Differential Response of Fanconi Anemia Hematopoietic Stem and Progenitor Cells to Oxidative and Oncogenic Stresses

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3593-3593
Author(s):  
Wei Du ◽  
Surya Amarachintha ◽  
Erden Ozlem ◽  
Qishen Pang

Abstract Members of the Fanconi anemia (FA) protein family are involved in DNA damage response. A common damage to DNA in vivo is oxidative stress, and compelling evidence suggests that FA cells are in an in vivo pro-oxidant state. In response to oncogenic activation, normal cells induce genetically encoding programs that prevent deregulated proliferation and thus protect multicellular organisms from cancer progression. How FA cells respond to oxidative DNA damage and oncogenic stress is largely unknown. By employing an in vivo stress-response model expressing the Gadd45b-luciferase transgene, we show here that hematopoietic stem and progenitor cells (HSPCs) from mice deficient for the FA gene Fanca or Fancc differentially responded to oxidative and oncogenic stresses. Compared to wild-type controls, Fanca-/- or Fancc-/- HSPCs exhibited a persistent response to oxidative stress. Mechanistically, we demonstrated that accumulation of unrepaired DNA damage, particularly in oxidative damage-sensitive genes, was responsible for the long-lasting response in FA HSPCs. In contrast, using two inducible models of oncogenic activation (LSL-K-rasG12D and MycER), we identify a short-lived response of FA HSPCs to oncogenic insults both in vitro and in vivo. Mechanistic studies revealed that loss of Fanca or Fancc impaired oncogenic stress-induced senescence (OIS), and genetic correction of Fanca or Fancc deficiency restored OIS in HSPCs. Furthermore, FA deficiency compromised K-rasG12D-induced arginine methylation of p53 mediated by the protein arginine methyltransferase 5 (PRMT5). Finally, forced expression of PRMT5 in HSPCs from LSL-K-rasG12D/CreER-Fanca-/- mice prolonged oncogenic response and delayed leukemia development in recipient mice. Taken together, our study demonstrates differential responses of HSPCs to oxidative and oncogenic stresses and identifies the FA pathway as an integral part of this versatile cellular mechanism. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 34-35
Author(s):  
Fengjiao Wang ◽  
Jiahuan He ◽  
Yanni Ma ◽  
Sha Hao ◽  
Siqi Liu ◽  
...  

RNA editing, adenosine (A)-to-inosine (I), plays a vital role in many biological processes. Our previous study has demonstrated that the hematopoietic stem and progenitor cells (HSPCs) deficient in adenosine deaminase acting on RNA 1 (Adar1), an RNA-editing enzyme, cannot reconstitute the irradiated recipients in vivo and form colonies in vitro (Xufeng R et al, PNAS 2009). However, the overall profile of RNA editome in hematopoiesis has not been established and the underlying mechanism how RNA editing governs the function of HSPCs is poorly defined. In this study, we sorted 12 murine adult hematopoietic cell populations and performed RNA sequencing. We depicted the landscape of RNA editome in hematopoietic cells and identified 30,796 editing sites in total. The dynamic landscape of RNA editome comprised of stage/group-specific as well as house-keeping editing patterns. Notably, antizyme inhibitor 1 (Azin1) was uncovered to be highly edited in HSPCs. To understand whether edited Azin1 was required for functioning of HSPCs, we transduced c-Kit+ HSPCs with the lentivirus carrying Azin1 cDNAs with distinct editing frequencies. c-Kit+ cells transduced with fully edited Azin1 showed enhanced reconstitution, compared to that transduced with partially edited or non-edited Azin1. Specifically, inability of RNA editing in Azin1 blocked the differentiation of hematopoietic stem cells (HSCs) in vivo. Moreover, a similar finding was obtained when Azin1 was knocked down. In conclusion, RNA editing of Azin1 (i) results in amino acid change to induce AZIN1 translocation to the nucleus, (ii) enhances AZIN1 binding affinity for DEAD box polypeptide 1 (DDX1) to alter the DDX1 chromatin distribution, and (iii) changes the expression of multiple hematopoietic regulators to ultimately promote HSPC differentiation. This work provides a valuable resource for studying RNA editing and delineates an essential role of Azin1 RNA editing in HSPCs. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3629-3629
Author(s):  
Yan Li ◽  
Shi Chen ◽  
Yongzheng He ◽  
Xiaohong Li ◽  
Fengchun Yang

Abstract Abstract 3629 Poster Board III-565 Fanconi anemia (FA) is a heterogeneous genetic disorder characterized by progressive bone marrow failure (BMF) and acquisition of malignancies. The only cure for BMF is a human leukocyte antigen (HLA)-matched BM transplantation from a family member or autologous stem cells before BMF develops. Therefore, mobilization of hematopoietic stem/progenitor cells (HSPCs) from BM into peripheral blood (PB) for collection has been a prerequisite for the therapy. However, patients with FA show a markedly decreased HSPC mobilization in response to the traditional mobilizing drug G-CSF and the mechanism(s) underlying the defect remains unknown. Mesenchymal stem/progenitor cells (MSPCs) have been known to be the common progenitor of a variety of cellular components in the bone marrow microenvironment. MSPCs express/secrete cytokines, extracellular matrix proteins and cell adhesion molecules, which regulate the homing, migration, proliferation and survival of HSPCs in vitro and in vivo. Recently, we reported that Fancg-/- MSPCs have a defect in hematopoietic supportive activity both in vitro and in vivo (Li et al. Blood, 2009). In the current studies, we show that Fancg-/- MSPCs have significant reduction in HSPC recruitment as compared to WT MSPCs in a transwell assay. Furthermore, Fancg-/- MSPCs have an alteration in the production of multiple cytokines/chemokines. Application of a neutralizing antibody to the cytokine blocked WT MSPC mediated HSPC migration in vitro. Furthermore, administration of the specific cytokine significantly increased HSPC mobilization in the Fancg-/- mice in vivo. These results demonstrated that an impaired BM microenvironment, specifically MSPCs in Fancg-/- mice, is contributory to defective HSPC mobilization. This study provides evidence of alternative clinical therapeutics for the mobilization of HSPCs in FA patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 864-864 ◽  
Author(s):  
Fukun Guo ◽  
Jie Li ◽  
Wei Du ◽  
Shuangmin Zhang ◽  
Wei Liu ◽  
...  

Abstract Abstract 864 The mammalian target of rapamycin (mTOR) integrates signals from nutrients, growth factors, and cellular energy status to control protein synthesis, cell growth, proliferation, survival and metabolism in various cancer cells, but its physiological function in the hematopoiesis process and signaling role in hematopoietic stem cell (HSC) regulation remain unknown. By using the inhibitor rapamycin, mTOR has previously been suggested to regulate megakaryocyte and dendritic cell proliferation and differentiation. Hyperactivation of mTOR by deletion of the negative regulators of mTOR, TSC1/TSC2 or PTEN, causes a loss of quiescence and long-term exhaustion of HSCs. Since conventional gene targeting of mTOR leads to early embryonic lethality, a conditional mTOR knockout mouse model has recently been generated. We have produced mTORflox/flox; Mx-Cre compound mice that allow interferon-induced mTOR deletion in bone marrow (BM) following a transplantation and polyI:C induction protocol. We found that depletion of mTOR drastically affected hematopoiesis: the mTORflox/flox;Mx-Cre BM recipient mice showed a marked reduction in total BM cellularity and in the numbers and frequency of myeloid and lymphoid cells, erythrocytes, and platelets in peripheral blood, bone marrow, and thymus, after induced mTOR deletion, resulting in bone marrow failure and lethality. Interestingly, the numbers of hematopoietic stem and progenitor cells (HSPCs; Lin−Sca-1+c-Kit+) and HSCs (CD150+ CD41−CD48− Lin−Sca-1+c-Kit+) in bone marrow increased transiently by approximately 5- and 8-fold, respectively, while the numbers of early progenitors (CMP, GMP, MEP, CLP) were mildly affected in the mutant mice 7–14 days after polyI:C treatment. While the more mature lineage committed mTOR−/− blood cells showed a cell cycle blockage and significantly increased apoptosis, mTOR−/− HSPCs and HSCs displayed a loss of quiescence and increased proliferation, as assessed by 5-bromodeoxyuridine incorporation assays, and a normal survival index. Transplantation of mTOR−/− BM cells into immunodeficient or syngeneic mice demonstrated that the mTOR−/− HSPCs failed to engraft and repopulate in the recipients. At the molecular level, mRNA microarray, quantitative real-time PCR and immunoblotting analyses of mTOR−/− HSPCs or Lin− cells revealed that the cell cycle inhibitor Rb was downregulated while the positive regulator of cell cycle E2F5 and pro-survival regulators MCL1 and BCL-xL were upregulated. mTOR deficiency abolished the activation of translational regulators S6K and 4E-BP but led to an increased activation of Akt. In addition, mTOR deficiency sensitized Lin− cells to DNA damage induced in vitro or in vivo by melphalan or mitomycin C (MMC), evidenced by a marked increase in γH2AX foci as well as DNA double-strand breaks (comet-tailed value of 30.2 ± 7.6 in mTOR−/− cells treated in vitro with melphalan and 37.6 ± 3.4 in mTOR−/− cells treated in vivo with MMC compared to 7.6 ± 2.1 in melphalan-treated WT cells and 17.3 ± 6.7 in MMC-treated WT cells, respectively). The increased DNA damage response can be attributed to an ∼300-fold reduction of the expression of FANCD2, a key component of the Fanconi DNA damage repair complex. Significantly, the effect of mTOR deficiency on Fanconi gene expression was specific to FANCD2, because the expression of other Fanconi proteins such as FANCA and FANCC was not affected in mTOR−/− Lin− cells. Intriguingly, the mTOR−/− Lin− cells phenocopied the DNA damage response of FANCD2−/− Lin− cells in vitro and in vivo. Similar effects of reduced FANCD2 expression and dampened DNA damage response were observed in human lymphoblasts treated with pp242, a mTOR kinase inhibitor. FANCD2-deficient human Fanconi anemia patient cells recapitulated the pp242-induced DNA damage phenotypes that could be rescued by FANCD2 reconstitution. Taken together, these results demonstrate that mTOR is a critical regulator of HSC quiescence and engraftment through the regulation of cell cycle machinery and is essential in multiple stages of hematopoiesis. Moreover, mTOR is required for maintaining genomic stability of HSPCs through modulation of the Fanconi anemia DNA damage response pathway. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (17) ◽  
pp. 3197-3207 ◽  
Author(s):  
Kirsteen J. Campbell ◽  
Mary L. Bath ◽  
Marian L. Turner ◽  
Cassandra J. Vandenberg ◽  
Philippe Bouillet ◽  
...  

Abstract Diverse human cancers with poor prognosis, including many lymphoid and myeloid malignancies, exhibit high levels of Mcl-1. To explore the impact of Mcl-1 overexpression on the hematopoietic compartment, we have generated vavP-Mcl-1 transgenic mice. Their lymphoid and myeloid cells displayed increased resistance to a variety of cytotoxic agents. Myelopoiesis was relatively normal, but lymphopoiesis was clearly perturbed, with excess mature B and T cells accumulating. Rather than the follicular lymphomas typical of vavP-BCL-2 mice, aging vavP-Mcl-1 mice were primarily susceptible to lymphomas having the phenotype of a stem/progenitor cell (11 of 30 tumors) or pre-B cell (12 of 30 tumors). Mcl-1 overexpression dramatically accelerated Myc-driven lymphomagenesis. Most vavP-Mcl-1/ Eμ-Myc mice died around birth, and transplantation of blood from bitransgenic E18 embryos into unirradiated mice resulted in stem/progenitor cell tumors. Furthermore, lethally irradiated mice transplanted with E13 fetal liver cells from Mcl-1/Myc bitransgenic mice uniformly died of stem/progenitor cell tumors. When treated in vivo with cyclophosphamide, tumors coexpressing Mcl-1 and Myc transgenes were significantly more resistant than conventional Eμ-Myc lymphomas. Collectively, these results demonstrate that Mcl-1 overexpression renders hematopoietic cells refractory to many cytotoxic insults, perturbs lymphopoiesis and promotes malignant transformation of hematopoietic stem and progenitor cells.


2015 ◽  
Vol 15 (1) ◽  
pp. 130-140 ◽  
Author(s):  
Qing-Shuo Zhang ◽  
Matthew Deater ◽  
Kathryn Schubert ◽  
Laura Marquez-Loza ◽  
Carl Pelz ◽  
...  

mBio ◽  
2020 ◽  
Vol 11 (3) ◽  
Author(s):  
Cristina Bono ◽  
Alba Martínez ◽  
Javier Megías ◽  
Daniel Gozalbo ◽  
Alberto Yáñez ◽  
...  

ABSTRACT Toll-like receptor (TLR) agonists drive hematopoietic stem and progenitor cells (HSPCs) to differentiate along the myeloid lineage. In this study, we used an HSPC transplantation model to investigate the possible direct interaction of β-glucan and its receptor (dectin-1) on HSPCs in vivo. Purified HSPCs from bone marrow of B6Ly5.1 mice (CD45.1 alloantigen) were transplanted into dectin-1−/− mice (CD45.2 alloantigen), which were then injected with β-glucan (depleted zymosan). As recipient mouse cells do not recognize the dectin-1 agonist injected, interference by soluble mediators secreted by recipient cells is negligible. Transplanted HSPCs differentiated into macrophages in response to depleted zymosan in the spleens and bone marrow of recipient mice. Functionally, macrophages derived from HSPCs exposed to depleted zymosan in vivo produced higher levels of inflammatory cytokines (tumor necrosis factor alpha [TNF-α] and interleukin 6 [IL-6]). These results demonstrate that trained immune responses, already described for monocytes and macrophages, also take place in HSPCs. Using a similar in vivo model of HSPC transplantation, we demonstrated that inactivated yeasts of Candida albicans induce differentiation of HSPCs through a dectin-1- and MyD88-dependent pathway. Soluble factors produced following exposure of HSPCs to dectin-1 agonists acted in a paracrine manner to induce myeloid differentiation and to influence the function of macrophages derived from dectin-1-unresponsive or β-glucan-unexposed HSPCs. Finally, we demonstrated that an in vitro transient exposure of HSPCs to live C. albicans cells, prior to differentiation, is sufficient to induce a trained phenotype of the macrophages they produce in a dectin-1- and Toll-like receptor 2 (TLR2)-dependent manner. IMPORTANCE Invasive candidiasis is an increasingly frequent cause of serious and often fatal infections. Understanding host defense is essential to design novel therapeutic strategies to boost immune protection against Candida albicans. In this article, we delve into two new concepts that have arisen over the last years: (i) the delivery of myelopoiesis-inducing signals by microbial components directly sensed by hematopoietic stem and progenitor cells (HSPCs) and (ii) the concept of “trained innate immunity” that may also apply to HSPCs. We demonstrate that dectin-1 ligation in vivo activates HSPCs and induces their differentiation to trained macrophages by a cell-autonomous indirect mechanism. This points to new mechanisms by which pathogen detection by HSPCs may modulate hematopoiesis in real time to generate myeloid cells better prepared to deal with the infection. Manipulation of this process may help to boost the innate immune response during candidiasis.


Blood ◽  
2000 ◽  
Vol 95 (2) ◽  
pp. 700-704 ◽  
Author(s):  
Kimberly A. Gush ◽  
Kai-Ling Fu ◽  
Markus Grompe ◽  
Christopher E. Walsh

Fanconi anemia (FA) is a genetic disorder characterized by bone marrow failure, congenital anomalies, and a predisposition to malignancy. FA cells demonstrate hypersensitivity to DNA cross-linking agents, such as mitomycin C (MMC). Mice with a targeted disruption of the FANCC gene (fancc −/− nullizygous mice) exhibit many of the characteristic features of FA and provide a valuable tool for testing novel therapeutic strategies. We have exploited the inherent hypersensitivity offancc −/− hematopoietic cells to assay for phenotypic correction following transfer of the FANCC complementary DNA (cDNA) into bone marrow cells. Murine fancc −/− bone marrow cells were transduced with the use of retrovirus carrying the humanfancc cDNA and injected into lethally irradiated recipients. Mitomycin C (MMC) dosing, known to induce pancytopenia, was used to challenge the transplanted animals. Phenotypic correction was determined by assessment of peripheral blood counts. Mice that received cells transduced with virus carrying the wild-type gene maintained normal blood counts following MMC administration. All nullizygous control animals receiving MMC exhibited pancytopenia shortly before death. Clonogenic assay and polymerase chain reaction analysis confirmed gene transfer of progenitor cells. These results indicate that selective pressure promotes in vivo enrichment offancc-transduced hematopoietic stem/progenitor cells. In addition, MMC resistance coupled with detection of the transgene in secondary recipients suggests transduction and phenotypic correction of long-term repopulating stem cells.


Sign in / Sign up

Export Citation Format

Share Document