scholarly journals Loss of Atg2b and Gskip impairs the maintenance of the hematopoietic stem cell pool size

Author(s):  
Shun-suke Sakai ◽  
Atsushi Hasegawa ◽  
Ryosuke Ishimura ◽  
Naoki Tamura ◽  
Shun Kageyama ◽  
...  

A germline copy number duplication of chromosome 14q32, which contains ATG2B and GSKIP , was identified in families with myeloproliferative neoplasm (MPN). Herein, we show that mice lacking both Atg2b and Gskip , but not either alone, exhibited decreased hematopoiesis, resulting in death in utero accompanied by anemia. In marked contrast to MPN patients with duplication of ATG2B and GSKIP , the number of hematopoietic stem cells (HSCs), in particular long-term HSCs, in double knockout fetal livers were significantly decreased due to increased cell death. Although the remaining HSCs still had the ability to differentiate into hematopoietic progenitor cells, the differentiation efficiency was quite low. Remarkably, mice with knockout of Atg2b or Gskip alone did not show any hematopoietic abnormality. Mechanistically, while loss of both genes had no effect on autophagy, it increased the expression of genes encoding enzymes involved in oxidative phosphorylation. Taken together, our results indicate that Atg2b and Gskip play a synergistic effect in maintaining the pool size of HSCs.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1166-1166 ◽  
Author(s):  
Zhenrui Li ◽  
Keiyo Takubo ◽  
Pengxu Qian ◽  
Toshio Suda ◽  
Linheng Li

Abstract Hematopoietic stem cells (HSCs) maintenance is required to preserve stem cell pool and compensate the dynamic loss of blood cells. Previous studies of HSCs maintenance mainly focus on the quiescent versus active state of HSCs and accumulated evidence indicates that metabolism plays a critical role in coordinating divergent stem cell states. While recent reports largely emphasized the role of catabolic glycolysis on long-term (LT) HSC maintenance, we found that free amino acids are enriched in primitive stem cell by ~1.5 fold. Given that amino acid metabolism in HSCs is largely unknown, we first cultured bone marrow (BM) cells with individual amino acid deprived medium to study the function of individual amino acids on HSCs in vitro. Surprisingly, we found that specific amino acids, including valine, methionine and threonine (VMT), are essential for maintaining primitive HSCs, as removing them (VMT) individually from media dramatically reduced primitive HSC number by over 95%. Thus, we hypothesize that specific amino acids are critical for preserving the stem cell pool and maintaining their function. To test it, we transplanted equal number of cells cultured with complete or individual VMT deprived media into lethally irradiated recipient mice and found VMT deprivation in vitro impaired stem cell repopulation ability. We also identified the amino acid transporter X (AATX) that is specifically expressed in HSCs and maintain VMT levels within the cell. Furthermore, inhibition of AATX reduced LT-HSC (LSK CD34- Flk2-) number in vivo. BM transplantation indicated that AATX inhibition impaired stem cell long-term reconstitution ability by over 2 fold. Our studies uncovered a role of amino acid metabolism in HSC maintenance and discovered the underlying molecular mechanism related to the amino acid transport. This finding may impact clinical treatment of blood disorders including leukemia. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4749-4749
Author(s):  
Shanti Rojas-Sutterlin ◽  
André Haman ◽  
Trang Hoang

Abstract Abstract 4749 Hematopoietic stem cell (HSC) transplantation is the first successful cellular therapy and remains the only treatment providing long-term cure in acute myeloblastic leukemia. At the apex of the hematopoietic system, quiescent HSCs are spared by chemotherapeutic treatments that target proliferating cells and therefore can regenerate the entire blood system of a patient after drug exposure. Nevertheless, the consequence of repeated chemotherapy regimen on HSC function remains to be clarified. We previously showed that Scl/Tal1 gene dosage regulates HSC quiescence and functions when transplanted at limiting dilutions (Lacombe et al., 2010). In the present study, we investigate how massive expansion in vivo influences stem cell functions. To address this question, we optimized a protocol based on 5-fluorouracil (5-FU), an antimetabolite that has been used to treat colon, rectum, and head and neck cancers. In addition, we used Scl+/− mice to address the role of Scl in controlling HSCs expansion post-5-FU. We show that within 7 days following 5-FU treatment, HSCs exit quiescence and enter the cell cycle. To deplete cycling HSCs, we injected a second dose of 5-FU and showed that the stem cell pool was disseminated. Nonetheless, the remaining HSCs proliferated extensively to re-establish the HSC pool, which was twice larger than that of untreated mice. At this point, most HSCs have exited the cell cycle and were back to quiescence. Despite a near normal stem cell pool size and a quiescent status, HSCs from these 5-FU treated mice could not compete against untreated cells to regenerate the host in transplantation assays. Furthermore, we show that this extensive proliferation in vivo severely impaired the clonal expansion of individual HSC as measured by the mean activity of stem cell (MAS). Our results demonstrate that HSCs lose their competitive potential after two 5-FU treatments, suggesting that HSCs have an intrinsic expansion limit beyond which their regenerative potential is impaired. In addition, Scl is haplodeficient for cell cycle entry and cell division but Scl gene dosage does not affect this expansion limit. Therefore, our data dissociate the control of HSC expansion under extensive proliferative stress from cell cycle control during steady state. We surmise that chemotherapy regimen based on repeated administration of 5-FU or other antimetabolites are likely to severely impair long-term stem cell functions. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 170-170
Author(s):  
Owen J Tamplin ◽  
Jonathan E Henninger ◽  
Hunter L Elliott ◽  
Douglas S Richardson ◽  
Leonard I. Zon

Abstract The entire blood system is supported throughout life by a small number of hematopoietic stem and progenitor cells (HSPCs) that are produced exclusively during embryonic development. These stem cells are generated from the hemogenic endothelium of the dorsal aorta, then migrate to the fetal liver where they expand, before making a final migration to the fetal bone marrow. After seeding the bone marrow, the stem cell pool stabilizes and the total number of HSPCs remains relatively constant. Very little is known about this early stage of bone marrow niche colonization. A better understanding of native stem cell pool establishment will likely lead to improved clinical HSPC transplantation that depends on repopulation of the bone marrow niche. Currently, imaging technology does not allow direct visualization of the bone marrow niche during colonization because it occurs in utero in the long bones of the fetus. The zebrafish is an advantageous model because the embryos develop externally and are transparent. To quantify the early stem cell pool, we employed long term fate mapping with clonal analysis using the multicolor Zebrabow system, which imparts a unique fluorescent hue to stem cells and their progeny. Our findings reveal that 21 HSPC clones exist prior to HSPC emergence (24 hours post fertilization) and 30 clones are present during peak production from the aorta (48 hours post fertilization). Seeding of the presumptive adult marrow niche in zebrafish begins 4-5 days post fertilization, versus 16.5 days in the mouse. We previously described a transgenic zebrafish line (Runx:mCherry) that marks long term repopulating HSPCs throughout development and into adulthood. HSPC-specific expression is driven by the well-characterized Runx1 +23 kb mouse enhancer element. We used this line to directly observe the earliest immigration events of HSPCs as they arrive in the marrow. To achieve this, we immobilized the zebrafish by injection of the snake venom protein alpha-bungarotoxin directly into circulation. This allowed long term live imaging of the niche (~16 hours) so we could quantify the dynamics of HSPC colonization and expansion. To rapidly acquire high resolution imaging data for this deep tissue we applied lightsheet microscopy. By simultaneously illuminating the sample in the X plane, while taking images in the Z plane, hundreds of optical sections can be captured in seconds. The high pixel and temporal resolution of lightsheet microscopy in a large volume of tissue provides a highly dynamic view of the entire marrow niche. We could assess the localization of HSPCs in relation to other cell types within the niche. For example, HSPCs were closely associated with endothelial cells in a perivascular niche, similar to what has been described in mammalian bone marrow. Furthermore, we could quantify single Runx+ nuclei over time on one side of the bilateral kidney marrow. During this early stage of niche colonization, we found the number of HSPCs per side was ~50 (so ~100 total) and that remained relatively constant. This was in fact a dynamic equilibrium achieved by ingress and egress of cells, as well as occasional cell divisions. This cell number was independently validated using another transgenic zebrafish line, cd41:GFP, that also marks HSPCs. This quantification, combined with our data from earlier development, suggests that HSPCs undergo around two population doublings between emergence from the aorta and engraftment in the marrow. This unique platform for the quantification of a total stem cell pool will allow further functional and mechanistic studies using both genetics and chemical biology. Our goal is to gain insights into the establishment of the stem cell pool within the niche microenvironment and how this could improve clinical transplantation outcomes. Disclosures Zon: Marauder Therapeutics: Equity Ownership, Other: Founder; Scholar Rock: Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Other: Founder; Fate, Inc.: Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Other: Founder.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 585-585
Author(s):  
Owen J. Tamplin ◽  
Ellen M. Durand ◽  
Logan A. Carr ◽  
Pulin Li ◽  
Leonard I. Zon

Abstract Hematopoietic stem cells (HSC) reside in the bone marrow niche and sustain the production of blood throughout life. The entire pool of these rare and important cells is generated during a brief window of embryonic development. HSC are produced by the hemogenic endothelium of the dorsal aorta, migrate to and expand in the fetal liver, and then migrate again to seed the bone marrow. The zebrafish is a highly conserved and well-established model for HSC development. Similar to mammals, HSC emerge from the dorsal aorta, but then colonize a vascular plexus in the tail of the embryo—the caudal hematopoietic tissue (CHT). It is difficult to directly observe the interactions between an endogenous HSC and its niche, so we have developed the CHT as a model for HSC-niche interactions. To track HSC in vivo we have generated a transgenic reporter using the previously described mouse Runx1 +23 kb intronic enhancer. The purity of the stem cell pool marked by this reporter was determined. Using adult-to-adult limiting dilution transplantation with as few as one Runx1+23 positive cell, we have estimated the HSC purity to be approximately 1/35 (without immune matching), or similar to Kit+Sca1+Lin- (KSL) in mouse. This is the most pure stem cell population defined in the zebrafish system. Using embryo-to-embryo transplantation, a technique that is unique to zebrafish, we sorted Runx1+23 positive cells from one group of embryos and transplanted them to another by injection directly into circulation. Embryos are then grown to adulthood and marrow is tested for long-term engraftment between 3 and 5 months. This transplantation technique precedes formation of the thymus, thereby removing any chance of immune rejection. Highly stringent dilution of HSC in our embryo-to-embryo transplants has estimated a stem cell purity of one in two cells. Next, we applied our highly specific reporter to visualize HSC migration to the CHT niche. After arrival of the HSC, we have described 5 distinct steps during colonization: 1) adherence; 2) extravasation; 3) abluminal migration; 4) endothelial niche formation (“cuddling”); and 5) cell fate decisions. Live imaging analysis of HSC together with endothelial and stromal transgenic reporters has allowed us to quantify the relationship between different cell types within the CHT. For example, we observe preferential localization of HSC in close proximity to cxcl12a positive stromal cells. Lastly, we have sought to identify the molecular mechanisms involved in interactions between HSC and their niche. A chemical genetic screen identified the natural product lycorine as a small molecule that increases hematopoiesis in the CHT and promotes HSC-endothelial cell interactions. Combined chemical treatment and live imaging revealed that lycorine significantly increased the residence time of HSC in the niche. To test if treatment during the window of CHT colonization (2-3 days post fertilization) had long-term effects on HSC and the stem cell pool, the compound was washed off at 3 days and the Runx1+23 positive population was quantified by FACS. At 7 days post fertilization, after colonization of the marrow, there was a sustained and significant increase in Runx1+23 positive HSC. Strikingly, after 3 months, when treated embryos were raised to adulthood, we discovered that the increased HSC-endothelial cell interactions we observed in the CHT niche had in fact had an impact on the number of HSC in the adult. Our studies establish that the Runx1+23 transgenic is a highly specific reporter of HSC both in the embryo and adult, and that we can use this reporter for in vivo observation of an endogenous HSC niche. Furthermore, we show that the size of the adult stem cell pool can be altered by a transient signal during development. Disclosures: Tamplin: Boston Children's Hospital: Patents & Royalties. Zon:FATE Therapeutics, Inc: Consultancy, Equity Ownership, Founder Other, Membership on an entity’s Board of Directors or advisory committees, Patents & Royalties; Stemgent, Inc: Consultancy, Membership on an entity’s Board of Directors or advisory committees, Stocks, Stocks Other; Scholar Rock: Consultancy, Equity Ownership, Founder, Founder Other, Membership on an entity’s Board of Directors or advisory committees, Patents & Royalties.


Blood ◽  
2003 ◽  
Vol 102 (10) ◽  
pp. 3541-3547 ◽  
Author(s):  
Cheryl D. Helgason ◽  
Jennifer Antonchuk ◽  
Caroline Bodner ◽  
R. Keith Humphries

AbstractSH2-containing inositol 5-phosphatase (SHIP) is an important negative regulator of cytokine and immune receptor signaling. SHIP-deficient mice have a number of hematopoietic perturbations, including enhanced cytokine responsiveness. Because cytokines play an important role in the maintenance/expansion of the primitive hematopoietic cell pool, we investigated the possibility that SHIP also regulates the properties of cells in these compartments. Primitive hematopoietic cells were evaluated in SHIP-deficient mice and wild-type littermate controls using the colony-forming unit-spleen (CFU-S) and competitive repopulating unit (CRU) assays for multipotent progenitors and long-term lympho-myeloid repopulating cells, respectively. Absence of SHIP was found to affect homeostasis of CFU-S and CRU compartments. Numbers of primitive cells were increased in extramedullary sites such as the spleen of SHIP-deficient mice, although total body numbers were not significantly changed. In vivo cell cycle status of the CRU compartment was further evaluated using 5-fluorouracil (5-FU). SHIP-deficient CRUs were more sensitive to 5-FU killing, indicating a higher proliferative cell fraction. More strikingly, SHIP was found to regulate the ability of primitive cells to regenerate in vivo, as CRU recovery was approximately 30-fold lower in mice that received transplants of SHIP-deficient cells compared with controls. These results support a major role for SHIP in modulating pathways important in homeostasis and regeneration of hematopoietic stem cells, and emphasize the importance of negative cytokine regulation at the earliest stages of hematopoiesis. (Blood. 2003;102:3541-3547)


2018 ◽  
Vol 2018 ◽  
pp. 1-4 ◽  
Author(s):  
Paola Villafuerte-Gutiérrez ◽  
Montserrat López Rubio ◽  
Pilar Herrera ◽  
Eva Arranz

Hematopoietic myeloproliferative neoplasms with FGFR1 rearrangement result in the 8p11 myeloproliferative syndrome that in the current Word Health Organization classification is designated as “myeloid and lymphoid neoplasm with FGFR1 abnormalities.” We report the case of a 66-year-old man who had clinical features that resembled chronic myeloid leukaemia (CML), but bone marrow cytogenetic and fluorescent in situ hybridization (FISH) studies showed t(8;22)(p11;q11) and BCR-FGFR1 fusion gene. He was initially managed with hydroxyurea, and given the aggressive nature of this disease, four months later, the patient underwent an allogeneic hematopoietic stem-cell transplantation (HSCT) from an HLA-haploidentical relative. Currently, HSCT may be the only therapeutic option for long-term survival at least until more efficacious tyrosine kinase inhibitors (TKIs) become available.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5014-5014
Author(s):  
Irina N. Shipounova ◽  
Nataliya A. Petinati ◽  
Nina J. Drize ◽  
Aminat A. Magomedova ◽  
Ekaterina A. Fastova ◽  
...  

Introduction. Stromal microenvironment of the bone marrow (BM) is essential for normal hematopoiesis; the very same cells are involved in the interaction with the leukemic stem cells. The aim of the study was to reveal the alterations in stromal microenvironment of patients in debut and after the therapy using multipotent mesenchymal stromal cells (MSC) as a model. Methods. MSC of patients with acute myeloid leukemia (AML, N=32), acute lymphoblastic leukemia (ALL, N=20), chronic myeloid leukemia (CML, N=19), and diffuse large B-cell lymphoma without BM involvement (DLBCL, N=17) were isolated by standard method from the patients' BM. Each BM sample was acquired during diagnostic aspiration after the informed signed consent was obtained from the patient. Groups of BM donors comparable by age and gender were used as controls for each nosology. Gene expression was analyzed with real-time RT-PCR. The significance of differences was evaluated with Mann-Whitney U-test. Results. The results of gene expression analysis are summarized in Table. The expression of genes regulating hematopoietic stem and precursor cells (JAG1, LIF, IL6) was significantly upregulated in MSC of the patients in debut, except for DLBCL. The latter was characterized with upregulation of osteogenic marker gene SPP1 and downregulation of FGFR1 gene. The upregulation of the expression of genes regulating proliferation of stromal cells (PDGFRA, FGFR1) and adipogenic marker gene (PPARG) was common for AML and CML. Both acute leukemias were characterized by the upregulation of genes associated with inflammation and regulation of hematopoietic precursors (CSF1, IL1B, IL1BR1) and by the downregulation of chondrogenic differentiation marker gene (SOX9). CML and DLBCL demonstrated the upregulation of FGFR2. BM of the DLBCL patients did not contain any malignant cells; nevertheless, stromal precursors from the BM were significantly affected. This indicates the distant effects of DLBCL malignant cells on the patients' BM. Myeloid malignancies seem to affect MSC more profoundly then lymphoid ones. Effect of leukemic cells on stromal microenvironment in case of myeloid leukemia was more pronounced. The treatment significantly affected gene expression in MSC of patients. In all studied nosologies the IL6 gene expression was upregulated, which may reflect the inflammation processes ongoing in the organism. The expression of LIF was upregulated and ICAM1, downregulated in MSCs of AML, ALL, and CML patients. In the MSC of patients with AML, who had received the highest doses of cytostatic drugs to achieve remission, a significant decrease in the expression of most studied genes was found. In patients with ALL with long-term continuing treatment in combination with lower doses of drugs, IL1B expression was increased, while the decrease in expression was detected for a number of genes regulating hematopoietic stem cells (SDF1, TGFB1), differentiation and proliferation (SOX9, FGFR1, FGFR2). Treatment of CML patients is based on tyrosine kinase inhibitors in doses designed for long-term use, and is less damaging for MSC. The upregulation of TGFB1, SOX9, PDGFRA genes and downregulation of IL1B gene was revealed. MCS of DLBCL patients, unlike the other samples, were analyzed after the end of treatment. Nevertheless, significant upregulation of IL8 and FGFR2 genes was found. Thus, both the malignant cells and chemotherapy affect stromal precursor cells. The changes are not transient; they are preserved for a few months at least. MSCs comprise only a minor subpopulation in the BM in vivo. When expanded in vitro, they demonstrate significant changes between groups of patients and healthy donors. Conclusions. Leukemia cells adapt the stromal microenvironment. With different leukemia, the same changes are observed in the expression of genes in MSC. MSC of patients with acute forms have a lot of changes which coincide among these two diseases. MSC of AML patients are most affected both in debut and after the therapy. Treatment depends on the nosology and in varying degrees changes the MSC. This work was supported by the Russian Foundation for Basic Research, project no. 17-00-00170. Disclosures Chelysheva: Novartis: Consultancy, Honoraria; Fusion Pharma: Consultancy. Shukhov:Novartis: Consultancy; Pfizer: Consultancy. Turkina:Bristol Myers Squibb: Consultancy; Novartis: Consultancy, Speakers Bureau; Pfizer: Consultancy; Novartis: Consultancy, Speakers Bureau; fusion pharma: Consultancy.


Blood ◽  
2013 ◽  
Vol 121 (25) ◽  
pp. 5015-5024 ◽  
Author(s):  
Sarah J. Stein ◽  
Albert S. Baldwin

Key Points p65 is an important factor in hematopoiesis through the regulation of hematopoietic stem cell function and lineage commitment. p65 controls the expression of genes encoding key factors that promote hematopoietic stem cell homeostasis.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1394-1394
Author(s):  
Sachiko Ezoe ◽  
Itaru Matsumura ◽  
Hirokazu Tanaka ◽  
Yusuke Satoh ◽  
Takafumi Yokota ◽  
...  

Abstract Sir2 (silent information regulator 2) is a member of a gene family (sirtuins) encoding NAD(+)-dependent histone deacetylases, which leads to increased DNA stability and prolonged lifespan in Saccharomyces cerevisiae and Caenorhabditis elegans. In mammalians, SIRT1 has also been found to function as a deacetylase for numerous protein targets involved in various cellular pathways, including stress responses, apoptosis, and neural axonal degeneration. However, the effects of SIRT1 on hematopoiesis remains unknown. We previously reported that the SIRT1 inhibitor, nicotinamide(NA), promoted the differentiation of murine hematopoietic stem/progenitor cells, and its activator, Resveratrol, suppressed the differntiation. In this report, we analysed the differentiation of stem/progenitor cells derived from SIRT1 KO mice. Because SIRT1 KO mice can survive less than a week after birth, we performed experiments using c-Kit(+)Lineage(−)Sca-1(+) cells (KSLs) derived from KO mice fetal liver. At first, we cultured KSLs with the cytokine cocktail containing SCF, IL-6, Flt3L, and TPO, which is utilized for the expansion of stem cells. After five day culture, we estimated the population which remains in KSL fraction. As a result, percentage of KSLs from KO fetal liver was less than 5%, while that from WT was about 15%. We also examined the colony formation of KO and WT fetal liver KSL cells using replating assays. At the first plating, total number of colonies developed from KO fetal liver KSLs was smaller than that from WT by 30–40%, and at the third plating, there could be detected no colonies from KO, while 20–30 colonies were observed from WT. Furthermore, we performed serial transplantation assays using WT and KO fetal liver KSLs. Although after primary transplant, we detected no significant difference in repopulation from KO KSLs compared to WT controls, three weeks after secondary transplant, % chimerism from KO KSLs was reduced to 1/2 compared with that from WT KSLs. These results suggested that Sirt1 suppresses the differentiation and promotes self-renew of hematopoietic stem/progenitor cells. To dissect the roles of target molecules of Sirt1 in suppression of differentiation, we first examined the mRNA expressions of some cell cycle-relating molecules in KO and WT fetal liver KSLs. As consequence, p16Ink4A and p19Arf were detected only in KO KSLs. Then we analyzed the roles of molecules which may effect those expressions. First, we examined the effects of MAPkinases inhibitors on the differentiation of KO and WT fetal liver KSLs. During the culture with SCF, IL-6, Flt3L, and TPO, the addition of p38 inhibitor(SB202190), or MEK1 inhibitor(PD98059), or JNK inhibitor did not change the effects of the SIRT1 targeting. Then it was suggested that MAPkinase pathways have little relation with the SIRT1-induced suppression of differentiation. Next we examined the role of p53, which was reported to combine with SIRT1 and to be deacetylated and repressed by SIRT1. KO and WT fetal liver KSLs were cultured with p53 inhibitor (pifithrin?), which partially cancelled the promotion of differntiation in SIRT1 KO KSLs. This result suggested that SIRT1 might inhibit differentiation of KSLs partially by antagonizing p53 activity. Next we examined the role of Foxo3a, a downstream molecule of SIRT1. Enforced expression of constitutive active form of Foxo3a(FKHRL1TM) also cancelled the promotion of differentiation in SIRT1 KO KSLs. As conclusion, we demonstrate that SIRT1 suppresses the differentiation of hematopoietic stem/progenitor cells by antagonizing p53 and enhancing Foxo3a activities, and contributes to maintenance of stem cell properties and stem cell pool.


Sign in / Sign up

Export Citation Format

Share Document