Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Blood ; 140(14): 1592-1606, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35767701

ABSTRACT

Adult hematopoietic stem cells (HSCs) are predominantly quiescent and can be activated in response to acute stress such as infection or cytotoxic insults. STAT1 is a pivotal downstream mediator of interferon (IFN) signaling and is required for IFN-induced HSC proliferation, but little is known about the role of STAT1 in regulating homeostatic hematopoietic stem/progenitor cells (HSPCs). Here, we show that loss of STAT1 altered the steady state HSPC landscape, impaired HSC function in transplantation assays, delayed blood cell regeneration following myeloablation, and disrupted molecular programs that protect HSCs, including control of quiescence. Our results also reveal STAT1-dependent functional HSC heterogeneity. A previously unrecognized subset of homeostatic HSCs with elevated major histocompatibility complex class II (MHCII) expression (MHCIIhi) displayed molecular features of reduced cycling and apoptosis and was refractory to 5-fluorouracil-induced myeloablation. Conversely, MHCIIlo HSCs displayed increased megakaryocytic potential and were preferentially expanded in CALR mutant mice with thrombocytosis. Similar to mice, high MHCII expression is a feature of human HSCs residing in a deeper quiescent state. Our results therefore position STAT1 at the interface of stem cell heterogeneity and the interplay between stem cells and the adaptive immune system, areas of broad interest in the wider stem cell field.


Subject(s)
Hematopoietic Stem Cells , Megakaryocytes , STAT1 Transcription Factor , Animals , Cell Proliferation , Fluorouracil/pharmacology , Hematopoietic Stem Cells/metabolism , Humans , Interferons , Megakaryocytes/metabolism , Mice , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
2.
Blood ; 132(8): 791-803, 2018 08 23.
Article in English | MEDLINE | ID: mdl-29991556

ABSTRACT

Recent advances in single-cell technologies have permitted the investigation of heterogeneous cell populations at previously unattainable resolution. Here we apply such approaches to resolve the molecular mechanisms driving disease in mouse hematopoietic stem cells (HSCs), using JAK2V617F mutant myeloproliferative neoplasms (MPNs) as a model. Single-cell gene expression and functional assays identified a subset of JAK2V617F mutant HSCs that display defective self-renewal. This defect is rescued at the single HSC level by crossing JAK2V617F mice with mice lacking TET2, the most commonly comutated gene in patients with MPN. Single-cell gene expression profiling of JAK2V617F-mutant HSCs revealed a loss of specific regulator genes, some of which were restored to normal levels in single TET2/JAK2 mutant HSCs. Of these, Bmi1 and, to a lesser extent, Pbx1 and Meis1 overexpression in JAK2-mutant HSCs could drive a disease phenotype and retain durable stem cell self-renewal in functional assays. Together, these single-cell approaches refine the molecules involved in clonal expansion of MPNs and have broad implications for deconstructing the molecular network of normal and malignant stem cells.


Subject(s)
Cell Self Renewal , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/metabolism , Hematopoietic Stem Cells/metabolism , Janus Kinase 2/metabolism , Mutation, Missense , Myeloproliferative Disorders/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Amino Acid Substitution , Animals , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Hematopoietic Stem Cells/pathology , Janus Kinase 2/genetics , Mice , Mice, Transgenic , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology
3.
Blood ; 131(6): 649-661, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29282219

ABSTRACT

Somatic mutations in the endoplasmic reticulum chaperone calreticulin (CALR) are detected in approximately 40% of patients with essential thrombocythemia (ET) and primary myelofibrosis (PMF). Multiple different mutations have been reported, but all result in a +1-bp frameshift and generate a novel protein C terminus. In this study, we generated a conditional mouse knockin model of the most common CALR mutation, a 52-bp deletion. The mutant novel human C-terminal sequence is integrated into the otherwise intact mouse CALR gene and results in mutant CALR expression under the control of the endogenous mouse locus. CALRdel/+ mice develop a transplantable ET-like disease with marked thrombocytosis, which is associated with increased and morphologically abnormal megakaryocytes and increased numbers of phenotypically defined hematopoietic stem cells (HSCs). Homozygous CALRdel/del mice developed extreme thrombocytosis accompanied by features of MF, including leukocytosis, reduced hematocrit, splenomegaly, and increased bone marrow reticulin. CALRdel/+ HSCs were more proliferative in vitro, but neither CALRdel/+ nor CALRdel/del displayed a competitive transplantation advantage in primary or secondary recipient mice. These results demonstrate the consequences of heterozygous and homozygous CALR mutations and provide a powerful model for dissecting the pathogenesis of CALR-mutant ET and PMF.


Subject(s)
Calreticulin/genetics , Cell Self Renewal/genetics , Hematopoietic Stem Cells/physiology , Primary Myelofibrosis/genetics , Thrombocytosis/genetics , Animals , Cells, Cultured , Homozygote , Leukocytosis/genetics , Leukocytosis/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Splenomegaly/genetics , Splenomegaly/pathology , Thrombocythemia, Essential/genetics , Thrombocythemia, Essential/pathology
5.
Cell Rep ; 19(8): 1503-1511, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28538171

ABSTRACT

Aging of the hematopoietic stem cell (HSC) compartment is characterized by lineage bias and reduced stem cell function, the molecular basis of which is largely unknown. Using single-cell transcriptomics, we identified a distinct subpopulation of old HSCs carrying a p53 signature indicative of stem cell decline alongside pro-proliferative JAK/STAT signaling. To investigate the relationship between JAK/STAT and p53 signaling, we challenged HSCs with a constitutively active form of JAK2 (V617F) and observed an expansion of the p53-positive subpopulation in old mice. Our results reveal cellular heterogeneity in the onset of HSC aging and implicate a role for JAK2V617F-driven proliferation in the p53-mediated functional decline of old HSCs.


Subject(s)
Cell Compartmentation , Cellular Senescence , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Cell Cycle , Cell Proliferation , Janus Kinase 2 , Mice , Myeloid Cells/metabolism , Transcription Factors/metabolism
6.
Nat Commun ; 7: 11208, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27052461

ABSTRACT

The production of megakaryocytes (MKs)--the precursors of blood platelets--from human pluripotent stem cells (hPSCs) offers exciting clinical opportunities for transfusion medicine. Here we describe an original approach for the large-scale generation of MKs in chemically defined conditions using a forward programming strategy relying on the concurrent exogenous expression of three transcription factors: GATA1, FLI1 and TAL1. The forward programmed MKs proliferate and differentiate in culture for several months with MK purity over 90% reaching up to 2 × 10(5) mature MKs per input hPSC. Functional platelets are generated throughout the culture allowing the prospective collection of several transfusion units from as few as 1 million starting hPSCs. The high cell purity and yield achieved by MK forward programming, combined with efficient cryopreservation and good manufacturing practice (GMP)-compatible culture, make this approach eminently suitable to both in vitro production of platelets for transfusion and basic research in MK and platelet biology.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cellular Reprogramming , GATA1 Transcription Factor/genetics , Megakaryocytes/cytology , Pluripotent Stem Cells/cytology , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cryopreservation/methods , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/genetics , Megakaryocytes/metabolism , Microarray Analysis , Pluripotent Stem Cells/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transduction, Genetic , Transgenes
7.
Blood ; 123(20): 3139-51, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24692758

ABSTRACT

Genomic regions of acquired uniparental disomy (UPD) are common in malignancy and frequently harbor mutated oncogenes. Homozygosity for such gain-of-function mutations is thought to modulate tumor phenotype, but direct evidence has been elusive. Polycythemia vera (PV) and essential thrombocythemia (ET), 2 subtypes of myeloproliferative neoplasms, are associated with an identical acquired JAK2V617F mutation but the mechanisms responsible for distinct clinical phenotypes remain unclear. We provide direct genetic evidence and demonstrate that homozygosity for human JAK2V617F in knock-in mice results in a striking phenotypic switch from an ET-like to PV-like phenotype. The resultant erythrocytosis is driven by increased numbers of early erythroid progenitors and enhanced erythroblast proliferation, whereas reduced platelet numbers are associated with impaired platelet survival. JAK2V617F-homozygous mice developed a severe hematopoietic stem cell defect, suggesting that additional lesions are needed to sustain clonal expansion. Together, our results indicate that UPD for 9p plays a causal role in the PV phenotype in patients as a consequence of JAK2V617F homozygosity. The generation of a JAK2V617F allelic series of mice with a dose-dependent effect on hematopoiesis provides a powerful model for studying the consequences of mutant JAK2 homozygosity.


Subject(s)
Janus Kinase 2/genetics , Mutation , Polycythemia Vera/genetics , Thrombocythemia, Essential/genetics , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Erythroblasts/metabolism , Erythroblasts/pathology , Female , Gene Knock-In Techniques , Homozygote , Humans , Male , Megakaryocytes/metabolism , Megakaryocytes/pathology , Mice , Mice, Inbred C57BL , Phenotype , Polycythemia Vera/pathology , Thrombocythemia, Essential/pathology , Uniparental Disomy/genetics , Uniparental Disomy/pathology
8.
PLoS Biol ; 11(6): e1001576, 2013.
Article in English | MEDLINE | ID: mdl-23750118

ABSTRACT

Recent descriptions of significant heterogeneity in normal stem cells and cancers have altered our understanding of tumorigenesis, emphasizing the need to understand how single stem cells are subverted to cause tumors. Human myeloproliferative neoplasms (MPNs) are thought to reflect transformation of a hematopoietic stem cell (HSC) and the majority harbor an acquired V617F mutation in the JAK2 tyrosine kinase, making them a paradigm for studying the early stages of tumor establishment and progression. The consequences of activating tyrosine kinase mutations for stem and progenitor cell behavior are unclear. In this article, we identify a distinct cellular mechanism operative in stem cells. By using conditional knock-in mice, we show that the HSC defect resulting from expression of heterozygous human JAK2V617F is both quantitative (reduced HSC numbers) and qualitative (lineage biases and reduced self-renewal per HSC). The defect is intrinsic to individual HSCs and their progeny are skewed toward proliferation and differentiation as evidenced by single cell and transplantation assays. Aged JAK2V617F show a more pronounced defect as assessed by transplantation, but mice that transform reacquire competitive self-renewal ability. Quantitative analysis of HSC-derived clones was used to model the fate choices of normal and JAK2-mutant HSCs and indicates that JAK2V617F reduces self-renewal of individual HSCs but leaves progenitor expansion intact. This conclusion is supported by paired daughter cell analyses, which indicate that JAK2-mutant HSCs more often give rise to two differentiated daughter cells. Together these data suggest that acquisition of JAK2V617F alone is insufficient for clonal expansion and disease progression and causes eventual HSC exhaustion. Moreover, our results show that clonal expansion of progenitor cells provides a window in which collaborating mutations can accumulate to drive disease progression. Characterizing the mechanism(s) of JAK2V617F subclinical clonal expansions and the transition to overt MPNs will illuminate the earliest stages of tumor establishment and subclone competition, fundamentally shifting the way we treat and manage cancers.


Subject(s)
Amino Acid Substitution/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Janus Kinase 2/genetics , Mutation/genetics , Animals , Antigens, CD/metabolism , Cell Count , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Proliferation , Clone Cells , Gene Knock-In Techniques , Hematopoietic Stem Cell Transplantation , Humans , Mice , Myeloproliferative Disorders/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...