Gene expression profile in bone marrow and hematopoietic stem cells in mice exposed to inhaled benzene

Author(s):  
Brenda Faiola ◽  
Elizabeth S Fuller ◽  
Victoria A Wong ◽  
Leslie Recio
Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4694-4694
Author(s):  
Jana Michalova ◽  
Ludek Sefc ◽  
Filipp Savvulidi ◽  
Katarina Forgacova ◽  
Katerina Faltusova ◽  
...  

Abstract Abstract 4694 Background: Quiescent hematopoietic stem cells (HSCs) located in stem cell niches are characterized by a relative resistance to hypoxia. This study is focused primarily on maintainance of the repopulating ability of HSCs in structurally intact BM exposed to anoxia, lack of metabolic substrates and accumulation of metabolic waste products during a period of ischemia at three different temperatures. In the case of a warm ischemia at 37°C, changes in gene expression profile in the whole bone marrow has been also examined. Methods: Murine congenic model C57Bl/6 Ly5.1/Ly5.2 was used in the experiments. Normal mice or mice recovering from a bone marrow damage induced either by cyclophosphamide or a sublethal irradiation were sacrified. Their BM was maintained in intact femurs at 37°C for different time periods up to 6 hours. For normal bone marrow, exposure to ischemia at 20°C and 4°C was also used for up to 20 and 48 hours, respectively. Afterwards, bone marrow cells were harvested and cells corresponding to a half of the femur were transplanted to sublethally (6 Gy) irradiated recipients in a competitive repopulation assay. Resulting chimerism was examined up to 6 months after transplantation to test for STRCs and LTRCs (Short and Long Term Repopulating Cells). Subpopulations of erythropoietic (Ter119+), B-lymphopoietic (B220+), granulo- and monocytopoietic (Gr-1/Mac+), and LSK (Lin-Sca-1+c-Kit+) bone marrow cells were analyzed for dead cells and apoptosis. Total RNA was isolated from bone marrow exposed to warm ischemia ranging 0 to 4 hours and dynamics of changes in its gene expression profile was determined by Illumina MouseRef8 BeadChip. Results: Repopulating ability of ischemic BM was fully preserved for 2 hour of the warm (37°C) ischemia and for 6 hours and 8 hours of 20°C and 4°C ischemia, respectively. There was no difference between STRCs and LTRCs in survival. STRCs and LTRCs from the bone marrow collected 2 days or 5 days after a single dose of cyclophosphamide exposed to warm ischemia showed decreased repopulating ability in comparison with those of normal mice. STRCs significantly prevailed over LTRCs in bone marrow collected 20 days after a sublethal irradiation and showed increased sensitivity to warm ischemia. B220+ cells were the most sensitive cells of the bone marrow to warm ischemia, LSK and Ter119 cells being the most resistant ones. Gene expression profile in bone marrow exposed to warm ischemia changed progressively over time. Despite the highly unfavorable metabolic conditions, hypoxia and lack of energy, a set of overexpressed genes equaled in number the one inhibited. Conclusions: HSCs exposed to warm or cold ischemia maintain their repopulating ability for a considerable time. Bone marrow ischemia activates specific gene expression in paralel with supression of others. Supported by projects LC06044, MSM 0021620806 and the grant SVV-2010-254260507. Disclosures: No relevant conflicts of interest to declare.


2010 ◽  
Vol 10 (1) ◽  
pp. 12 ◽  
Author(s):  
Leilei Tang ◽  
Saskia M Bergevoet ◽  
Christian Gilissen ◽  
Theo de Witte ◽  
Joop H Jansen ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3433-3433
Author(s):  
Nan Xiao ◽  
Kaushal Jani ◽  
Jonathan L Jesneck ◽  
Glen D Raffel

Abstract Abstract 3433 With age, hematopoietic stem cells (HSCs) have numerical expansion, skewing towards myeloid development, loss of lymphoid potential, an underlying pro-inflammatory state and loss of self-renewal potential thus severely limiting responses to hematopoietic stress, ultimately leading to bone marrow failure. The mechanisms and pathways responsible for these changes in aged HSCs are incompletely understood. Using a conditional allele of Ott1, a gene originally isolated as the 5' fusion partner in t(1;22) acute megakaryocytic leukemia, we previously found a global regulatory role for the gene in hematopoiesis. Deletion of Ott1 in adult mice utilizing Mx1-cre recapitulated certain aspects of aging hematopoiesis including increased Lin−Sca1+c-Kit+ (LSK) population, myeloid expansion and decreased lymphopoiesis. The LSK compartment was further characterized using SLAM and CD34/Flk2 markers and demonstrated normal levels of LT-HSCs and increased ST-HSCs. Despite sufficient LT-HSC numbers, Ott1-deleted bone marrow was unable to competitively or non-competitively repopulate irradiated recipients. To exclude a homing or engraftment effect, Ott1flox/null Mx1-cre bone marrow was transplanted with competitor then excised post-engraftment. The rapid loss of the Ott1-deficient graft demonstrated Ott1 is required for maintenance under competitive stress. In contrast, primary mice undergoing Ott1 excision lived a normal lifespan and were able to maintain sufficient hematopoiesis although with a partial reduction in bone marrow clonagenicity showing loss of Ott1 is not limiting under steady state conditions. To test the HSC requirement for Ott1 under replicative stress, Ott1 knockout mice were challenged with 5-fluorouracil (5-FU). Ott1-deleted mice treated with 5-FU displayed delayed peripheral blood neutrophil recovery and showed accelerated bone marrow failure. Cell cycle analysis of steady state Ott1 knockout HSCs showed a similar profile to wild type controls, however, after 5-FU treatment, the G0 fraction was dramatically reduced. The G0 fraction is associated with the quiescent, self-renewing HSC population, therefore, Ott1 is required for maintaining HSC quiescence during replicative stress but not steady state hematopoiesis. To more specifically assess whether the functional hematopoietic changes seen after loss of Ott1 were accompanied by alterations in known aging-associated pathways, Gene Set Enrichment Analysis comparing Ott1-deleted HSCs in steady state to aged HSCs was performed and showed a highly enriched gene expression signature (NES 2.02 p<0.0001). Physiologic sequelae of HSC aging were observed after Ott1 excision including activation of NFκβ, elevation of reactive oxygen species (ROS), increase in DNA damage (γH2A.X levels) and activation of p38Mapk. Although ROS was elevated under steady state conditions, neither apoptosis, senescence or proliferation was significantly different from wild type control HSCs. Furthermore, anti-oxidant treatment with N-acetyl-cysteine was unable to rescue the HSC maintenance defect of the Ott1 knockout, signifying additional requirements in HSCs for Ott1 beyond regulation of ROS. An observed increase of mitochondrial mass in Ott1-deleted HSCs suggests an upstream function for Ott1 in metabolic control, potentially contributing to ROS generation or degradation. In summary, we have demonstrated an essential role for Ott1 in maintaining HSC quiescence during replicative stress and shown loss of Ott1 leads to the acquisition of key gene expression patterns and pathophysiologic changes associated with aging. These data suggest Ott1 functions in part to oppose specific consequences of aging in the hematopoietic compartment. Ott1 and Ott1-dependent pathways therefore represent a potential therapeutic target to prevent the morbidity and mortality arising from age-related defects in hematopoiesis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 603-603 ◽  
Author(s):  
Masayuki Yamashita ◽  
Eriko Nitta ◽  
Toshio Suda

Abstract Accumulation of DNA damage in hematopoietic stem cells (HSCs) is associated with aging, bone marrow failure and development of hematological malignancies. Mutation accumulation in HSCs precedes the development of leukemia and lymphoma, and these “pre-leukemic HSCs” can survive after chemotherapy, contributing to the relapse of the disease. Thus, understanding for the DNA damage response at a HSC level is a matter of critical importance for lifelong hematopoiesis, yet the protection mechanism for HSCs from DNA damage accumulation remains to be elucidated. During our study on the response of HSCs to ionizing radiation (IR), we have detected higher responsiveness of HSCs to DNA damage compared with committed progenitor cells: higher p53 activation was observed in HSC-enriched LSK (Lin-Sca1+cKit+) cells and LT-HSCs (CD150+CD41-CD48-LSK) than in myeloid progenitor-enriched LKS- cells. Of note, when treated with 4 Gy IR, LSK cells exhibited stronger upregulation of pro-apoptotic genes Bax, Noxa and Puma compared with LKS- cells, whereas upregulation of survival-contributing p21 and Mdm2 genes was comparable between the two populations. Corresponding to such characteristic behavior, we have identified apoptosis-stimulating protein of p53 1 (Aspp1) as a novel specific regulator of HSCs that provides HSCs with high sensitivity to apoptosis. We found that mRNA and protein of Aspp1 were specifically detected in LSK cells and LT-HSCs. To uncover the roles of Aspp1 in the regulation of HSCs, we evaluated HSCs of adult Aspp1 knockout (KO) mice. These mutant mice exhibited a major increase in the absolute number of LSK cells (1.5-fold; P<0.05) and LT-HSCs (2-fold; P<0.0005). Furthermore, self-renewal capacity of Aspp1-null HSCs was significantly enhanced as measured by serial competitive bone marrow (BM) transplantation assays (P<0.01). To assess the cause of enhanced self-renewal of Aspp1-null HSCs, we examined gene expression profile of Aspp1-null LSK cells before and after BM transplantation using multiplex quantitative RT-PCR array. Aspp1-null LSK cells showed higher expression of multiple quiescence-related genes including Tek, Mpl and Ndn. In line with this, Ki67 staining revealed that Aspp1-null LSK cells showed resistance to the loss of quiescence after serial BM transplantation (P<0.01), and Aspp1 KO mice showed accelerated recovery of peripheral blood and BM when treated with a single dose of 5-FU (P<0.05). Moreover, when serially transplanted or subjected to 4 Gy IR in vivo, Aspp1-null LSK cells exhibited higher resistance to apoptosis which was detected as decreased proportion of Annexin V-positive cells (P<0.05). Gene expression analysis consistently revealed that the induction of pro-apoptotic genes Bax, Noxa and Puma was impaired in irradiated Aspp1-null LSK cells. As a result of the reduced apoptosis, Aspp1-null LSK cells exhibited the tendency to retain persistent DNA damage after genotoxic stress as assessed by γH2AX and 53BP1 foci (chi-square test, P<0.05). Importantly, by breeding Aspp1 KO mice with Mx1-Cre mice and p53flox/flox mice, we verified that Aspp1 synergized with p53 to regulate self-renewal and genomic integrity of HSCs beyond its canonical p53-dependent function. Aspp1 loss further enhanced self-renewal capacity of HSCs in a p53-null background when assayed by serial BM transplantation (P<0.05). Likewise, Aspp1 deficiency further accentuated the accumulation of DNA damage after IR exposure in the absence of p53 (P<0.05). Consequently, whereas approximately half of the recipients receiving p53-null LSK cells died of thymic lymphoma, the recipient mice transplanted with LSK cells deficient for both Aspp1 and p53 were 100% lethal within 6 months after BM transplantation (log-rank test, P<0.01). These mice succumbed to hematological malignancies, mostly T-cell acute lymphoblastic lymphoma and leukemia (ALL) (88%) but also B-cell (6%) and myeloid (6%) malignancies. Taken together, our study demonstrates that Aspp1 attenuates HSC quiescence and induces apoptosis in damaged HSCs, in both p53-dependent and -independent manners, thereby inhibiting the development of leukemia and lymphoma in conjunction with p53 in HSCs. As loss of Aspp1 expression due to aberrant methylation of its promoter has already been proven to be an independent poor prognosis factor in ALL patients, Aspp1 may be a potential target for stem cell-directed therapy of leukemia and lymphoma. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 113 (8) ◽  
pp. 1661-1669 ◽  
Author(s):  
Ivan Maillard ◽  
Ya-Xiong Chen ◽  
Ann Friedman ◽  
Yuqing Yang ◽  
Anthony T. Tubbs ◽  
...  

Abstract Men1 is a tumor suppressor gene mutated in endocrine neoplasms. Besides its endocrine role, the Men1 gene product menin interacts with the mixed lineage leukemia (MLL) protein, a histone H3 lysine 4 methyltransferase. Although menin and MLL fusion proteins cooperate to activate Homeobox (Hox) gene expression during transformation, little is known about the normal hematopoietic functions of menin. Here, we studied hematopoiesis after Men1 ablation. Menin loss modestly impaired blood neutrophil, lymphocyte, and platelet counts. Without hematopoietic stress, multilineage and myelo-erythroid bone marrow progenitor numbers were preserved, while B lymphoid progenitors were decreased. In contrast, competitive transplantation revealed a marked functional defect of long-term hematopoietic stem cells (HSC) in the absence of menin, despite normal initial homing of progenitors to the bone marrow. HoxA9 gene expression was only modestly decreased in menin-deficient HSCs. These observations reveal a novel and essential role for menin in HSC homeostasis that was most apparent during situations of hematopoietic recovery, suggesting that menin regulates molecular pathways that are essential during the adaptive HSC response to stress.


2011 ◽  
Vol 41 (4) ◽  
pp. 192 ◽  
Author(s):  
Su-Hwan Kim ◽  
Young-Sung Kim ◽  
Su-Yeon Lee ◽  
Kyoung-Hwa Kim ◽  
Yong-Moo Lee ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document