Transcription Factor Induction of Ectopic Vascular Blood Stem Cell Niches In Vivo

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 525-525
Author(s):  
Elliott J. Hagedorn ◽  
Julie R. Perlin ◽  
Rebecca J. Freeman ◽  
Clara Mao ◽  
Inés Fernández-Maestre ◽  
...  

The hematopoietic stem and progenitor cell (HSPC) niche is a supportive microenvironment comprised of distinct cell types, including specialized vascular endothelial cells (ECs) that directly interact with HSPCs and promote stem cell function. Utilizing spatial transcriptomics, in combination with tissue-specific RNA-seq, we identified 29 genes selectively enriched in ECs of the zebrafish fetal hematopoietic niche. Using upstream regulatory sequences for two of these genes, mrc1a and selectin E (sele), we generated GFP reporter lines that allowed us to selectively isolate niche ECs for ATAC-seq. This analysis identified 6,848 regions of chromatin that were accessible in niche ECs but not ECs from other tissues. Several of these regions were associated with the 29 genes. To evaluate whether these regions might be enhancers we coupled them to GFP and injected them into embryos. 12/15 sequences drove GFP expression in niche ECs. Upon closer examination of the mrc1a and sele genes, we identified enhancer sequences as short as 125 bp and 158 bp, respectively, which drove niche EC-specific expression. A genome-wide motif enrichment analysis of the 6,848 uniquely open chromatin regions revealed that Ets, SoxF and Nuclear Hormone Receptor (RXRA/NR2F2, specifically) sites were most enriched. In contrast, 4,522 pan-EC elements were enriched for Ets sites but not SoxF or NHR motifs. Using mutant variants of the 125 bp and 158 bp enhancer sequences, we demonstrated that Ets, SoxF and RXRA/NR2F2 sites were independently required for specific transgene expression. Gel shift experiments demonstrated that NR2F2 could bind the 125 bp and 158 bp zebrafish enhancers and this binding was disrupted upon mutation of the NR2F2 binding sites. Knockdown of the endogenous zebrafish nr2f2 gene resulted in a loss of expression of the 125 bp mrc1a enhancer-GFP construct and a significantly reduced number of HSPCs in the fetal niche. We next injected pools of human transcription factors, including at least one member from each of the three families, under the control of a ubiquitous promoter. Strikingly, we found that a combination of ETV2 or ETS1 with SOX7 and NR2F2 generated ectopic patches of mrc1a+ niche ECs that recruited runx1+ HSPCs outside of the endogenous niche. Using high-resolution live cell imaging we could observe HSPCs initially arriving at the ectopic sites, lodging for several hours and then eventually dividing and migrating away from the site through circulation. HSPCs localized to the ectopic regions were found in both intravascular and extravascular spaces, and were often enwrapped by ECs and in contact with cxcl12a+ stromal cells, similar to what is observed in the endogenous niche. Ectopic regions of niche EC gene expression were similarly observed when alternative regulatory elements were used for transcription factor overexpression, including a pan-EC enhancer (nrp1b), a muscle promoter (mylz2) and a heat shock promoter (hsp70). These results suggest the three-factor combinations are sufficient to reprogram niche EC identify in vivo. Lastly, we evaluated by RNA-seq the expression of our niche EC signature in the zebrafish kidney marrow (the site of adult hematopoiesis) and in ECs from multiple organs of the mouse, including the heart, kidney, liver, lung and bone marrow, at multiple stages of development (E11-13, E14-15, E16-17, P2-P4 and adult). Strikingly, 23/29 genes were highly expressed in ECs of the zebrafish kidney and 21/29 genes were enriched in the ECs of a mammalian hematopoietic organ - the fetal liver and/or adult bone marrow - relative to their expression in ECs from non-hematopoietic organs at the same stage. Notably, for a subset of the genes the expression patterns mirrored the temporal dynamics of HSPC ontogeny in the mouse, showing robust expression in fetal liver ECs and then later in adult bone marrow ECs with a concomitant reduction in liver ECs. An analysis of transcription factor expression within these EC populations revealed that Ets1, the SoxF factor Sox18, and Nr2f2 were the most highly expressed members of the Ets, Sox and NHR families. Collectively our work has uncovered a conserved gene expression signature and transcriptional regulatory program unique to the vascular niche of hematopoietic organs. These findings have important implications for designing a synthetic vascular niche for blood stem cells or for modulating the niche in a therapeutic context. Disclosures Zon: Fate Therapeutics: Equity Ownership; Scholar Rock: Equity Ownership; CAMP4: Equity Ownership.

2004 ◽  
Vol 200 (7) ◽  
pp. 871-882 ◽  
Author(s):  
Kam-Wing Ling ◽  
Katrin Ottersbach ◽  
Jan Piet van Hamburg ◽  
Aneta Oziemlak ◽  
Fong-Ying Tsai ◽  
...  

GATA-2 is an essential transcription factor in the hematopoietic system that is expressed in hematopoietic stem cells (HSCs) and progenitors. Complete deficiency of GATA-2 in the mouse leads to severe anemia and embryonic lethality. The role of GATA-2 and dosage effects of this transcription factor in HSC development within the embryo and adult are largely unexplored. Here we examined the effects of GATA-2 gene dosage on the generation and expansion of HSCs in several hematopoietic sites throughout mouse development. We show that a haploid dose of GATA-2 severely reduces production and expansion of HSCs specifically in the aorta-gonad-mesonephros region (which autonomously generates the first HSCs), whereas quantitative reduction of HSCs is minimal or unchanged in yolk sac, fetal liver, and adult bone marrow. However, HSCs in all these ontogenically distinct anatomical sites are qualitatively defective in serial or competitive transplantation assays. Also, cytotoxic drug-induced regeneration studies show a clear GATA-2 dose–related proliferation defect in adult bone marrow. Thus, GATA-2 plays at least two functionally distinct roles during ontogeny of HSCs: the production and expansion of HSCs in the aorta-gonad-mesonephros and the proliferation of HSCs in the adult bone marrow.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 358-358 ◽  
Author(s):  
Gang Huang ◽  
Pu Zhang ◽  
Steffen Koschmieder ◽  
Joseph D. Growney ◽  
D. Gary Gilliland ◽  
...  

Abstract PU.1 is expressed in hematopoietic stem cells (HSC), progenitors and differentiating blood cells except terminally differentiated T cells, erythrocytes and megakaryocytes. PU.1 is required for commitment of HSC to multiple lineages. PU.1 −/− embryos die perinatally and fail to generate myeloid and B cells. We previously reported that a DNase I hypersensitive site located 14 kb upstream of the PU.1 transcription start site (−14 DHS) confers myelomonocytic specific gene expression. Targeted deletion this DHS fragment in mice results in a decrease in PU.1 expression in bone marrow to 20% of wild type levels, subsequently leading to a profound decrease in macrophages and B cells. Within the DHS fragment is a “core” consisting of a distal (296bp) and a proximal (253bp) region, which are highly conserved among different species. The PU.1 promoter by itself cannot direct gene expression in vivo. However, −14 DHS confers to the promoter the ability to direct expression of a reporter gene in granulocytes, monocytes, and B-cells of transgenic mice. The proximal region can itself direct high-level gene expression. The proximal region contains 3 AML1 sites. These results, along with data indicating that PU.1 expression is selectively absent from Aml1 −/− embryos (Okada, et al, Oncogene. 1998), suggested that AML1 is likely to be upstream of PU.1. Electro-mobility gel shift assays and chromatin immunoprecipitation assays confirmed that AML1 binds to all 3 AML1 sites both in vitro and in vivo. Mutation of the 3 AML1 sites dramatically reduced the DHS activity of conferring gene expression. We used real time PCR to quantitatively measure PU.1 expression in both embryonic and adult hematopoiesis. We found that PU.1 expression was completely lost in the 9.5 dpc yolk sac, 10.5 dpc AGM and fetal liver of Aml1−/− embryos, suggesting that AML1 is required for PU.1 expression during embryonic hematopoiesis. To evaluate the effects of AML1 loss in the adult hematopoiesis, we employed a conditional Aml1 knockout allele in which LoxP flanked Aml1 (Aml1F/F) was excised by Mx1 promoter driven Cre expression following injection of pIpC. These mice show that Aml1 is not required for maturation of myeloid lineages in adult mice. However, these mice develop a mild myeloproliferative phenotype characterized by increasing in bone marrow and peripheral blood (PB) neutrophils, a 5 fold increasing in HSC, and 2–3 fold increasing myeloid progenitors. Spleen and liver contain infiltration by myeloid cells. These mice also display a dramatic decrease (~80%) in PB platelets and bone marrow megakaryocytes. Furthermore, there are significant blocks in lymphoid development, including reduced numbers of pre-B, pro-B and mature B cells, as well a block in T cell maturation at the DN2 (CD4−;CD8−;CD44+;CD25+) stage. We observed a 70% reduction of PU.1 expression in sorted HSC, progenitors, Gr1+/Mac1+ and B-cells from these mice relative to control mice. In contrast, upregulation of 3–5 fold expression in Ter119+, CD41+, and T cells in these mice compared to controls. Our data shows that PU.1 is a critical target gene of AML1, and AML1 regulates PU.1 in both positive and negative way. We are currently testing the ability of restoration of PU.1 expression to rescue specific defects in Aml1F/F; Tg (Mx1-cre) mice, as well as investigating the role of decreased PU.1 expression in human AML in which the function of AML1 is disrupted.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1357-1357
Author(s):  
Andrew C. Perkins ◽  
Janelle R. Keys ◽  
Denise J. Hodge ◽  
Michael R. Tallack

Abstract Erythroid Kruppel-Like Factor (EKLF) is a zinc finger transcription factor which is essential for β-globin gene expression. Knockout mice die from anemia at E15, but restoration of globin chain imbalance does not rescue anemia or increase survival. Cell lines derived from EKLF null mice undergo proliferation arrest upon reactivation of a conditional EKLF-ER fusion protein, suggesting a role in cell cycle control. A transcriptional profiling experiment comparing the global gene expression in EKLF null and wild type fetal liver identified many differentially expressed genes, a number of which function in G1 and at the G1/S checkpoint of the cell cycle. The Cyclin dependent kinase (Cdk) inhibitor, p18, and the S phase transcription factor E2F4 were both found to be significantly down regulated in EKLF null mice and this result was confirmed by real-time PCR. Interestingly, E2F4 knockout mice have a similar phenotype to EKLF knockout mice. Bioinformatic searches of the p18 and E2F4 genes shows that each contains phylogenetically conserved CACC box motifs capable of binding EKLF within longer regions of conservation in promoter and intron regions. The p18 gene contains two conserved CACCC sites upstream of the start of transcription, which are required for EKLF dependent promoter activity in luciferase reporter assays. The transcription factor E2F4 contains a conserved EKLF-binding CACC site within an intron that is closely associated with two conserved GATA1 binding sites. We show by a chromatin immunoprecipitation (ChIP) assays that the E2F4 intron and p18 promoter are occupied by EKLF in vivo. Together, these results suggest that EKLF is likely to directly regulate expression of key cell cycle genes in vivo to drive the switch from proliferation to differentiation of erythrocytes. The loss of EKLF is likely to result in aberrant proliferation and predisposition to leukemia.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1208-1208 ◽  
Author(s):  
Karen Wieland ◽  
Alan B. Cantor

Abstract About ten percent of infants with Down syndrome are born with a transient myeloproliferative disorder (DS-TMD) that spontaneously resolves within the first few months of life. Twenty to thirty percent of these infants subsequently develop acute megakaryoblastic leukemia (DS-AMKL), typically within a year or two following resolution of their DS-TMD. Recent work has shown that both DS-TMD and DS-AMKL cells harbor acquired mutations in the key megakaryocyte transcription factor GATA-1 that lead to the exclusive production of a short GATA-1 isoform (GATA-1s). The mechanism by which GATA-1s acts in DS-TMD/AMKL remains incompletely understood. Mice engineered to produce only GATA-1s exhibit hyperproliferation of fetal liver-derived megakaryocytes, but normal growth of post-natal bone marrow-derived megakaryocytes. This suggests that unique microenvironmental features of fetal liver compared to bone marrow differentially influence the effects of GATA-1s on megakaryopoiesis. In order to further understand the mechanisms involved in these stage-specific effects, we compared gene expression profiles of wild type megakaryocyte progenitors (MkPs) isolated directly from embryonic day 13.5 (e13.5) murine fetal liver (FL) and from adult bone marrow (BM). Cells were FACS sorted based on the immunophenotype lin-sca-1-c-kit+ CD41+ CD150+, which has recently been shown to mark megakaryocyte-selective progenitors. Colony forming assays of the sorted cells revealed 92–100% growth of megakaryocyte colonies in culture medium containing multilineage cytokines (SCF, IL3, IL11, GM-CSF, EPO and TPO), confirming strong enrichment for megakaryocyte lineage cells. RNA from sorted FL and BM MkPs was then used to perform Affymetrix 3′ cDNA expression microarray analysis. Expression of early-stage megakaryocyte factors, such as c-mpl, FOG1, GATA1, Runx-1 and Fli-1 were found in both sets of samples, confirming selection of megakaryocyte progenitor cells. Importantly, we observed a striking up regulation of interferon alpha (IFN alpha) inducible genes belonging to the p200 family in BM MkPs compared to FL MkPs. Gene set enrichment analyses (GSEA) confirmed broad up regulation of the IFN alpha pathway, and as well up regulation of the JAK-STAT pathway in BM MkPs compared to FL MkPs. These findings were validated by quantitative RT-PCR and in situ immunohistochemistry. Given that STAT1 is a direct GATA-1 target gene and a major downstream effector of IFN alpha signaling, we hypothesize that enhanced IFN alpha signaling in the bone marrow may compensate for potential deficiencies of STAT1 during fetal liver megakaryopoiesis in the setting of GATA-1s. Experiments are underway to test this hypothesis.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 387-387
Author(s):  
Morgan Jones ◽  
Michelle L Brinkmeier ◽  
Julien Schira ◽  
Ann Friedman ◽  
Sami Malek ◽  
...  

Abstract Abstract 387 The Trithorax family of epigenetic regulators is intimately linked to normal and malignant hematopoiesis. While substantial work indicates that Mixed lineage leukemia (Mll) is required for hematopoietic stem cell (HSC) homeostasis, the functions of many other Trithorax family members have not been evaluated. We have discovered that the mammalian Trithorax group gene absent, small, or homeotic 1-like (ash1l) is required for the maintenance of adult, but not fetal HSCs. Mice homozygous for a gene trap insertion into the first intron of ash1l (GT/GT) had a ca. 90% reduction in ash1l transcripts. These animals had normal numbers of phenotypically defined fetal liver HSCs (CD150+CD48−Lin−Sca-1hic-Kithi cells), but a 10-fold reduction in adult bone marrow HSCs already apparent by 6 weeks after birth. GT/GT bone marrow HSC depletion began in the first three weeks of life, the period during which HSCs turn off their fetal homeostasis program and enter a state of increased quiescence in the bone marrow. Cell cycle analysis revealed that GT/GT HSCs had an increased cycling fraction with a profound reduction in quiescent HSCs in the G0 phase of the cell cycle. This suggested that ash1l is essential to establish and/or maintain a quiescent population of adult-type HSCs. Furthermore, competitive and non-competitive transplantation assays showed that both fetal liver and adult bone marrow were incapable of providing long-term reconstitution in lethally irradiated recipients, indicating that neither compartment could sustain long-term HSC activity after reaching the recipient's bone marrow. To understand this profound HSC defect, we next sought to analyze the in vivo function of Ash1l. Like MLL, Ash1l is a SET domain-containing histone methyltransferase. Although MLL functions as an H3K4 methyltransferase, the in vivo specificity of the Ash1l SET domain has not been described. Transcriptional analysis of GT/GT HSCs indicated that expression of Hoxa9, a known target of MLL, was reduced by 50%. This suggested that Hoxa9 could be a useful locus at which to evaluate the biochemical activity of Ash1l by chromatin immunoprecipitation-quantitative PCR (ChIP-qPCR). ChIP-qPCR revealed that GT/GT bone marrow cells had reduced H3K36 dimethylation but preserved H3K4 trimethylation at the Hoxa9 locus. This was consistent with previous in vitro analysis of Ash1l SET domain activity and computer predictions showing a high degree of conservation between Ash1l and SET2, an H3K36 methyltransferase. Thus, Ash1l and MLL are both required for Hoxa9 expression although their enzymatic activities differ, suggesting that Ash1l and MLL may act cooperatively at Hoxa9 and other target loci. To evaluate the functional consequences of this interaction, we studied mice deficient for ash1l and lacking Menin, a factor required for proper MLL targeting to target loci. Strikingly, these animals rapidly progressed to hematopoietic failure with a complete obliteration of the hematopoietic stem and progenitor compartment, a phenotype not observed in either genetic background independently. These data indicate that Ash1l and MLL/menin work cooperatively in hematopoiesis. Together, our findings reveal an essential function of ash1l in the maintenance of adult HSCs. Furthermore, they indicate that Ash1l functions in vivo as an H3K36 methyltransferase and suggest that different Trithorax family members can coregulate target genes by providing distinct activating histone marks. Future work will reveal the full extent and mechanisms of these cooperative effects in normal and malignant hematopoiesis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 583-583 ◽  
Author(s):  
Jalal Ahmed ◽  
Yuya Kunisaki ◽  
Miriam Merad ◽  
Paul S. Frenette

Abstract Although most hematopoietic stem cells (HSCs) are quiescent under homeostasis in the adult bone marrow, they are actively proliferating during development. Definitive HSCs, marked by the ability to repopulate a lethally irradiated adult mouse, are first detectable in the aorta-gonad-mesonephros region around E10.5, and then colonize the fetal liver (FL) to expand in this organ until E15 when hematopoietic activity shifts to the fetal bone marrow. The role of the microenvironment, or niche, in the regulation of HSCs in the FL, a site of physiological expansion, is unclear. Fetal liver sinusoidal cells as well as hepatic progenitors have been proposed as sources of supportive signals that drive the exponential expansion of HSCs, however, exact cell type(s) supporting HSC expansion have not been defined. Since stromal cells marked by Nestin-GFP form niches for HSCs in the BM, we have hypothesized that similar cells exist in the FL. Indeed, a rare population of Nestin+ cells (CD31-CD45-Ter119-) comprising 0.02 ± 0.01% of nucleated FL cells was isolated. FL Nestin+ cells express surface markers similar to their BM counterparts, including PDGFRα, CD51, and endoglin. Since BM Nestin+ cells are enriched in mesenchymal stem and progenitor cell (MSPC) activity, we next tested this activity in FL Nestin+ cells. We found that the entire CFU-F forming capacity of the FL was contained within FL Nestin+ cells, further suggesting similarities of these cells between these two hematopoietic organs. In addition, FL Nestin+ cells were enriched for the HSC maintenance genes SCF and CXCL12, suggesting that they may also serve as the HSC niche in the FL. FL Nestin+ cells were localized on the abluminal side of large-bore arteries and expressed the pericyte markers, α-smooth muscle actin and NG2. To investigate the function of the FL Nestin+NG2+ cells in vivo, we generated a genetic depletion mouse model of NG2+ cells (NG2-cre / inducible diphtheria toxin-A (iDTA) transgenic mice). We found that E14.5 NG2-Cre;iDTA mouse embryos developed normally compared to fl-DTA control littermates. The frequency and absolute numbers of HSCs per FL, however, were reduced by about 40% in NG2-Cre;iDTA embryos (control / depleted; 0.01146 ± 0.0008% / 0.00660 ± 0.0008% of nucleated cells, p=0.0015; 606 ± 76 / 377 ± 48 HSCs/FL, p=0.03, , N=6-7 per group) suggesting that Nestin+ cells are required to maintain HSCs/progenitors in vivo. To interrogate further the relationship between FL Nestin+ cells and HSCs, we adopted the re-aggregate organ culture assay (Sheridan, Genesis 2009). Sorted lineage- FL hematopoietic progenitor cells and FL parenchymal cells either with or without sorted Nestin+ cells were re-aggregated and cultured for 7 days in serum-free, cytokine-free, media. After 7 days of culture, we found that CD150+ CD48− CD41− Lineage− HSCs were maintained in re-aggregates containing Nestin+ cells, but not when Nestin+ cells were absent. To confirm that FL Nestin+ cells were essential to maintain functional HSCs, re-aggregated cells cultured for 7 days were transplanted together with competitor bone marrow into lethally irradiated mice. We found that the contribution to the peripheral blood at 8 weeks post-transplant was only observed in the re-aggregated group containing Nestin+ cells. These preliminary data indicate that factors derived from FL Nestin+ cells are required to maintain HSCs in re-aggregate cultures. These findings suggest that HSCs inhabit similar microenvironments in temporally and spatially distinct hematopoietic organs. Further studies on differences between Nestin+ cells in these tissues may shed light on the mechanisms that determine the finely tuned quiescence, self-renewal and differentiation of HSCs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 735-735
Author(s):  
Per Holmfeldt ◽  
Pardieck Jennifer ◽  
Shannon McKinney-Freeman

Abstract Hematopoietic stem cells (HSCs) are responsible for life-long maintenance of hematopoiesis. HSC transplantation represents one of the most heavily exploited cell based therapies, routinely used to treat a myriad of life threating disorders, such as leukemia and bone marrow failure. Identifying the molecular pathways that regulate HSC engraftment is crucial to further improving outcomes in patients that rely on HSC transplantation as a curative therapy. By examining the global gene expression profiles of highly purified HSC (Lineage-Sca-1+c-Kit+CD150+CD48-), we recently identified the following members of the Nfi gene family of transcription factors as highly expressed by HSC (McKinney-Freeman et al., Cell Stem Cell, 2012): Nfix, Nfia, and Nfic. These data suggest that Nfi genes may play a novel role in regulating HSC function. To test this hypothesis, HSCs were enriched from adult bone marrow (Lineage-, c-kit+, Sca-1+ (LSK) cells) and then transduced, individually, with lentiviruses carrying shRNAs targeting each Nfi gene. Twenty-four hours post-transduction, cells were injected into lethally irradiated mice along with untransduced bone marrow LSK competitor cells congenic at the CD45 allele. The peripheral blood of recipient mice was then analyzed periodically over 16 weeks for engraftment of the Nfi-depleted cells. Although shRNA mediated knockdown of Nfi gene expression had no effect on the in vitro cell growth or viability of LSK cells, Nfi-depleted HSCs displayed a significant loss of short- and long-term in vivo hematopoietic repopulating activity. This was true for Nfia-, Nfic-, and Nfix-deficient HSC. While Nfia and Nfic are only expressed by bone marrow HSC, Nfix is highly expressed by both bone marrow and fetal liver HSC. When Nfix was depleted by shRNAs from LSK cells purified from E14.5 fetal liver, a similar loss in competitive repopulating potential was seen. Lineage analysis of peripheral blood of recipients showed no significant differences in the distribution of the major blood lineages derived from LSK cells transduced with Nfi-specific shRNAs compared to controls. When the bone marrow of recipients transplanted with Nfix- depleted cells was examined 4 and 16 weeks post-transplant, a general loss of all hematopoietic stem- and progenitor compartments examined was seen relative to control. Thus, the observed decrease in repopulating activity occurs at the level of HSCs and multipotent progenitors. To confirm an essential role for an Nfi gene family member in the regulation of HSC engraftment post-transplant, LSK cells were purified from Nfix fl/fl mice, transduced with lentiviral Cre recombinase and subsequently introduced into lethally irradiated recipients alongside congenic competitor cells. Like LSK transduced with Nfix-specific shRNAs, Nfix-/- LSK cells failed to repopulate the peripheral blood of recipient mice as efficiently as control and similar trends were detected in all stem- and progenitor cell populations examined. Time-course experiments immediately following transplantation revealed that Nfix-depleted LSK cells establish themselves in the marrow of recipient mice as efficiently as control at 5 days post-transplant, but thereafter exhausted rapidly. Examination 10 days post-transplant revealed a 5-fold increase in apoptosis specifically in the LSK compartment, but not in its differentiated progeny, in recipients transplanted with Nfix-depleted LSK cells compared to control. The increase in apoptosis was not associated with any apparent change in the cell cycle status of the LSK cells. These data suggest that Nfi genes are necessary for the survival of HSC post-transplantation. In an effort to identify the molecular pathways regulated by Nfi genes in HSC, we acquired the global gene expression profiles of Nfix-depleted HSC. In agreement with our observation that Nfix-deficient HSC displays elevated levels of apoptosis following transplantation in vivo, we observed a significant decrease in multiple genes known to be important for HSC survival, such as Erg, Mecom and Mpl, in Nfix-depleted HSC. In summary, we have for the first time established a role for the Nfi gene family in HSC biology, as evident by a decrease in bone marrow repopulating activity in Nfi-depleted HSCs. By dissecting the precise role of Nfi genes in HSC biology, we will glean insights that could improve our understanding of graft failure in clinical bone marrow transplantations. Disclosures: No relevant conflicts of interest to declare.


2005 ◽  
Vol 83 (4) ◽  
pp. 535-547 ◽  
Author(s):  
Gareth N Corry ◽  
D Alan Underhill

To date, the majority of the research regarding eukaryotic transcription factors has focused on characterizing their function primarily through in vitro methods. These studies have revealed that transcription factors are essentially modular structures, containing separate regions that participate in such activities as DNA binding, protein–protein interaction, and transcriptional activation or repression. To fully comprehend the behavior of a given transcription factor, however, these domains must be analyzed in the context of the entire protein, and in certain cases the context of a multiprotein complex. Furthermore, it must be appreciated that transcription factors function in the nucleus, where they must contend with a variety of factors, including the nuclear architecture, chromatin domains, chromosome territories, and cell-cycle-associated processes. Recent examinations of transcription factors in the nucleus have clarified the behavior of these proteins in vivo and have increased our understanding of how gene expression is regulated in eukaryotes. Here, we review the current knowledge regarding sequence-specific transcription factor compartmentalization within the nucleus and discuss its impact on the regulation of such processes as activation or repression of gene expression and interaction with coregulatory factors.Key words: transcription, subnuclear localization, chromatin, gene expression, nuclear architecture.


Sign in / Sign up

Export Citation Format

Share Document