Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
J Anesth ; 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138698

ABSTRACT

PURPOSE: Prone position has recently gained renewed importance as a treatment for acute respiratory distress syndrome and spine and brain surgeries. Our study aimed to perform an error grid analysis to examine the clinical discrepancies between arterial blood pressure (ABP) and non-invasive blood pressure (NIBP) in the prone position and to investigate the risk factors influencing these differences. METHODS: Error grid analysis was performed retrospectively on 1389 pairs of 100 consecutive prone positioning cases. This analysis classifies the difference between the two methods into five clinically relevant zones, from "no risk" to "dangerous risk". Additionally, multivariable ordinal logistic regression analysis was conducted to evaluate the relationship between the risk zones of mean blood pressure (MBP), as classified by error grid analysis and the covariate of interest. RESULTS: Error grid analysis showed that the proportions of measurement pairs in risk zones A-E for systolic blood pressure were 96.8%, 3.2%, 0.1%, 0%, and 0%, respectively. In contrast, the MBP proportions were 74.0%, 25.1%, 0.9%, 0.1%, and 0%. Multivariable ordinal logistic regression analysis revealed that the position of arms (next to the head) was a significant factor (adjusted odds ratio: 4.35, 95% CI: 2.38-8.33, P < 0.001). CONCLUSION: Error grid analysis revealed a clinically unacceptable discrepancy between ABP and NIBP for MBP during prone positioning surgery. The position of the arms next to the head was associated with increased clinical discrepancy between the two MBP measurement methods.

2.
bioRxiv ; 2024 May 24.
Article in English | MEDLINE | ID: mdl-38826462

ABSTRACT

Normal hematopoietic stem and progenitor cells (HSPCs) inherently accumulate somatic mutations and lose clonal diversity with age, processes implicated in the development of myeloid malignancies 1 . The impact of exogenous stressors, such as cancer chemotherapies, on the genomic integrity and clonal dynamics of normal HSPCs is not well defined. We conducted whole-genome sequencing on 1,032 single-cell-derived HSPC colonies from 10 patients with multiple myeloma (MM), who had undergone various chemotherapy regimens. Our findings reveal that melphalan treatment distinctly increases mutational burden with a unique mutation signature, whereas other MM chemotherapies do not significantly affect the normal mutation rate of HSPCs. Among these therapy-induced mutations were several oncogenic drivers such as TET2 and PPM1D . Phylogenetic analysis showed a clonal architecture in post-treatment HSPCs characterized by extensive convergent evolution of mutations in genes such as TP53 and PPM1D . Consequently, the clonal diversity and structure of post-treatment HSPCs mirror those observed in normal elderly individuals, suggesting an accelerated clonal aging due to chemotherapy. Furthermore, analysis of matched therapy-related myeloid neoplasm (t-MN) samples, which occurred 1-8 years later, enabled us to trace the clonal origin of t-MNs to a single HSPC clone among a group of clones with competing malignant potential, indicating the critical role of secondary mutations in dictating clonal dominance and malignant transformation. Our findings suggest that cancer chemotherapy promotes an oligoclonal architecture with multiple HSPC clones possessing competing leukemic potentials, setting the stage for the selective emergence of a singular clone that evolves into t-MNs after acquiring secondary mutations. These results underscore the importance of further systematic research to elucidate the long-term hematological consequences of cancer chemotherapy.

3.
Int J Hematol ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750343

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous hematologic malignancy primarily driven by an immature population of AML cells termed leukemia stem cells (LSCs) that are implicated in AML development, chemoresistance, and relapse. An emerging area of research in AML focuses on identifying and targeting the aberrant metabolism in LSCs. Dysregulated metabolism is involved in sustaining functional properties of LSCs, impeding myeloid differentiation, and evading programmed cell death, both in the process of leukemogenesis and in response to chemotherapy. This review discusses recent discoveries regarding the aberrant metabolic processes of AML LSCs that have begun to change the therapeutic landscape of AML.

4.
Nat Commun ; 15(1): 538, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38225226

ABSTRACT

Hematopoietic stem cells (HSCs) are capable of regenerating the blood system, but the instructive cues that direct HSCs to regenerate particular lineages lost to the injury remain elusive. Here, we show that iron is increasingly taken up by HSCs during anemia and induces erythroid gene expression and regeneration in a Tet2-dependent manner. Lineage tracing of HSCs reveals that HSCs respond to hemolytic anemia by increasing erythroid output. The number of HSCs in the spleen, but not bone marrow, increases upon anemia and these HSCs exhibit enhanced proliferation, erythroid differentiation, iron uptake, and TET2 protein expression. Increased iron in HSCs promotes DNA demethylation and expression of erythroid genes. Suppressing iron uptake or TET2 expression impairs erythroid genes expression and erythroid differentiation of HSCs; iron supplementation, however, augments these processes. These results establish that the physiological level of iron taken up by HSCs has an instructive role in promoting erythroid-biased differentiation of HSCs.


Subject(s)
Anemia , Dioxygenases , Humans , Spleen , Hematopoietic Stem Cells/metabolism , Cell Differentiation , Iron/metabolism , Anemia/metabolism , Erythroid Cells , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism
5.
Genes Dev ; 36(15-16): 887-900, 2022 08 01.
Article in English | MEDLINE | ID: mdl-36167470

ABSTRACT

The polycomb complex component Bmi1 promotes the maintenance of stem cells in multiple postnatal tissues, partly by negatively regulating the expression of p16Ink4a and p19Arf, tumor suppressors associated with cellular senescence. However, deficiency for p16Ink4a and p19Arf only partially rescues the function of Bmi1-deficient stem cells. We conditionally deleted Bmi1 from adult hematopoietic cells and found that this slowly depleted hematopoietic stem cells (HSCs). Rather than inducing senescence, Bmi1 deficiency increased HSC division. The increased cell division was caused partly by increased Aristaless-related homeobox (ARX) transcription factor expression, which also increased ribosomal RNA expression. However, ARX deficiency did not rescue HSC depletion. Bmi1 deficiency also increased protein synthesis, protein aggregation, and protein ubiquitylation independent of its effects on cell division and p16Ink4a, p19Arf, and ARX expression. Bmi1 thus promotes HSC quiescence by negatively regulating ARX expression and promotes proteostasis by suppressing protein synthesis. This highlights a new connection between the regulation of stem cell maintenance and proteostasis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16 , Proteostasis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Hematopoietic Stem Cells , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Protein Aggregates , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Ribosomal/metabolism
7.
Cell Stem Cell ; 29(3): 386-399.e7, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35108519

ABSTRACT

Deregulation of transcription is a hallmark of acute myeloid leukemia (AML) that drives oncogenic expression programs and presents opportunities for therapeutic targeting. By integrating comprehensive pan-cancer enhancer landscapes with genetic dependency mapping, we find that AML-enriched enhancers encode for more selective tumor dependencies. We hypothesized that this approach could identify actionable dependencies downstream of oncogenic driver events and discovered a MYB-regulated AML-enriched enhancer regulating SEPHS2, a key component of the selenoprotein production pathway. Using a combination of patient samples and mouse models, we show that this enhancer upregulates SEPHS2, promoting selenoprotein production and antioxidant function required for AML survival. SEPHS2 and other selenoprotein pathway genes are required for AML growth in vitro. SEPHS2 knockout and selenium dietary restriction significantly delay leukemogenesis in vivo with little effect on normal hematopoiesis. These data validate the utility of enhancer mapping in target identification and suggest that selenoprotein production is an actionable target in AML.


Subject(s)
Leukemia, Myeloid, Acute , Selenium , Animals , Carcinogenesis/genetics , Enhancer Elements, Genetic/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Oncogenes , Selenium/therapeutic use
8.
Commun Biol ; 5(1): 72, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058565

ABSTRACT

Early diagnosis of acute myeloid leukemia (AML) in the pre-leukemic stage remains a clinical challenge, as pre-leukemic patients show no symptoms, lacking any known morphological or numerical abnormalities in blood cells. Here, we demonstrate that platelets with structurally abnormal mitochondria emerge at the pre-leukemic phase of AML, preceding detectable changes in blood cell counts or detection of leukemic blasts in blood. We visualized frozen-hydrated platelets from mice at different time points during AML development in situ using electron cryo-tomography (cryo-ET) and identified intracellular organelles through an unbiased semi-automatic process followed by quantitative measurement. A large proportion of platelets exhibited changes in the overall shape and depletion of organelles in AML. Notably, 23% of platelets in pre-leukemic cells exhibit abnormal, round mitochondria with unfolded cristae, accompanied by a significant drop in ATP levels and altered expression of metabolism-related gene signatures. Our study demonstrates that detectable structural changes in pre-leukemic platelets may serve as a biomarker for the early diagnosis of AML.


Subject(s)
Blood Platelets/cytology , Hematopoiesis , Leukemia, Myeloid, Acute/diagnosis , Tomography, X-Ray Computed/methods , Animals , Female , Mice
9.
Stem Cell Reports ; 16(8): 2014-2028, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34242617

ABSTRACT

Histone variants contribute to the complexity of the chromatin landscape and play an integral role in defining DNA domains and regulating gene expression. The histone H3 variant H3.3 is incorporated into genic elements independent of DNA replication by its chaperone HIRA. Here we demonstrate that Hira is required for the self-renewal of adult hematopoietic stem cells (HSCs) and to restrain erythroid differentiation. Deletion of Hira led to rapid depletion of HSCs while differentiated hematopoietic cells remained largely unaffected. Depletion of HSCs after Hira deletion was accompanied by increased expression of bivalent and erythroid genes, which was exacerbated upon cell division and paralleled increased erythroid differentiation. Assessing H3.3 occupancy identified a subset of polycomb-repressed chromatin in HSCs that depends on HIRA to maintain the inaccessible, H3.3-occupied state for gene repression. HIRA-dependent H3.3 incorporation thus defines distinct repressive chromatin that represses erythroid differentiation of HSCs.


Subject(s)
Adult Stem Cells/metabolism , Cell Cycle Proteins/genetics , Cell Differentiation/genetics , Erythroid Cells/metabolism , Hematopoietic Stem Cells/metabolism , Histone Chaperones/genetics , Transcription Factors/genetics , Age Factors , Animals , Animals, Newborn , Cell Cycle Proteins/metabolism , Cell Self Renewal/genetics , Gene Expression Profiling/methods , Gene Ontology , Hematopoiesis/genetics , Histone Chaperones/metabolism , Histones/genetics , Histones/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA-Seq/methods , Transcription Factors/metabolism
10.
Sci Adv ; 7(30)2021 07.
Article in English | MEDLINE | ID: mdl-34290089

ABSTRACT

Metabolic dysregulation underlies malignant phenotypes attributed to cancer stem cells, such as unlimited proliferation and differentiation blockade. Here, we demonstrate that NAD+ metabolism enables acute myeloid leukemia (AML) to evade apoptosis, another hallmark of cancer stem cells. We integrated whole-genome CRISPR screening and pan-cancer genetic dependency mapping to identify NAMPT and NMNAT1 as AML dependencies governing NAD+ biosynthesis. While both NAMPT and NMNAT1 were required for AML, the presence of NAD+ precursors bypassed the dependence of AML on NAMPT but not NMNAT1, pointing to NMNAT1 as a gatekeeper of NAD+ biosynthesis. Deletion of NMNAT1 reduced nuclear NAD+, activated p53, and increased venetoclax sensitivity. Conversely, increased NAD+ biosynthesis promoted venetoclax resistance. Unlike leukemia stem cells (LSCs) in both murine and human AML xenograft models, NMNAT1 was dispensable for hematopoietic stem cells and hematopoiesis. Our findings identify NMNAT1 as a previously unidentified therapeutic target that maintains NAD+ for AML progression and chemoresistance.


Subject(s)
Leukemia, Myeloid, Acute , Nicotinamide-Nucleotide Adenylyltransferase , Animals , Apoptosis/genetics , Homeostasis , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mice , NAD/metabolism , Neoplastic Stem Cells/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism
12.
Stem Cell Reports ; 16(4): 741-753, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33770496

ABSTRACT

Hematopoiesis serves as a paradigm for how homeostasis is maintained within hierarchically organized cell populations. However, important questions remain as to the contribution of hematopoietic stem cells (HSCs) toward maintaining steady state hematopoiesis. A number of in vivo lineage labeling and propagation studies have given rise to contradictory interpretations, leaving key properties of stem cell function unresolved. Using processed flow cytometry data coupled with a biology-driven modeling approach, we show that in vivo flux experiments that come from different laboratories can all be reconciled into a single unifying model, even though they had previously been interpreted as being contradictory. We infer from comparative analysis that different transgenic models display distinct labeling efficiencies across a heterogeneous HSC pool, which we validate by marker gene expression associated with HSC function. Finally, we show how the unified model of HSC differentiation can be used to simulate clonal expansion in the early stages of leukemogenesis.


Subject(s)
Hematopoietic Stem Cells/metabolism , Leukemia/pathology , Models, Biological , Animals , Biomarkers/metabolism , Carcinogenesis/pathology , Cell Self Renewal , Guanine Nucleotide Exchange Factors/metabolism , Integrases/metabolism , Kinetics , Mice, Transgenic , Receptor, TIE-2/metabolism , Staining and Labeling
14.
Nat Commun ; 11(1): 5327, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087716

ABSTRACT

Clonal diversity is a consequence of cancer cell evolution driven by Darwinian selection. Precise characterization of clonal architecture is essential to understand the evolutionary history of tumor development and its association with treatment resistance. Here, using a single-cell DNA sequencing, we report the clonal architecture and mutational histories of 123 acute myeloid leukemia (AML) patients. The single-cell data reveals cell-level mutation co-occurrence and enables reconstruction of mutational histories characterized by linear and branching patterns of clonal evolution, with the latter including convergent evolution. Through xenotransplantion, we show leukemia initiating capabilities of individual subclones evolving in parallel. Also, by simultaneous single-cell DNA and cell surface protein analysis, we illustrate both genetic and phenotypic evolution in AML. Lastly, single-cell analysis of longitudinal samples reveals underlying evolutionary process of therapeutic resistance. Together, these data unravel clonal diversity and evolution patterns of AML, and highlight their clinical relevance in the era of precision medicine.


Subject(s)
Clonal Evolution/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Aged , Animals , Clonal Evolution/drug effects , Cohort Studies , Female , Genetic Association Studies , Genomics , Heterografts , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/drug therapy , Male , Mice , Mice, Inbred NOD , Middle Aged , Models, Genetic , Mutation , Sequence Analysis, DNA , Single-Cell Analysis
15.
Nat Cell Biol ; 22(10): 1162-1169, 2020 10.
Article in English | MEDLINE | ID: mdl-32958856

ABSTRACT

Stem cells need to be protected from genotoxic and proteotoxic stress to maintain a healthy pool throughout life1-3. Little is known about the proteostasis mechanism that safeguards stem cells. Here we report endoplasmic reticulum-associated degradation (ERAD) as a protein quality checkpoint that controls the haematopoietic stem cell (HSC)-niche interaction and determines the fate of HSCs. The SEL1L-HRD1 complex, the most conserved branch of ERAD4, is highly expressed in HSCs. Deletion of Sel1l led to niche displacement of HSCs and a complete loss of HSC identity, and allowed highly efficient donor-HSC engraftment without irradiation. Mechanistic studies identified MPL, the master regulator of HSC identity5, as a bona fide ERAD substrate that became aggregated in the endoplasmic reticulum following ERAD deficiency. Restoration of MPL signalling with an agonist partially rescued the number and reconstitution capacity of Sel1l-deficient HSCs. Our study defines ERAD as an essential proteostasis mechanism to safeguard a healthy stem cell pool by regulating the stem cell-niche interaction.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/metabolism , Hematopoietic Stem Cells/cytology , Intracellular Signaling Peptides and Proteins/physiology , Receptors, Thrombopoietin/metabolism , Stem Cell Niche , Ubiquitin-Protein Ligases/metabolism , Animals , Female , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Thrombopoietin/genetics , Ubiquitin-Protein Ligases/genetics
16.
Nat Cell Biol ; 22(2): 225-234, 2020 02.
Article in English | MEDLINE | ID: mdl-32029897

ABSTRACT

Energy stress depletes ATP and induces cell death. Here we identify an unexpected inhibitory role of energy stress on ferroptosis, a form of regulated cell death induced by iron-dependent lipid peroxidation. We found that ferroptotic cell death and lipid peroxidation can be inhibited by treatments that induce or mimic energy stress. Inactivation of AMP-activated protein kinase (AMPK), a sensor of cellular energy status, largely abolishes the protective effects of energy stress on ferroptosis in vitro and on ferroptosis-associated renal ischaemia-reperfusion injury in vivo. Cancer cells with high basal AMPK activation are resistant to ferroptosis and AMPK inactivation sensitizes these cells to ferroptosis. Functional and lipidomic analyses further link AMPK regulation of ferroptosis to AMPK-mediated phosphorylation of acetyl-CoA carboxylase and polyunsaturated fatty acid biosynthesis. Our study demonstrates that energy stress inhibits ferroptosis partly through AMPK and reveals an unexpected coupling between ferroptosis and AMPK-mediated energy-stress signalling.


Subject(s)
AMP-Activated Protein Kinases/genetics , Acetyl-CoA Carboxylase/genetics , Ferroptosis/genetics , Kidney/enzymology , Lipid Peroxidation/genetics , Reperfusion Injury/genetics , A549 Cells , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Cell Line, Tumor , Cyclohexylamines/pharmacology , Embryo, Mammalian , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Acids, Unsaturated/biosynthesis , Ferroptosis/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Glucose/deficiency , Glucose/pharmacology , Humans , Iron/metabolism , Kidney/drug effects , Kidney/pathology , Lipid Peroxidation/drug effects , MCF-7 Cells , Mice , Mice, Transgenic , Phenylenediamines/pharmacology , Phosphorylation , Piperazines/antagonists & inhibitors , Piperazines/pharmacology , Primary Cell Culture , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction , Stress, Physiological/drug effects , Stress, Physiological/genetics
17.
Exp Hematol ; 83: 66-73, 2020 03.
Article in English | MEDLINE | ID: mdl-31893524

ABSTRACT

Clonal hematopoiesis (CH) has emerged as an important factor linked to adverse health conditions in the elderly. CH is characterized by an overrepresentation of genetically distinct hematopoietic stem cell clones in the peripheral blood. Whereas the genetic mutations that underlie CH have been closely scrutinized, relatively little attention has been paid to the environmental factors that may influence the emergence of one dominant stem cell clone. As there is huge individual variation in latency between acquisition of a genetic mutation and emergence of CH, environmental factors likely play a major role. Indeed, environmental stressors such as inflammation, chemotherapy, and metabolic syndromes are known to affect steady-state hematopoiesis. To date, epidemiologic studies point toward smoking and prior chemotherapy exposure as likely contributors to some forms of CH, though the impact of other environmental factors is also being investigated. Mechanistic studies in murine models indicate that the role of different environmental factors in CH emergence may be highly specific to the mutation that marks each stem cell clone. For instance, recent studies have found that clones with mutations in the PPM1D gene are more resistant to genotoxic stress induced by chemotherapy. These clones thus have a competitive advantage in the setting of chemotherapy, but not in other types of stress. Here we review currently available literature on the interplay between environment and the genetic landscapes in CH and highlight critical areas for future study. Improved understanding of the effects of environmental stress on emergence of CH with mutation-specific clarity will guide future efforts to provide preventive medicine to individuals with CH.


Subject(s)
Clonal Evolution , Environmental Exposure/adverse effects , Hematologic Diseases , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Mutation , Animals , DNA Damage , Disease Models, Animal , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Hematopoietic Stem Cells/pathology , Humans , Mice , Protein Phosphatase 2C/genetics , Protein Phosphatase 2C/metabolism
18.
Blood ; 134(24): 2183-2194, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31697807

ABSTRACT

Altered metabolism fuels 2 hallmark properties of cancer cells: unlimited proliferation and differentiation blockade. Adenosine monophosphate-activated protein kinase (AMPK) is a master regulator of bioenergetics crucial for glucose metabolism in acute myeloid leukemia (AML), and its inhibition delays leukemogenesis, but whether the metabolic function of AMPK alters the AML epigenome remains unknown. Here, we demonstrate that AMPK maintains the epigenome of MLL-rearranged AML by linking acetyl-coenzyme A (CoA) homeostasis to Bromodomain and Extra-Terminal domain (BET) protein recruitment to chromatin. AMPK deletion reduced acetyl-CoA and histone acetylation, displacing BET proteins from chromatin in leukemia-initiating cells. In both mouse and patient-derived xenograft AML models, treating with AMPK and BET inhibitors synergistically suppressed AML. Our results provide a therapeutic rationale to target AMPK and BET for AML therapy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acetyl Coenzyme A/metabolism , Cell Cycle Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Transcription Factors/metabolism , Acetylation , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Leukemic , Histones/metabolism , Homeostasis , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/pathology , Mice , Neoplasm Grading , Protein Binding , Xenograft Model Antitumor Assays
19.
Blood ; 134(7): 614-625, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31270104

ABSTRACT

Oncogenic mutations confer on cells the ability to propagate indefinitely, but whether oncogenes alter the cell fate of these cells is unknown. Here, we show that the transcriptional regulator PRDM16s causes oncogenic fate conversion by transforming cells fated to form platelets and erythrocytes into myeloid leukemia stem cells (LSCs). Prdm16s expression in megakaryocyte-erythroid progenitors (MEPs), which normally lack the potential to generate granulomonocytic cells, caused AML by converting MEPs into LSCs. Prdm16s blocked megakaryocytic/erythroid potential by interacting with super enhancers and activating myeloid master regulators, including PU.1. A CRISPR dropout screen confirmed that PU.1 is required for Prdm16s-induced leukemia. Ablating PU.1 attenuated leukemogenesis and reinstated the megakaryocytic/erythroid potential of leukemic MEPs in mouse models and human AML with PRDM16 rearrangement. Thus, oncogenic PRDM16 s expression gives MEPs an LSC fate by activating myeloid gene regulatory networks.


Subject(s)
Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/pathology , Megakaryocyte-Erythroid Progenitor Cells/pathology , Transcription Factors/genetics , Animals , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Leukemic , Gene Regulatory Networks , Humans , Leukemia, Myeloid, Acute/genetics , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Mice, Inbred C57BL , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Translocation, Genetic
20.
Stem Cell Reports ; 13(3): 545-558, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31257132

ABSTRACT

Bone marrow stromal cells (BMSCs) that express high levels of stem cell factor (SCF) and CXC chemokine ligand 12 (CXCL12) are one crucial component of the hematopoietic stem cell (HSC) niche. While the secreted factors produced by BMSCs to support HSCs have been well described, little is known regarding the transcriptional regulators controlling the cell fate of BMSCs and thus indirectly maintaining HSCs. BMI1 is a polycomb group protein that regulates HSCs both cell intrinsically and extrinsically, but it is unknown in which cell type and how BMI1 functions to maintain HSCs extrinsically. Here we show that Bmi1 maintains HSCs by preventing adipogenic differentiation of BMSCs. Bmi1 is highly expressed in BMSCs but becomes downregulated upon adipogenic differentiation and during aging. Deleting Bmi1 from BMSCs increased marrow adipocytes, induced HSC quiescence and depletion, and impaired hematopoiesis. We found that BMI1 repressed multiple developmental programs in BMSCs by safeguarding the repressive epigenetic marks histone H2A ubiquitylation and H3 lysine 27 trimethylation. We identified a novel adipogenic program governed by Pax3, which BMI1 repressed in BMSCs. Our results establish Bmi1 as a critical regulator of BMSC cell fate that suppresses marrow adipogenesis to create a supportive niche for HSCs.


Subject(s)
Adipocytes/metabolism , Mesenchymal Stem Cells/cytology , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Adipocytes/cytology , Adipogenesis , Animals , Cell Differentiation , Cellular Senescence , Female , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Male , Mesenchymal Stem Cells/metabolism , Methylation , Mice , Mice, Inbred C57BL , PAX3 Transcription Factor/genetics , PAX3 Transcription Factor/metabolism , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL