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.
Stem Cell Reports ; 14(4): 561-574, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32243840

ABSTRACT

Hematopoietic stem cells (HSCs) exist in a dormant state and progressively lose regenerative potency as they undergo successive divisions. Why this functional decline occurs and how this information is encoded is unclear. To better understand how this information is stored, we performed RNA sequencing on HSC populations differing only in their divisional history. Comparative analysis revealed that genes upregulated with divisions are enriched for lineage genes and regulated by cell-cycle-associated transcription factors, suggesting that proliferation itself drives lineage priming. Downregulated genes are, however, associated with an HSC signature and targeted by the Polycomb Repressive Complex 2 (PRC2). The PRC2 catalytic subunits Ezh1 and Ezh2 promote and suppress the HSC state, respectively, and successive divisions cause a switch from Ezh1 to Ezh2 dominance. We propose that cell divisions drive lineage priming and Ezh2 accumulation, which represses HSC signature genes to consolidate information on divisional history into memory.


Subject(s)
Cell Division , Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells/cytology , Animals , Cell Division/genetics , Cell Lineage/genetics , Cell Self Renewal , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Gene Expression Regulation , Hematopoiesis/genetics , Homeostasis , Male , Mice, Inbred C57BL , Models, Biological , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
2.
Cell Stem Cell ; 26(3): 359-376.e7, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32109377

ABSTRACT

Quiescence is a fundamental property that maintains hematopoietic stem cell (HSC) potency throughout life. Quiescent HSCs are thought to rely on glycolysis for their energy, but the overall metabolic properties of HSCs remain elusive. Using combined approaches, including single-cell RNA sequencing (RNA-seq), we show that mitochondrial membrane potential (MMP) distinguishes quiescent from cycling-primed HSCs. We found that primed, but not quiescent, HSCs relied readily on glycolysis. Notably, in vivo inhibition of glycolysis enhanced the competitive repopulation ability of primed HSCs. We further show that HSC quiescence is maintained by an abundance of large lysosomes. Repression of lysosomal activation in HSCs led to further enlargement of lysosomes while suppressing glucose uptake. This also induced increased lysosomal sequestration of mitochondria and enhanced the competitive repopulation ability of primed HSCs by over 90-fold in vivo. These findings show that restraining lysosomal activity preserves HSC quiescence and potency and may be therapeutically relevant.


Subject(s)
Hematopoietic Stem Cells , Mitochondria , Cell Division , Glycolysis , Hematopoietic Stem Cells/metabolism , Lysosomes , Mitochondria/metabolism
3.
FEBS Lett ; 593(23): 3266-3287, 2019 12.
Article in English | MEDLINE | ID: mdl-31557312

ABSTRACT

Transcription factor (TF)-based reprogramming of somatic tissues holds great promise for regenerative medicine. Previously, we demonstrated that the TFs GATA2, GFI1B, and FOS convert mouse and human fibroblasts to hemogenic endothelial-like precursors that generate hematopoietic stem progenitor (HSPC)-like cells over time. This conversion is lacking in robustness both in yield and biological function. Herein, we show that inclusion of GFI1 to the reprogramming cocktail significantly expands the HSPC-like population. AFT024 coculture imparts functional potential to these cells and allows quantification of stem cell frequency. Altogether, we demonstrate an improved human hemogenic induction protocol that could provide a valuable human in vitro model of hematopoiesis for disease modeling and a platform for cell-based therapeutics. DATABASE: Gene expression data are available in the Gene Expression Omnibus (GEO) database under the accession number GSE130361.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Hemangioblasts/cytology , Hematopoietic Stem Cells/cytology , Animals , Coculture Techniques/methods , Fibroblasts/cytology , Fibroblasts/metabolism , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Developmental/genetics , Hemangioblasts/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fos/genetics , Repressor Proteins/genetics , Transcription Factors/genetics
4.
Proc Natl Acad Sci U S A ; 115(47): E11128-E11137, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30385632

ABSTRACT

Osteosarcoma (OS), the most common primary bone tumor, is highly metastatic with high chemotherapeutic resistance and poor survival rates. Using induced pluripotent stem cells (iPSCs) generated from Li-Fraumeni syndrome (LFS) patients, we investigate an oncogenic role of secreted frizzled-related protein 2 (SFRP2) in p53 mutation-associated OS development. Interestingly, we find that high SFRP2 expression in OS patient samples correlates with poor survival. Systems-level analyses identified that expression of SFRP2 increases during LFS OS development and can induce angiogenesis. Ectopic SFRP2 overexpression in normal osteoblast precursors is sufficient to suppress normal osteoblast differentiation and to promote OS phenotypes through induction of oncogenic molecules such as FOXM1 and CYR61 in a ß-catenin-independent manner. Conversely, inhibition of SFRP2, FOXM1, or CYR61 represses the tumorigenic potential. In summary, these findings demonstrate the oncogenic role of SFRP2 in the development of p53 mutation-associated OS and that inhibition of SFRP2 is a potential therapeutic strategy.


Subject(s)
Bone Neoplasms/genetics , Carcinogenesis/genetics , Li-Fraumeni Syndrome/pathology , Membrane Proteins/genetics , Osteosarcoma/genetics , Tumor Suppressor Protein p53/genetics , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , Cysteine-Rich Protein 61/antagonists & inhibitors , Cysteine-Rich Protein 61/genetics , Forkhead Box Protein M1/antagonists & inhibitors , Forkhead Box Protein M1/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Li-Fraumeni Syndrome/genetics , Male , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Osteoblasts/cytology , Osteosarcoma/pathology
5.
Blood ; 129(14): 1901-1912, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28179275

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is used clinically to treat leukopenia and to enforce hematopoietic stem cell (HSC) mobilization to the peripheral blood (PB). However, G-CSF is also produced in response to infection, and excessive exposure reduces HSC repopulation capacity. Previous work has shown that dormant HSCs contain all the long-term repopulation potential in the bone marrow (BM), and that as HSCs accumulate a divisional history, they progressively lose regenerative potential. As G-CSF treatment also induces HSC proliferation, we sought to examine whether G-CSF-mediated repopulation defects are a result of increased proliferative history. To do so, we used an established H2BGFP label retaining system to track HSC divisions in response to G-CSF. Our results show that dormant HSCs are preferentially mobilized to the PB on G-CSF treatment. We find that this mobilization does not result in H2BGFP label dilution of dormant HSCs, suggesting that G-CSF does not stimulate dormant HSC proliferation. Instead, we find that proliferation within the HSC compartment is restricted to CD41-expressing cells that function with short-term, and primarily myeloid, regenerative potential. Finally, we show CD41 expression is up-regulated within the BM HSC compartment in response to G-CSF treatment. This emergent CD41Hi HSC fraction demonstrates no observable engraftment potential, but directly matures into megakaryocytes when placed in culture. Together, our results demonstrate that dormant HSCs mobilize in response to G-CSF treatment without dividing, and that G-CSF-mediated proliferation is restricted to cells with limited regenerative potential found within the HSC compartment.


Subject(s)
Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Platelet Membrane Glycoprotein IIb/biosynthesis , Animals , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/cytology , Mice , Mice, Transgenic , Platelet Membrane Glycoprotein IIb/genetics
6.
Cell ; 167(5): 1296-1309.e10, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27839867

ABSTRACT

The ability of cells to count and remember their divisions could underlie many alterations that occur during development, aging, and disease. We tracked the cumulative divisional history of slow-cycling hematopoietic stem cells (HSCs) throughout adult life. This revealed a fraction of rarely dividing HSCs that contained all the long-term HSC (LT-HSC) activity within the aging HSC compartment. During adult life, this population asynchronously completes four traceable symmetric self-renewal divisions to expand its size before entering a state of dormancy. We show that the mechanism of expansion involves progressively lengthening periods between cell divisions, with long-term regenerative potential lost upon a fifth division. Our data also show that age-related phenotypic changes within the HSC compartment are divisional history dependent. These results suggest that HSCs accumulate discrete memory stages over their divisional history and provide evidence for the role of cellular memory in HSC aging.


Subject(s)
Aging/pathology , Bone Marrow Cells/cytology , Hematopoietic Stem Cells/cytology , Animals , Bone Marrow Transplantation , Cell Cycle , Cell Division , Mice , Mice, Inbred C57BL , Platelet Membrane Glycoprotein IIb/metabolism
7.
Dev Cell ; 36(5): 525-39, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26954547

ABSTRACT

Definitive hematopoiesis emerges via an endothelial-to-hematopoietic transition in the embryo and placenta; however, the precursor cells to hemogenic endothelium are not defined phenotypically. We previously demonstrated that the induction of hematopoietic progenitors from fibroblasts progresses through hemogenic precursors that are Prom1(+)Sca1(+)CD34(+)CD45(-) (PS34CD45(-)). Guided by these studies, we analyzed mouse placentas and identified a population with this phenotype. These cells express endothelial markers, are heterogeneous for early hematopoietic markers, and localize to the vascular labyrinth. Remarkably, global gene expression profiles of PS34CD45(-) cells correlate with reprogrammed precursors and establish a hemogenic precursor cell molecular signature. PS34CD45(-) cells are also present in intra-embryonic hemogenic sites. After stromal co-culture, PS34CD45(-) cells give rise to all blood lineages and engraft primary and secondary immunodeficient mice. In summary, we show that reprogramming reveals a phenotype for in vivo precursors to hemogenic endothelium, establishing that direct in vitro conversion informs developmental processes in vivo.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Cellular Reprogramming , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Mouse Embryonic Stem Cells/cytology , Animals , Cells, Cultured , Endothelium/metabolism , Female , Fibroblasts/cytology , Mice , Mice, Inbred C57BL , Pregnancy
8.
J Vis Exp ; (118)2016 12 16.
Article in English | MEDLINE | ID: mdl-28060340

ABSTRACT

This protocol details the induction of a hemogenic program in mouse embryonic fibroblasts (MEFs) via overexpression of transcription factors (TFs). We first designed a reporter screen using MEFs from human CD34-tTA/TetO-H2BGFP (34/H2BGFP) double transgenic mice. CD34+ cells from these mice label H2B histones with GFP, and cease labeling upon addition of doxycycline (DOX). MEFS were transduced with candidate TFs and then observed for the emergence of GFP+ cells that would indicate the acquisition of a hematopoietic or endothelial cell fate. Starting with 18 candidate TFs, and through a process of combinatorial elimination, we obtained a minimal set of factors that would induce the highest percentage of GFP+ cells. We found that Gata2, Gfi1b, and cFos were necessary and the addition of Etv6 provided the optimal induction. A series of gene expression analyses done at different time points during the reprogramming process revealed that these cells appeared to go through a precursor cell that underwent an endothelial to hematopoietic transition (EHT). As such, this reprogramming process mimics developmental hematopoiesis "in a dish," allowing study of hematopoiesis in vitro and a platform to identify the mechanisms that underlie this specification. This methodology also provides a framework for translation of this work to the human system in the hopes of generating an alternative source of patient-specific hematopoietic stem cells (HSCs) for a number of applications in the treatment and study of hematologic diseases.


Subject(s)
Fibroblasts/physiology , Hematopoiesis/genetics , Transcription Factors/metabolism , Animals , Cell Differentiation , Fibroblasts/cytology , Gene Expression Regulation , Hematopoietic Stem Cells , Mice
9.
Stem Cells Dev ; 23(22): 2673-86, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25075441

ABSTRACT

Patient-specific induced pluripotent stem cells (iPSCs) are considered a versatile resource in the field of biomedicine. As iPSCs are generated on an individual basis, iPSCs may be the optimal cellular material to use for disease modeling, drug discovery, and the development of patient-specific cellular therapies. Recently, to gain an in-depth understanding of human pathologies, patient-specific iPSCs have been used to model human diseases with some iPSC-derived cells recapitulating pathological phenotypes in vitro. However, complex multigenic diseases generally have not resulted in concise conclusions regarding the underlying mechanisms of disease, in large part due to genetic variations between disease-state and control iPSCs. To circumvent this, the use of genomic editing tools to generate perfect isogenic controls is gaining momentum. To date, DNA binding domain-based zinc finger nucleases and transcription activator-like effector nucleases have been utilized to create genetically defined conditions in patient-specific iPSCs, with some examples leading to the successful identification of novel mechanisms of disease. As the feasibility and utility of genomic editing tools in iPSCs improve, along with the introduction of the clustered regularly interspaced short palindromic repeat system, understanding the features and limitations of genomic editing tools and their applications to iPSC technology is critical to expending the field of human disease modeling.


Subject(s)
Genetic Engineering/methods , Genetic Phenomena/genetics , Genome, Human/genetics , Induced Pluripotent Stem Cells/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...