scholarly journals Emerging Mechanisms of Cellular Iron Transport and Trafficking

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. SCI-2-SCI-2
Author(s):  
Caroline Philpott ◽  
Moon-Suhn Ryu

Abstract Iron is an essential nutrient required by every cell in the human body, yet it can also be a potent cellular toxin. Iron is essential because enzymes that require iron co-factors (namely, heme, iron-sulfur clusters, mono- and dinuclear iron centers) are involved in virtually every major metabolic process in the cell. Iron deficiency continues to be the most common nutritional deficiency in the world, while iron overload is a feature of an increasing number of human diseases, including genetic disorders such as hereditary hemochromatosis, thalassemias, and Friedreich ataxia, as well as chronic inflammatory diseases of the liver, such as hepatitis C and steatohepatitis. The biochemical mechanisms by which iron causes toxicity are not completely known. Previously, we identified Poly rC-binding protein 1 (Pcbp1) as a protein that directly binds and delivers iron to ferritin, the major iron storage protein in mammalian cells. Pcbp1 and its paralog Pcbp2 are multifunctional adaptors that bind cellular RNA, DNA, proteins, and iron, altering the fate of their binding partners. Ferritin is a large polypeptide, containing 24 subunits of H- and L-chains assembled into a hollow sphere. Reduced (ferrous) iron enters the sphere through pores formed between the subunits and is oxidized on the interior surface to form nanocrystals of ferric oxyhydroxides. Pcbp1 is the first example of an iron chaperone- a protein that specifically binds iron ions and delivers them to target proteins, such as ferritin, through direct protein-protein interactions. In cultured cells, Pcbp1 is also required for efficient iron delivery to non-heme enzymes in addition to ferritin. These enzymes include the mononuclear iron-containing prolyl hydroxylases that regulate HIF and the dinuclear iron-containing deoxyhypusine hydroxylase, which is required for the modification of lysine to hypusine. In the adult human, 70% of total body iron is present in circulating erythrocytes, which are produced by the bone marrow at a rate of 2 million reticulocytes per second. We examined the roles of Pcbp1 and Ncoa4 in this extraordinary flux of iron through the erythron. Ncoa4 is the cargo receptor that recruits ferritin into the autophagosome for degradation in the lysosome. Using an in vitro model of erythroid differentiation, we showed that depletion of Pcbp1 or Ncoa4 impeded trafficking of iron through ferritin, which impaired the synthesis of heme and hemoglobin. Mice with tamoxifen-induced Pcbp1 deficiency exhibited a microcytic anemia typical of iron deficiency, with activation of compensatory erythropoiesis. The role of ferritin in erythropoiesis has been controversial, but our studies indicate that iron flux through ferritin is an obligatory process in the early stages of erythroid terminal differentiation. Our in vitro studies of erythrocytes revealed that the interactions of Pcbp1 and Ncoa4 with ferritin changed during differentiation, with maximal binding of Pcbp1 at early stages and Ncoa4 at late stages. These binding activities are regulated by cellular iron in mechanistically distinct ways. Ongoing studies of Pcbp1 deficiency in other murine tissues suggest the importance of iron chaperones in maintaining the bioavailable pool of iron in mammalian cells. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1862-1862
Author(s):  
Gregory J. Cost ◽  
Morayma Temoche-Diaz ◽  
Janet Mei ◽  
Cristina N. Butterfield ◽  
Christopher T. Brown ◽  
...  

Abstract RNA guided CRISPR genome editing systems can make specific changes to the genomes of mammalian cells and have the potential to treat a range of diseases including those that can be addressed by editing hepatocytes. Attempts to edit the liver in vivo have relied almost exclusively on the Cas9 nucleases derived from the bacteria S treptococcus pyogenes or Staphylococcus aureus to which humans are commonly exposed. Pre-existing immunity to both these proteins has been reported in humans which raises concerns about their in vivo application. In silico analysis of a large metagenomics database followed by testing in mammalian cells in culture identified MG29-1, a novel CRISPR system which is a member of the Type V family but exhibits only 41 % amino acid identity to Francisella tularensis Cas12a/cpf1. MG29-1 is a 1280 amino acid RNA programmable nuclease that utilizes a single guide RNA comprised of a 22 nucleotide (nt) constant region and a 20 to 25 nt spacer, recognizes the PAM KTTN (predicted frequency 1 in 16 bp) and generates staggered cuts. MG29-1 was derived from a sample taken from a hydrothermal vent and it is therefore unlikely that humans will have developed pre-existing immunity to this protein. A screen for sgRNA targeting serum albumin in the mouse liver cell line Hepa1-6 identified 6 guides that generated more than 80% INDELS. The MG29-1 system was optimized for in vivo delivery by screening chemical modifications to the guide that improve stability in mammalian cell lysates while retaining or improving editing activity. Two lead guide chemistries were evaluated in mice using MG29-1 mRNA and sgRNA packaged in lipid nanoparticles (LNP). Three days after a single IV administration on-target editing was evaluated in the liver by Sanger sequencing. The sgRNA that was the most stable in the in vitro assay generated INDELS that ranged from 20 to 25% while a sgRNA with lower in vitro stability failed to generate detectable INDELs. The short sgRNA and small protein size compared to spCas9 makes MG29-1 an attractive alternative to spCas9 for in vivo editing applications. Evaluation of the potential of MG29-1 to perform gene knockouts and gene additions via non-homologous end joining is ongoing. Disclosures No relevant conflicts of interest to declare.


2002 ◽  
Vol 115 (10) ◽  
pp. 2165-2177
Author(s):  
Khristy J. Thompson ◽  
Michael G. Fried ◽  
Zheng Ye ◽  
Phillip Boyer ◽  
James R. Connor

Ferritin is traditionally considered a cytoplasmic iron-storage protein,but recent reports indicate that it is also found in cell nuclei. Nuclear ferritin has been proposed to be involved in both the protection of DNA and the exacerbation of iron-induced oxidative damage to DNA. We demonstrate that H-rich ferritin is present in the nucleus of human astrocytoma tumor cells. To study the mechanism and regulation of ferritin translocation to the nucleus,we developed a cell culture model using SW1088 human astrocytoma cells. Changes in cellular iron levels, cytokine treatments and hydrogen peroxide exposure affected the distribution of ferritin between the cytosol and the nucleus. Ferritin enters the nucleus via active transport through the nuclear pore and does not require NLS-bearing cytosolic factors for transport. Furthermore, H-rich ferritin is preferred over L-rich ferritin for uptake into the nucleus. Whole cell crosslinking studies revealed that ferritin is associated with DNA. Ferritin protected DNA from iron-induced oxidative damage in both in vitro and in cell culture models. These results strongly suggest a novel role for ferritin in nuclear protection. This work should lead to novel characterization of ferritin functions in the context of genomic stability and may have unparalleled biological significance in terms of the accessibility of metals to DNA. The knowledge generated as a result of these studies will also improve our understanding of iron-induced damage of nuclear constituents.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 780-780
Author(s):  
Hal E. Broxmeyer ◽  
Maegan L. Capitano ◽  
Liang Zhao ◽  
Scott Cooper ◽  
Charles S. Abrams

Abstract There are still unknowns regarding homeostatic regulation of hematopoietic stem (HSC) and progenitor (HPC) cells. Deciphering these processes are important for understanding and treating hematopoietic diseases. Phosphatidylinositol is a rare membrane structure lipid, but is critical for cellular signaling upon phosphorylation by lipid kinases to generate phosphoinositide. While phosphoinositide pathways contribute to events linked to the cytoskeleton, little is known of these pathways in regulating hematopoiesis. Critical to this pathway are phosphatidylinositol transfer proteins (PITPs) that in vitro enhance transfer of aqueous insoluble phosphatidylinositol from one membrane to another. Class I PITP proteins PITP α and β are highly conserved, small, and ubiquitously expressed in mammalian cells. To test the hypothesis that phosphatidylinositol signaling contributes to hematopoiesis, we generated conditional knock out mice that lack either PITPα single isoform (PITPαfl/fl PF4Cre+) or both PITPα and PITPβ (PITPαfl/fl βfl/fl PF4Cre+) specifically in their platelets and megakaryocytes, and observed a bone marrow (BM) HSC/HPC phenotype. BM from these mice and their littermate controls were evaluated for absolute numbers of nucleated cells, HSC, and HPC. Cells were analyzed by rigorous phenotyping for long-term (LT)-HSC, short-term (ST)-HSC, multipotential (MPP), common myeloid (CMP), and granulocyte macrophage (GMP) progenitors. They were also assessed for functional HPC by colony assays in vitro for multi-cytokine (Epo, GM-CSF, IL-3, SCF, hemin) stimulated granulocyte macrophage (CFU-GM), erythroid (BFU-E), and multipotential (CFU-GEMM) progenitors, and for their cycling status using a high specific activity tritiated thymidine kill assay. PITPα-/-, and to a greater extent PITPα/β-/-, progenitor cells demonstrated significant decreases in LT-HSC and ST-HSC per femur. While there were no significant changes in numbers of MPP, CMP, and GMP in the PITPα and PITPα/β-/- BM compared to controls, there were significant decreases of approximately 50% in numbers of CFU-GM, BFU-E, and CFU-GEMM per femur. PITC-/- HPC were in a slow or non-cycling state compared to the rapid cell cycle (40-57% in S-phase) of control HPC. Thus PITPα-/- and PITPα/β -/- BM cells were associated with decreased HSC and functional HPC numbers. To evaluate mechanisms for this phenotype, we focused on BM megakaryocytes, as they have been implicated in microenvironmental regulation of hematopoiesis, and PITPα and PITPα/β activities are associated with megakaryocyte/platelet function. BM derived TPO-culture expanded megakaryocytes were allowed to condition medium for 48 hours, and conditioned medium (CM) from PITPα-/-, PITPα/β-/-, and control BM megakaryocytes were assayed for effects on colony formation by multicytokine stimulated BM cells derived from normal mice. CM from PITPα-/- and PITPα/β-/- megakaryocytes, but not from control mice, significantly suppressed colony formation by CFU-GM, BFU-E and CFU-GEMM (by ~50%). Limiting dilution analysis of the CM demonstrated that PITPα/β-/- cells had more potent suppressor activity than PITPα-/- cells. Bioplex analysis of the CM from PITPα -/- and PITPα/β -/- megakaryocytes demonstrated significantly higher levels of cytokines/chemokines with known myelosuppressive activities (including: TNF-α, VEGF, LIF, IP-10, ENA-78, MDC, MIG, and MIP-1α). However, ELISA analysis of TGF-β1, demonstrated minimal protein in BM flushes from control mice, but large amounts of TGF-β (>350 pg/ml) in BM flushes from the PITPα/β -/- mice. CM from PITPα and α/β-/- megakaryocytes also contained highly elevated TGF-β protein. Thus, we hypothesized that the effect of PITP -/- on the suppression of HPC colony formation was mediated by TGF-β. The myelosuppressive CM derived from PITPα and PITPα/β -/- megakaryocytes was completely neutralized by a monoclonal TGF-β antibody. This demonstrates that PITPα and PITP α/β-/- megakaryocytes produce elevated TGF-β that at least in part, and possibly in synergy with other myelosuppressive cytokines/chemokines, decreases numbers of HSC and functional HPC. Our studies demonstrate a link between PITPα and α/β and TGF-β levels with significant effects on HSCs and HPCs, thus demonstrating involvement of the phosphoinositide pathway in homeostatic regulation of hematopoiesis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 3-3
Author(s):  
Areum Han ◽  
Alena V. Yermalovich ◽  
Vanessa Lundin ◽  
Daniel S. Pearson ◽  
Mariam Hachimi ◽  
...  

Erythropoiesis is an intricate process by which lineage-committed erythroid progenitors become mature red blood cells. Reticulocytes are terminal-staged, immature red blood cells with residual RNA after enucleation. In the absence of pathology, reticulocytes are efficiently processed into mature red blood cells and typically represent a small percentage of cells in human peripheral blood. In contrast, when differentiated in vitro from pluripotent stem cells or CD34+ progenitor cells, red cells tend to arrest at the reticulocyte stage. Recent studies have highlighted that uridylation by Terminal Uridylyl Transferases (TUTases) occurs on a broad spectrum of RNA classes in mammalian cells. Oligo-uridylated RNA is recognized by exoribonucleases and targeted for decay. We posited that the machinery behind RNA degradation that accompanies terminal erythropoiesis might involve RNA tail editors coupled to exonuclease activity. Utilizing constitutional murine knockout models, we observed that blood from the TUTase Zcchc6 RNA editor knockout embryos exhibited reticulocytosis and a terminal maturation defect, as documented by FACS, histology, and hematological profiling. Murine strains deficient in the downstream exonuclease Dis3l2 phenocopied the RNA decay defect of the Zcchc6 KO. Conditional knockout murine models of the TUTase-Dis3l2 axis driven by the red cell specific Erythropoietin Receptor-Cre exhibited comparable phenotypes, suggesting a cell intrinsic and niche-independent role for the TUTase-Dis3l2 axis in promoting red blood cell maturation. We are modulating the expression of this axis by various methods to optimize modeling of hemoglobinopathies such as sickle cell anemia. Disclosures No relevant conflicts of interest to declare.


1996 ◽  
Vol 314 (1) ◽  
pp. 139-144 ◽  
Author(s):  
Paolo SANTAMBROGIO ◽  
Sonia LEVI ◽  
Anna COZZI ◽  
Barbara CORSI ◽  
Paolo AROSIO

Mammalian ferritins are iron-storage proteins made of 24 subunits of two types: the H- and L-chains. L-chains, in contrast with H-chains, lack detectable ferroxidase activity. When ferritins were subjected to iron loading in vitro with increments near the saturation limit of 4000 Fe atoms per molecule, the homopolymers of human H-chains formed insoluble aggregates, caused by non-specific iron hydrolysis, whereas the homopolymers of L-chains remained soluble and incorporated most of the available iron. To analyse the molecular reasons for the difference, Glu-57 and Glu-60, which are conserved and exposed on the cavity of L-chains, were substituted with His, as in H-chains. The double substitution made the L-homopolymers as sensitive as the H-homopolymers to the iron-induced aggregation, whereas the opposite substitution in the H-chain increased homopolymer resistance to the aggregation only marginally. Millimolar concentrations of citrate and phosphate increased iron incorporation in H-homopolymers by reducing non-specific iron hydrolysis, but inhibited that in L-homopolymers by sequestering available iron. The data indicate that the specific iron incorporation into L-homopolymers is mainly due to the iron-nucleation capacity of Glu-57, Glu-60 and other carboxyl groups exposed on the cavity; in contrast, the specificity of iron incorporation into H-homopolymers is related to its ferroxidase activity, which determines rapid Fe(III) accumulation inside the cavity. The finding that ferroxidase centres are essential for the incorporation of iron in the presence of likely candidates of cellular iron transport, such as phosphate and citrate, confirms their importance in ferritin function in vivo.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1308-1308
Author(s):  
Dominique Bluteau ◽  
Larissa Lordier ◽  
Iléana Antony-Debré ◽  
Abdelali Jalil ◽  
Céline Legrand ◽  
...  

Abstract Abstract 1308 Megakaryocytes (MK) are unique mammalian cells that undergo polyploidization during differentiation, which leads to an increase in cell size and protein production that precedes platelet production. The molecular basis of MK polyploidization, denoted endomitosis, which is strongly altered in hematological malignancies and various other disease states, remain poorly understood. Recent evidence demonstrates that endomitosis is a consequence of a late failure in cytokinesis, associated with a contractile ring defect. Here, we demonstrate that the myosin non-muscle IIB heavy chain (MYH10) is expressed in immature human and mouse megakaryocytes and its expression is repressed during differentiation. In immature MK, MYH10 is specifically localized in the contractile ring while MYH9 is mainly present in the cytoplasm suggesting that they occupy two different functions during MK differentiation. Importantly, MYH10 down-modulation by shRNA or by addition of a chemical inhibitor blebbistatin, increases polyploidization by inhibiting the return of 4N cells to 2N. Conversely, re-expression of MYH10 in MKs prevents polyploidization and the transition of 2N cells to 4N cells. Furthermore, we demonstrated that RUNX1 directly repress the transcription of MYH10. In vitro and in vivo RUNX1 invalidation inhibits MK polyploidization and increases expression of MYH10. Accordingly, in patients with a germline mutation of RUNX1 (FPD/AML), MYH10 is still expressed in platelets. Altogether ours results demonstrated that the RUNX1-mediated silencing of MYH10 is needed for the switch from mitosis to endomitosis linking thus polyploidization with MK differentiation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3911-3911
Author(s):  
Akira Niwa ◽  
Tatsutoshi Nakahata ◽  
Megumu K Saito

Abstract Onset of acute myeloid leukemia (AML) has been accounted for by cooperation between multiple genetic alterations which induce abnormal control of various cellular pathways. Among the previously listed leukemogenic lesions, AML1-ETO fusion (AE) generated by translocation (8;21) (q22;q22) is one of the common mutations observed in 20-40% of patients. AE affects transcriptional regulation associated with hematopoietic differentiation, while 60% of AE-positive AML cases are shown to have together other types of mutation of genes involved in cell proliferation, such as receptor tyrosine kinase (RTK) c-kit and FLT3. However, the detailed mechanisms of how they work in the very early stages of leukemogenesis and what unknown "cooperative " cues function in those periods. From this viewpoint, in order to identify novel cellular molecules involved in the acquisition of leukemic phenotypes, we have conducted the gene-trap strategy-based phenomic screen in the use of pluripotent stem cell (PSC)-derived hematopoietic culture. Through our screening, we found that the functional knockdown of a gene NSFL1c, also known as p47, enhanced the activities of hematopoietic progenitor cells harboring AE to show leukemic phenotype both in vivo and in vitro: Cells differentiated from AE-PSCs which have additionally the poly A trapping sequence inserted in NSFL1c locus showed doubled efficiency in engraftment into immunodeficient NOG mice than cells without trapping (3.1% v.s. 1.3%, 16 weeks after intra bone marrow transplantation), and also showed the significantly higher colony replating efficacy in methylcellulose colony forming assay. In order to clear what lineages of cells were most responsible for those phenomena, we next performed those colony forming experiments after sorting of CD34+CD43+CD13- immature cells, CD34+/-CD43+CD13+ myeloid cells and CD71+CD41+ erythro-megakaryocytic subpopulations of cells. As a result, CD34+/-CD43+CD13+ myeloid precursors showed the strongest tendencies to emerge highly proliferative clones followed by CD34+CD43+CD13- immature progenitors in the presence of NSF1c trapping. Interestingly, those activities were cancelled in the absence of AE expression. NSFL1c is one of the component of NEMO complex which binds to ubiquitinated NEMO and induces its degradation, resulting in reduced IKK and elevated NFkB activities. In AML, elevated NF-κB pathways have been detected in more than 30% of patients. Although NF-κB signaling networks have proved induced by inflammatory and immune signals, and previous studies in vivo and in vitro showed their abnormal activities make leukemia cells escape from cell death and go into abnormal proliferation, the detailed mechanisms of how they are involved in the pathogenesis, in particular at the very early stages of the leukemogenesis, are well not defined. Our data therefore reflect the novel mechanisms behind the deviation of progenitor cell fate from normal to abnormal pathway leading to the emergence of leukemic initiating cells, and suggested the myeloid-biased leukemogenic potentials. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1485-1485
Author(s):  
Zheng Tao Hu ◽  
Zhi Hua Huang ◽  
Xiao Bo Cen ◽  
Tao Wang ◽  
Kun Zhuang ◽  
...  

Abstract Abstract 1485 Recombinant human granulocyte colony-stimulating factors (rhG-CSF) including filgrastim, lenograstim and pegfilgrastim are widely used to treat chemotherapy-induced neutropenia. However, it remains a challenge to manage severe neutropenia in cancer patients after high dose chemotherapy. The activation of G-CSFR by the G-CSF requires dimerization of two receptor chains bound to two G-CSF ligands. We hypothesized that a G-CSF dimer might generate a faster in vivo response, thus to benefit patients with severe neutropenia. F-627 is a recombinant human G-CSF dimer expressed in mammalian cells. In vitro, F-627 was able to activate STAT3 and to stimulate the proliferation of 32D-GCSFR and M-NSF60 cell lines. To evaluate the efficacy of F-627 in chemotherapy-induced neutropenia, 36 adult cynomolgus monkeys (3/sex) were injected with cyclophoshamide (CY, i.v., 60 mg/kg on day 0 and 65 mg/kg on day 1). Animals were randomized to receive (s.c.) either carrier, or F-627 on day 5 and day 10, at 25, 60 and 150 μg/kg, or rhG-CSF at 10 μg/kg /day (daily injection), or pegfilgrastim at 60 μg/kg on day 5 and day 10. Pharmacokinetics and pharmacodynamics (PK/PD) were evaluated. Significant absolute neutrophil count (ANC) increase was observed in animals treated with F-627 at 25 μg/kg compared to carrier-treated monkeys. At nadir, F-627 at 60 μg/kg generated optimal ANC response with 6.9- and 3.0-fold higher ANC compared to pegfilgrastim- (0.17 × 109/L) and rhG-CSF- (0.39 × 109/L) treated monkeys, respectively. The PK parameters of F627 at 60 ug/kg including MRT, Cmax, Tmax, AUC and CL were comparable to that of pegfilgrastim-treated monkeys, despite the relative G-CSF molar dose in F-627-treated animals was 2.5x lower than the pegfilgrastim-treated animals. The results demonstrate that the G-CSF dimer generated a faster response in CY-treated monkeys compared to the G-CSF monomer-treated animals, suggesting that F-627 could benefit cancer patients with severe neutropenia after high-dose chemotherapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. SCI-26-SCI-26
Author(s):  
Clara Camaschella ◽  
Antonella Nai ◽  
Alessia Pagani ◽  
Laura Silvestri

Abstract Abstract SCI-26 Hepcidin inhibition is a process essential to increase iron release from duodenal cells and macrophages to plasma in conditions of high iron requests such as iron deficiency, hypoxia and erythropoietic expansion. Among the reported potential inhibitors of hepcidin the serine protease matriptase-2 has a strong effect in vivo both in mice and in humans. Matriptase-2 is a member of the transmembrane serine protease (TTPS) family, encoded by TMPRSS6 gene, whose expression is tissue restricted, mainly to the liver and to a lesser extent to kidney and olfactory epithelium. The role of matriptase-2 in iron metabolism was first defined in the Mask mouse, which after birth develops a type of iron deficiency anemia refractory to oral iron and a peculiar pattern of hair loss, because of inappropriate overexpression of hepcidin and impaired iron absorption, a phenotype due to the deletion of the matriptase-2 serine protease domain. An identical phenotype is reported in Tmprss6 -/- mice and a correspondent phenotype is observed in iron-refractory iron-deficiency anemia (IRIDA) patients. We have shown that matriptase-2 cleaves through a proteolytic processing the bone morphogenetic protein (BMP) co-receptor hemojuvelin from plasma membrane in vitro, indicating that the suppression of the BMP signaling is essential for hepcidin inhibition. The finding also suggests that the BMP pathway requiring hemojuvelin as coreceptor is the main iron-dependent pathway of hepcidin regulation. The hepcidin inhibitory effect is observed also in zebrafish and is abolished in the human homologue of Mask. We have investigated several missense TMPRSS6 mutants affecting different domains of the protein, reported in IRIDA patients, in comparison with wild type matriptase-2 and Mask. Transient overexpression in hepatoma cells shows that all mutants are deficient in the ability to inhibit the hepcidin promoter in a classic luciferase-based assay and that the decreased hepcidin inactivation broadly corresponds to the inability to cleave hemojuvelin from plasma membrane. Studies of the in vitro processing of the mutants indicate that determinants of the pathogenic effect others than the protease activity are the intracellular processing and the ability of self-activation of matriptase-2. These results have implications for the molecular pathogenesis of IRIDA. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 701-701
Author(s):  
Jana Krosl ◽  
Jessica Lebrun ◽  
Jalila Chagraoui ◽  
Julie Lessard ◽  
Aline Mamo ◽  
...  

Abstract Abstract 701 Using Meis1 as a bait to screen the cDNA library from Hoxa9+Meis1 leukemia we identified Baf250a, a component of the chromatin remodeling Swi/Snf complex, as a Meis1-interacting protein. In mammalian cells, co-immunoprecipitation experiments confirmed this interaction and showed that only a minor fraction of cellular Meis1 interacts with Baf250a, suggesting a restriction of Meis1-Baf250a( Swi/Snf ) interactions to a specific cellular context. To reveal the role of Baf250a action in normal and leukemic HSC behavior we generated mouse strains expressing Baf250a protein lacking amino acids encoded by exons 2 and 3. Baf250a+/Δ mice were born in expected proportions and exhibited no hematological abnormalities. Intercrosses of Baf250a+/Δ (C57Bl/6J) adults yielded normal proportions of wild-type(WT), Baf250a+/Δ and Baf250aΔ/Δ day E14.5dpc embryos, but no viable Baf250aΔ /Δ newborn mice. These, however, could be obtained in a mixed C57Bl/J6 x Sve129 background, suggesting existance of a mouse strain-specific modifier(s) of Baf250a activity. Examination of E14.5 dpc fetal liver (FL) cell populations obtained from Baf250aΔ /Δ and WT littermates showed that cellularity and clonogenic progenitor content of Baf250aΔ/Δ FL was comparable to the WT controls, and that erythoid, megakaryocytic and myeloid cell lineages developed normally in the presence of Baf250aΔ. Transplantation experiments in conditions of limit dilution revealed that Baf250aΔ/Δ FL comprised 8-12-fold higher HSC numbers than WT controls. After transplantation into adult WT recipients the Baf250aΔ /Δ and WT HSCs exhibited comparable proliferation potential (output of mature cells per individual transplanted HSC), suggesting that of Baf250aΔ conferred no repopulation advantage to mutant HSC, but rather enhanced the ability of fetal liver microenvironment to support expansion of HSC populations. To explore this possibility we next examined the ability of Baf250aΔ /Δ stromal cell cultures to support the in vitro self-renewal of stem/progenitor cell populations identifiable as late cobblestone area forming cells (CAFCday28), and found that Baf250aΔ/Δ stromal cell layers increased the numbers of cobblestone areas 4-6-fold compared to controls, and increased the probability of CAFCday28 self renewal divisions (n=24, p<0.05) compared to controls as determined by their ability to form secondary late cobblestone areas. These observations thus suggested that Baf250a acts as a negative regulator of fetal HSC populations. However, we also found that Baf250aΔ/Δ genetic background accelerated the onset of Hoxa9 (n=7, mean latency 140 and 90 days for WT and Baf250aΔ/Δ, respectively, p<0.05) and NUP98-Hoxa9 + Meis1 (n=8, mean latency 125 and 49 days for WT and Baf250aΔ/Δ, respectively, p<0.005) induced AML, implying that Baf250a may also act as an intrinsic regulator of primitive hemopoietic cells. Together, results of our experiments identify a novel mechanism that limits the expansion FL HSC populations regulated by the Baf250a, and suggest a tumor suppressor function for this protein in Hoxa9-induced leukemogenesis. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document