Regulation of Hematopoietic Stem Cell Quiescence - A Novel Role for p53.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 92-92
Author(s):  
Yan Liu ◽  
Shannon E. Elf ◽  
Yasuhiko Miyata ◽  
Goro Sashida ◽  
Anthony D. Deblasio ◽  
...  

Abstract Although the p53 tumor suppressor can elicit cell-cycle arrest or apoptosis in hematopoietic cells upon DNA damage, its function during steady-state hematopoiesis is largely unknown. We demonstrated that the Ets transcription factor MEF/ELF4 regulates both HSC proliferation/self-renewal and quiescence, as Mef null mice exhibit greater numbers of hematopoietic stem cells and the Mef null HSCs are more quiescent than normal. As such, the hematopoietic compartment of Mef null mice shows significant resistance to chemotherapy and radiation (Lacorazza et al., Cancer Cell, 2006). In this study, we have investigated the mechanisms utilized by MEF/ELF4 to regulate the quiescence and self-renewal of hematopoietic stem cells, identifying p53 as a key regulator. We have recently found that Mef null mouse embryonic fibroblasts (mefs) accumulate p53 and undergo premature senescence; MEF appears to surpress the expression of p53 by directly upregulating Mdm2 (G. Sashida et al., submitted). We hypothesized that p53 may play a role in the enhanced stem cell quiescence or the increased HSC frequency seen in Mef null mice. To examine this, we generated p53−/− Mef −/− mice and compared HSC biology in the double knock out mice (p53−/− Mef −/−) vs p53 null mice, Mef null mice and wt mice. Loss of p53 decreased the fraction of Pyronin Ylow Lin-Sca-1+ cells, suggesting fewer quiescent HSCs, and staining of CD34-LSK cells for the proliferation marker Ki67 also showed enhanced HSC proliferation in the absence of p53 (with fewer quiescent cells present). These data suggest that p53 promotes quiescence in HSCs, and in the absence of p53, HSCs more readily enter the cell cycle. When we analyzed the DKO (p53−/− Mef −/−) mice, we observed that the percentage of G0 HSCs returned to normal, indicating that p53 is essential for maintaining the enhanced quiescence of MEF null HSCs. p21 is a major target gene of p53 in cells, and has been shown to play an important role in maintaining HSC quiescence. As expeceted, we found elevated levels of p21 mRNA in MEF null LSK cells and reasoned that p21 may account for their enhanced quiescence. We generated p21 −/− Mef −/− mice, which are viable, born at normal mendelian frequency and appear grossly normal. However, cell cycle analysis of HSCs obtained from p21 −/− Mef −/− mice showed that the enhanced quiescence in MEF null HSCs did not depend on p21, indicating that other p53 target genes play an important role in maintaining stem cell quiescence. We therefore utilized transcript profiling (Microarray studies and quantitative PCR analysis) to identify potential p53-regulated genes that may be differentialy expressed in LSK cells isolated from wild type, p53−/−, Mef −/−, and p53−/− Mef −/− mice. By ChiP and luciferase reporter assays, we show for the first time that Gfi-1 and Necdin are direct transcriptional targets of p53 in HSCs and both Gfi-1 and Necdin regulate the enhanced quiescence exhibited in MEF null HSCs. Taken together, our work defines a novel role for p53 in the maintenance of HSC quiescence. Furthermore, HSC quiescence and self-renewal appear to be mediated by different p53 target genes during steady state hematopoiesis.

2021 ◽  
Vol 12 ◽  
Author(s):  
Itamar B. Dias ◽  
Hjalmar R. Bouma ◽  
Robert H. Henning

Tissue-resident stem cells may enter a dormant state, also known as quiescence, which allows them to withstand metabolic stress and unfavorable conditions. Similarly, hibernating mammals can also enter a state of dormancy used to evade hostile circumstances, such as food shortage and low ambient temperatures. In hibernation, the dormant state of the individual and its cells is commonly known as torpor, and is characterized by metabolic suppression in individual cells. Given that both conditions represent cell survival strategies, we here compare the molecular aspects of cellular quiescence, particularly of well-studied hematopoietic stem cells, and torpor at the cellular level. Critical processes of dormancy are reviewed, including the suppression of the cell cycle, changes in metabolic characteristics, and cellular mechanisms of dealing with damage. Key factors shared by hematopoietic stem cell quiescence and torpor include a reversible activation of factors inhibiting the cell cycle, a shift in metabolism from glucose to fatty acid oxidation, downregulation of mitochondrial activity, key changes in hypoxia-inducible factor one alpha (HIF-1α), mTOR, reversible protein phosphorylation and autophagy, and increased radiation resistance. This similarity is remarkable in view of the difference in cell populations, as stem cell quiescence regards proliferating cells, while torpor mainly involves terminally differentiated cells. A future perspective is provided how to advance our understanding of the crucial pathways that allow stem cells and hibernating animals to engage in their ‘great slumbers.’


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 572-572
Author(s):  
Hitoshi Takizawa ◽  
Chandra S Boddupalli ◽  
Roland R Regoes ◽  
Sebastian Bonhoeffer ◽  
Markus G Manz

Abstract Abstract 572 Life-long blood production is maintained by a small fraction of hematopoietic stem cells (HSCs). Steady-state HSC cycling kinetics have been evaluated by in vivo labeling assays with 5-bromo-2-deoxyuridine (BrdU) (Cheshier et. al., PNAS 1999; Kiel et al., Nature 2007), biotin (Nygren et. al., 2008) and histon 2B-green fluorescent protein (H2B-GFP) transgenic mouse models (Wilson et. al., 2008; Foudi et. al., 2009). While the former studies showed that all HSCs equally divide and likely contribute to blood formation (clonal maintenance), the latter suggested that some HSCs divide frequently and contribute to blood formation until cell death or full differentiation, while some HSCs are quiescent and then get activated to follow the same fate as frequently dividing ones (clonal succession). However, due to low resolution, none of the labeling techniques used were able to track single cell divisions. Furthermore, methods used might have direct influence on cycling activity of HSCs. Thus it remains to be determined a) if HSC divide continuously, sequentially or repetitively and contribute to steady-state hematopoiesis, b) what is a relationship between divisional history and repopulating ability, and c) how self-renewal and differentiation capacity of HSC is impacted by naturally-occurring severe hematopoietic challenges as infections. To address this directly, we set up a high resolution non-invasive in vivo HSC divisional tracking assay with CFSE (carboxyfluorescein diacetate succinimidyl ester). We here show that i.v. transfer of CFSE-labeled HSCs into non-conditioned congenic recipient mice allows evaluation of steady-state HSC cycling-dynamics as CFSE is equally distributed to daughter cells upon cellular division. Transfer of Lin-c-kit+Sca-1+ cells (LKS) into non-irradiated mice revealed non- and 1–7x divided LKS in recipient bone marrow over 20 weeks. To test in vivo limiting dilution and single cell HSC potential, non- or ≥5x divided cells were sorted based on divisional history from primary recipients at different weeks after transplantation, and transplanted into lethally irradiated secondary recipients. Single non-divided LKS at 3 weeks post primary transfer was able to multi-lineage repopulate 24% of recipients long-term, while 50 of ≥5x divided LKS did not engraft. Interestingly, both non- and ≥5x divided LKS at 7 or 12–14 weeks after primary transfer engrafted and showed fluctuating contribution to multi-lineage hematopoiesis over serial transplantation. Mathematical modeling based on limiting dilution transplantation, revealed no evidence for a dichotomy of biologically defined HSCs in different groups. Instead, steady-state serial transplantation with temporary fast-cycling cells revealed that they can slow down over time, suggesting dynamically changing cycling activity of HSC. We next tested the effects of hemato-immunological challenge on HSC proliferation. Mice transplanted with CFSE-labeled LKS cells were repetitively treated with LPS. Analysis 8 days after final LPS injection, i.e. three weeks after steady-state transplantation revealed that all LKS entered cell cycle and the number of ≥5x divided LKS was increased. Secondary transplantation showed that 2–4 time and ≥5x divided LKS from LPS-treated mice reconstituted multi-lineage hematopoiesis whereas both fractions from control mice failed to engraft. This data clearly indicate that HSCs are activated from quiescence upon LPS challenge and provide evidence, that naturally-occurring hemato-immunological challenges, such as gram-negative bacterial infection induces proliferation and self-renewal of HSCs. Our data suggest in contrast to previously proposed concepts, a novel “dynamic repetition” model for HSC cycling activity and blood formation where some HSCs participate in hematopoiesis for a while, subsequently enter a resting phase and get reactivated again to contribute to blood formation in repetitive cycles, leading to homogenous total divisional history of all HSCs at end of life. These findings might represent a biological principle that could hold true for other somatic stem cell-sustained organ-systems and might have developed during evolution to ensure equal distribution of work-load, efficient recruitment of stem cells during demand, and reduction of risk to acquire genetic alterations or fatal damage to the whole HSC population at any given time. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2464-2464
Author(s):  
Grant Anthony Challen ◽  
Margaret A Goodell

Abstract Over the decades since hematopoietic stem cells (HSCs) were first identified, the traditional view has been that the hematopoietic system is regenerated by a single pool of multipotent, quiescent HSCs that are sequentially recruited into cell cycle and which then progressively divide and differentiate until they are exhausted and ultimately replaced by the next cohort of stem cells. However, recent evidence has challenged this classical clonal succession model of HSC hierarchy by suggesting that the hematopoietic system is maintained by a pool of different HSC subtypes, with distinct self-renewal and differentiation potentials (the clonal diversity model, Figure 1). The side population (SP), characterized by Hoechst dye efflux, has been used as a method for isolating HSCs for over a decade and the SP has been shown to be highly enriched for HSC activity. While the entire SP is strikingly homogeneous with respect to expression of canonical stem cell markers such as Sca-1 and c-Kit, we recently observed heterogeneous expression for the SLAM family molecule CD150 within the SP, with CD150+ cells more prevalent in the lower SP and CD150− cell more prevalent in the upper SP. We decided to examine this observation further by investigating the properties of cells from different regions of the SP. Functional capacity was assessed by competitive bone marrow transplantation of upper SP cells, lower SP cells, and a combination of the two populations. Lower SP cells showed better engraftment than upper SP cells in recipient mice, a trend that continued when donor HSCs were isolated from primary recipients and re-transplanted into secondary hosts. Lower SP cells showed 3-fold better engraftment than upper SP cells in secondary transplants, suggesting better self-renewal capacity. However, analysis of the hematopoietic lineages formed by donor cells in recipient mice demonstrated that while both upper and lower SP cells were capable of forming all mature lineages, lower SP cells were biased towards myeloid differentiation while upper SP cells were biased towards lymphoid differentiation. The lineage biases observed from transplantation of one cell population alone were exacerbated when both upper and lower SP cells were co-transplanted into the same recipient mouse, suggesting that while both populations are capable of forming all hematopoietic lineages, in the presence of the other stem cell type (as would be the case in normal homeostasis) that the majority of the output from each HSC subtype is almost exclusively lymphoid or myeloid. The lineage contribution trends observed in the peripheral blood were also reproduced when bone marrow of transplanted mice was analyzed, including at the level of progenitors with lower SP cells showing greater ability to make myeloid progenitors (megakaryocyte-erythrocyte progenitors and granulocyte-macrophage progenitors) and upper SP cells producing proportionately more common lymphoid progenitors. Microarray analysis of upper and lower SP cells to determine the molecular signatures underlying these functional differences found many genes critical for long-term HSC self-renewal to be highly expressed in lower SP cells including Rb1, Meis1, Pbx1 and TGFbr2 while upper SP cells showed higher expression of cell cycle and activation genes. Cell cycle analysis showed upper SP cells to be approximately 2-fold more proliferative than lower SP cells (18.9% to 8.3% Ki-67+, 39.4% to 20.1% BrdU+ 3-days post-BrdU administration). The clonal diversity model which proposes the adult HSC compartment consists of a fixed number of different HSC subtypes each with pre-programmed behavior has important implications for using HSCs in experimental and clinical settings. While other studies have provided functional evidence for the clonal diversity model, this is the first study to prospectively isolate the functionally distinct HSC subtypes prior to transplantation. Figure Figure


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4199-4199 ◽  
Author(s):  
Carolina L. Bigarella ◽  
Pauline Rimmele ◽  
Rebeca Dieguez-Gonzalez ◽  
Raymond Liang ◽  
Brigitte Izac ◽  
...  

Abstract Leukemic stem cells (LSCs) share many of the same properties of normal hematopoietic stem cells (HSCs) including their highly quiescent state, capacity to self-renew, low levels of reactive oxygen species (ROS) and enhanced DNA repair program. These properties make the efficient and specific eradication of these cells challenging. Foxo3 and p53 are two transcription factors essential for the modulation of HSC quiescence and self-renewal. While Foxo3 is inhibited by signaling from several oncoproteins but crucial for the maintenance of the LSCs in both chronic and acute myeloid leukemia (CML and AML respectively), mutations of p53 although rare, are associated with poor prognosis in advanced stages of these diseases. In vivo ROS-mediated activation of p53 is known to lead to loss of quiescence, alterations of cell cycle and exhaustion of the Foxo3-/- HSC pool. Seeking to understand the contribution of p53 to Foxo3-/- HSC cycling defects, we crossed p53+/- and Foxo3+/- mice. To our surprise we found the bone marrow (BM) frequency of both p53+/-Foxo3-/- and p53-/-Foxo3-/- LSK (Lin-Sca1+cKit+) and long-term-HSC (LT-HSC, LSK Flk2-CD34-) populations greatly increased as compared to their Foxo3-/- counterparts (n=5 mice per genotype; p<0.05). Using Ki67 and DAPI staining we found that loss of one or both alleles of p53 gradually rescued the cell cycle defect of Foxo3-/- HSC and increased the frequency of LSK cells in Go by 2-fold. Loss of p53 also rescued the impaired capacity of Foxo3-/- LSK cells to competitively repopulate multilineage blood over 16 weeks, as shown by the higher frequency of p53+/-Foxo3-/- and p53-/-Foxo3-/- donor-derived cells in the peripheral blood of recipient animals (∼47% recipients of double-mutant cells versus 20% in Foxo3-/- recipients, n=5 per group). Furthermore, loss of p53 significantly improved the compromised self-renewal of Foxo3 mutant HSC in serial BM transplantations. In our quest to identify mechanisms whereby p53 depletion improves Foxo3-/- HSC function, we noticed that the DNA damage accumulated in Foxo3-/- HSC at the steady-state was remarkably ameliorated by removal of one or both alleles of p53 from Foxo3-/- HSCs, as measured by flow cytometry levels of phospho-H2AX (gamma-H2AX) and DNA breaks by comet assay (n=3, p<0.05). Unexpectedly, ROS levels were also significantly reduced by 30% in p53+/-Foxo3-/- in comparison to Foxo3-/- LSK cells, while ROS levels in p53+/- LSK cells were similar to that in WT cells. Consistent with these results, the expression of several anti-oxidant enzymes including Sod1, Sod2, Catalase, Gpx1, Sesn1 and Sesn2 (n≥2), was highly upregulated while a number of genes implicated in mitochondrial generation of ROS were significantly deregulated as a result of loss of one or both alleles of p53. These combined findings suggest that a switch from anti-oxidant to pro-oxidant activity of p53 contributes to Foxo3-/- HSC defects. Despite their apparent normal stem cell function, p53+/-Foxo3-/- HSC were highly altered in their gene expression profile. Interestingly, Gene Set Enrichment Analysis (GSEA) of the microarray analysis (Illumina bead chip mouse-Ref8) of WT, p53+/-, Foxo3-/-, and p53+/-Foxo3-/- LSK cells showed that a cluster of genes associated with fatty acid metabolism was highly enriched in p53+/-Foxo3-/- HSCs (ES=0.746; p<0.01). In addition, from 3976 genes exclusively deregulated in p53+/-Foxo3-/- LSK cells, 201 (out of 1051) overlapped with genes downregulated, while 9 (out of 14) overlapped with genes exclusively upregulated in a LSC-gene signature. To evaluate whether this pre-leukemic profile was associated with increased susceptibility to malignancy, we compared the potential and timeline of BCR-ABL-transformed p53+/-Foxo3-/- HSC as compared to controls in establishing CML in mice. We found a shorter time to the onset of the disease and decreased survival of the recipients of p53+/-Foxo3-/- transformed HSCs (n=4 per group, p<0.05) as compared to WT and Foxo3-/- controls. We propose that the p53+/-Foxo3-/- double-mutant HSCs are enriched for preleukemic stem cells based on their quiescence and self-renewal capacity, low ROS, robust DNA repair, susceptibility to transformation and aberrant gene expression profile. These findings raise the possibility that the coordinated Foxo3 and p53 regulation of ROS wires together the stem cell program. Disclosures: No relevant conflicts of interest to declare.


2008 ◽  
Vol 2 (3) ◽  
pp. 274-283 ◽  
Author(s):  
Heather E. Fleming ◽  
Viktor Janzen ◽  
Cristina Lo Celso ◽  
Jun Guo ◽  
Kathleen M. Leahy ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (25) ◽  
pp. 6562-6571 ◽  
Author(s):  
Michihiro Kobayashi ◽  
Edward F. Srour

Abstract The cell-cycle status of hematopoietic stem cells (HSCs) is tightly regulated, most likely to balance maintenance of stem-cell status through quiescence and expansion/differentiation of the hematopoietic system. Tumor-suppressor genes (TSGs), with their cell cycle–regulatory functions, play important roles in HSC regulation. The cyclin-D binding myb-like transcription factor 1 (Dmtf1) was recently recognized as a TSG involved in human cancers by repressing oncogenic Ras/Raf signaling. However, the role of Dmtf1 in the hematopoietic system is entirely unknown. In the present study, we demonstrate that Dmtf1 regulates HSC function under both steady-state and stress conditions. Dmtf1−/− mice showed increased blood cell counts in multiple parameters, and their progenitor cells had increased proliferation and accelerated cell-cycle progression. In addition, long-term HSCs from Dmtf1−/− mice had a higher self-renewal capacity that was clearly demonstrated in secondary recipients in serial transplantation studies. Dmtf1−/− BM cells showed hyper proliferation after 5-fluorouracil–induced myeloablation. Steady-state expression and Induction of CDKN1a (p21) and Arf were impaired in HSCs from Dmtf1−/− mice. The function of Dmtf1 was mediated by both Arf-dependent and Arf-independent pathways. Our results implicate Dmtf1 in the regulation of HSC function through novel cell cycle–regulatory mechanisms.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. SCI-19-SCI-19
Author(s):  
Toshio Suda

Abstract Hematopoietic stem cells (HSCs) play a key role in the lifelong maintenance of hematopoiesis through self-renewal and multi-lineage differentiation. Adult HSCs reside within a specialized microenvironment of the bone marrow (BM), called "niche", in which they are maintained in a quiescent state in cell cycle. Most of HSCs within BM show quiescence under the hypoxic niche. Since the loss of HSC quiescence leads to the exhaustion or aging of stem cells through excess cell division, the regulation of quiescence in HSCs is essential for hematopoietic homeostasis. On the other hand, cellular metabolism has been suggested to play a critical role in many biological processes including the regulation of stem cell properties and functions. However, the metabolic condition and adaptation of stem cells remain largely unaddressed. First, we have analyzed HSC metabolism using metabolomics approaches. With step-wise differentiation of stem cells, the cell metabolism associated with each differentiation stage may be different. A feature of quiescent HSCs is their low baseline energy production; quiescent HSCs rely on glycolysis and exhibit low mitochondrial membrane potential (ΔΨm). Likewise, HSCs with a low ΔΨm show higher reconstitution activity in BM hematopoiesis, compared to cells with high ΔΨm. By contrast, upon stress hematopoiesis, HSCs actively divide and proliferate. However, the underlying mechanism for the initiation of HSC division still remains unclear. In order to elucidate the mechanism underlying the transition of cell cycle state in HSCs, we analyzed the change of mitochondria activity in HSCs after BM suppression induced by 5-fluoruracil (5-FU). Upon 5-FU treatment, cycling progenitors are depleted and then quiescent HSCs start to divide. We found that HSCs initiate cell division after exhibiting enhanced ΔΨm, as a result of increased intracellular Ca2+ level. We hypothesize that extracellular adenosine, derived from hematopoietic progenitors, inhibits the calcium influx and mitochondrial metabolism. While further activation of Ca2+-mitochondria pathway led to loss of the stem cell function after cell division, the appropriate suppression of intracellular Ca2+ level by nifedipine, a blocker of L-type voltage-gated Ca2+ channels, prolonged cell division interval in HSCs, and simultaneously achieved both cell division and HSC maintenance (self-renewal division). Thus, our results indicate that the adenosine-Ca2+-mitochondria pathway induces HSC division critically to determine HSC cell fate. Next, to examine the mitochondria oxidative metabolism and purinergic pathways, we introduced the study on a tumor suppressor, Folliculin (FLCN). Conditional deletion of FLCN in HSC compartment using the Mx1-Cre or Vav-iCre system disrupted HSC quiescence and BM homeostasis dependently on the lysosomal stress response induced by TFE3. Together all, we propose that the change in cellular metabolism involving mitochondria is crucial for HSC homeostasis in the stress settings. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5409-5409 ◽  
Author(s):  
Mariana Tereza Lira Benício ◽  
Priscila S. Scheucher ◽  
Aglair Bergamo Garcia ◽  
Roberto P. Falcao ◽  
Eduardo M. Rego

Abstract The clinical significance of LSCs has recently been confirmed by the finding that stem cell-like gene expression signatures can predict the clinical outcome of acute myeloid leukemia (AML) patients, what suggests that the targeted elimination of LSCs would be an efficient therapeutic approach in AML. LSCs are rather infrequent in patients’ bone marrow, and hence are difficult to be identified and isolated. As an attempt to overcome such limitations, it has been previously shown that cell lines might be considered as attractive models aimed to better characterize LSC biological properties and their chemotherapy resistance mechanisms. Many attempts have been made to identify an immunophenotype that allow to discriminate between LSCs and normal hematopoietic stem cells (HSCs), but there is still controversy regarding the surface markers proposed so far. As a functional rather than immunological approach, the assessment of intracellular enzyme activities associated with the protection of primitive cells from oxidative insult during hematopoietic development has been proven as successful strategies, such as the use of Aldefluor reagent in the identification of AML LSCs. By taking advantage of the Aldefluor reagent in the field of stem cells research, we sought to identify LSC among the following AML cell lines, based on their high expression levels of the enzyme aldehyde dehydrogenase (ALDH): Kasumi-1, THP-1, MV-4;11, NB4 and OCI-AML3. Cells were stained with the Aldefluor reagent, following manufacturer’s specifications, and cell subsets presenting high ALDH activity were identified in Kasumi-1 (13%), THP-1 (25%) and OCI-AML3 (12%). To assess if the Aldefluor staining was correlated to the expression of genes involved in stem cell maintenance or myeloid commitment, these cell lines were further sorted in two subpopulations, according to ALDH activity (ALDHhigh and ALDHlow/int), and the respective expression levels of the following genes were evaluated by RQ-PCR: CEBPA, BMI-1, NOTCH-1, C-MYC, HOXA9, E2F1, NANOG and OCT3/4. The delta-delta Ct method was used to determine the fold change in target genes between ALDHhigh and ALDHlow/int subpopulations. CEBPA, BMI-1 and NOTCH-1 genes were upregulated (9, 2.3 and 2 fold, respectively) in Kasumi-1 ALDHhigh cells when compared to ALDHlow/int, whereas C-MYC and E2F1 were downregulated (0.3 and 0.5 fold, respectively). Little is known about the physiological role of CEBPA in adult HSC biology, though it has been described as a central determinant in the switch from fetal to adult HSCs. Bmi-1 has been shown to be essential for the generation of self-renewing adult HSCs. Despite the roles of Notch signaling on myeloid development and AML are poorly understood, it can promote self-renewal, and induce growth arrest and apoptosis in hematopoietic stem cells. Some molecular mechanisms responsible for self-renewal, like Bmi-1 and Notch signaling pathways, were found to be shared by both HSC and LSC, suggesting that a higher expression of both genes in ALDHhigh cells would be in accordance with stemness properties. In line with these results, lower levels of C-MYC and E2F1 expression in ALDHhigh cells would suggest that they are somehow more quiescent than their ALDHlow/int counterparts. No significant differences were observed on the target genes expression levels among the other cell lines studied. In conclusion, our results reveal an important correlation between ALDH activity and the expression of genes associated with stemness properties in identifying stem cells in Kasumi-1 cell line, suggesting that like KG-1 cell line, which is also CD34-enriched, Kasumi-1 would be considered as a valuable model to better understand AML stem cells properties. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1283-1283
Author(s):  
Aldona A Karaczyn ◽  
Edward Jachimowicz ◽  
Jaspreet S Kohli ◽  
Pradeep Sathyanarayana

Abstract Several recent studies have showed that dysregulation of microRNA (miRNA) expression in hematopoietic stem cells (HSC) can affect self-renewal of HSCs, and indicated a role for miRNAs in development of acute myeloid leukemia (AML). We and others have reported a significant down-regulation of miR-199b in AML patients. Recently we found that miR-199b is enriched in long-term hematopoietic stem cells (LT-HSC), suggesting that miR199-b may regulate HSCs function. Therefore, to understand the physiologic role of miR-199 in hematopoiesis during homeostasis, we evaluated various hematopoietic stem and progenitor cells (HSPC) populations in mice harboring genetic deletion of miR199b-5p using CRISPR/Cas method. We found that ablation of miR199b resulted in markedly increased frequencies of primitive HSC and MPPs, and analyses of distribution pattern in myeloid progenitor populations showed reduced numbers of common myeloid progenitors (CMPs) biased toward granulocyte-monocyte (GMPs) linage with no changes in megakaryocytic-erythroid progenitors (MEPs). The elevated numbers of HSC and MPPs may indicate that increased proportion of HSC population is actively cycling, thus we analyzed LSK populations for expression of proliferation marker Ki67 along with DNA staining. We found that miR-199b deletion reduces proportion of primitive HSC and MPPs in cell-cycle, which may affect HSC cell self-renewal. Futher cell-cycle analyses revealed that miR-199b null HSCs leave G0 faster to accumulate in G1, but rather do not progress into mitosis, which was recovered upon 5-fluorouracil-induced cytokine burst. These results indicate that loss of miR-199b increases cell cycle duration. To verify that the absence of miR-199b influences proliferation of HSCs we pulsed miR-199b KO and WT mice with BrdU for 16 hours. We found the difference in the cell cycle distribution between HSCs and progenitors, namely reduction of BrdU-positive HSC and MPPs and progression of GMP compartment. These results show that miR-199b deletion decreases HSC active cell cycle by prolonging cell cycle transition during steady-state hematopoiesis and promotes proliferation of myeloid cells. Because quiescent cells only become susceptible to 5-FU during hematopoietic stress, driving them into cycle, we injected 5-FU into miR-199 KO and WT mice once per week until hematopoietic failure occurred. We found that miR199-b KO mice died soon after two subsequent injections, most likely due to the faster HSC exhaust as compared to WT mice. These results show that loss of miR199b produces HSC with reduced quiescence and prolonged cell cycle, however upon stress these cells progress into cell cycle, making them more susceptible for 5-FU treatment. These results demonstrate that miR-199b intrinsically regulates active cycling of HSCs. CFU-S assays showed that miR-199b KO donors showed decreased colonies in spleen, suggesting that miR-199b deletion affects short-term repopulation. In long-term repopulation assay, we observed a significant reduction of HSCs compartment, but elevated numbers of MPPs in host mice transplanted with BM from miR-199 KO mice. This data indicates that loss of miR-199b causes defects in HSC self-renewal and alters HSCs reconstitution potential. To identify potential miR-199b targets in HSCs under steady-state hematopoiesis, we performed a gene profiling in SLAM-HSCs. mRNA levels of several putative miR-199b targets were markedly elevated in miR-199b KO HSCs. These genes are known to be involved in cell adhesion, cell cycle, transcription regulation and chromatin remodeling including Klf12, Tox3 and Cdk18. Our findings reveal a novel functional role for miR-199b in governing HSC maintenance. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document