culture , Protein Binding , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Xenograft Model Antitumor Assays
9.
Curr Biol ; 24(16): 1909-17, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25127216

ABSTRACT

Tissue microenvironments are characterized not only in terms of chemical composition but also by collective properties such as stiffness, which influences the contractility of a cell, its adherent morphology, and even differentiation. The nucleoskeletal protein lamin-A,C increases with matrix stiffness, confers nuclear mechanical properties, and influences differentiation of mesenchymal stem cells (MSCs), whereas B-type lamins remain relatively constant. Here we show in single-cell analyses that matrix stiffness couples to myosin-II activity to promote lamin-A,C dephosphorylation at Ser22, which regulates turnover, lamina physical properties, and actomyosin expression. Lamin-A,C phosphorylation is low in interphase versus dividing cells, and its levels rise with states of nuclear rounding in which myosin-II generates little to no tension. Phosphorylated lamin-A,C localizes to nucleoplasm, and phosphorylation is enriched on lamin-A,C fragments and is suppressed by a cyclin-dependent kinase (CDK) inhibitor. Lamin-A,C knockdown in primary MSCs suppresses transcripts predominantly among actomyosin genes, especially in the serum response factor (SRF) pathway. Levels of myosin-IIA thus parallel levels of lamin-A,C, with phosphosite mutants revealing a key role for phosphoregulation. In modeling the system as a parsimonious gene circuit, we show that tension-dependent stabilization of lamin-A,C and myosin-IIA can suitably couple nuclear and cell morphology downstream of matrix mechanics.


Subject(s)
Extracellular Matrix/metabolism , Lamin Type A/genetics , Mesenchymal Stem Cells/metabolism , Nonmuscle Myosin Type IIA/genetics , Cell Differentiation , Elasticity , Feedback, Physiological , Humans , Lamin Type A/metabolism , Nonmuscle Myosin Type IIA/metabolism , Phosphorylation , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...