scholarly journals Transcriptional network orchestrating regional patterning of cortical progenitors

2021 ◽  
Vol 118 (51) ◽  
pp. e2024795118
Author(s):  
Athéna R. Ypsilanti ◽  
Kartik Pattabiraman ◽  
Rinaldo Catta-Preta ◽  
Olga Golonzhka ◽  
Susan Lindtner ◽  
...  

We uncovered a transcription factor (TF) network that regulates cortical regional patterning in radial glial stem cells. Screening the expression of hundreds of TFs in the developing mouse cortex identified 38 TFs that are expressed in gradients in the ventricular zone (VZ). We tested whether their cortical expression was altered in mutant mice with known patterning defects (Emx2, Nr2f1, and Pax6), which enabled us to define a cortical regionalization TF network (CRTFN). To identify genomic programming underlying this network, we performed TF ChIP-seq and chromatin-looping conformation to identify enhancer–gene interactions. To map enhancers involved in regional patterning of cortical progenitors, we performed assays for epigenomic marks and DNA accessibility in VZ cells purified from wild-type and patterning mutant mice. This integrated approach has identified a CRTFN and VZ enhancers involved in cortical regional patterning in the mouse.

2020 ◽  
Author(s):  
Athéna R Ypsilanti ◽  
Kartik Pattabiraman ◽  
Rinaldo Catta-Preta ◽  
Olga Golonzhka ◽  
Susan Lindtner ◽  
...  

SUMMARYWe uncovered a transcription factor (TF) network that regulates cortical regional patterning. Screening the expression of hundreds of TFs in the developing mouse cortex identified 38 TFs that are expressed in gradients in the ventricular zone (VZ). We tested whether their cortical expression was altered in mutant mice with known patterning defects (Emx2, Nr2f1 and Pax6), which enabled us to define a cortical regionalization TF network (CRTFN). To identify genomic programming underlying this network, we performed TF ChIP-seq and chromatin-looping conformation to identify enhancer-gene interactions. To map enhancers involved in regional patterning of cortical progenitors, we performed assays for epigenomic marks and DNA accessibility in VZ cells purified from wild-type and patterning mutant mice. This integrated approach has identified a CRTFN and VZ enhancers involved in cortical regional patterning.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4195-4195
Author(s):  
Elena Levantini ◽  
Francesca Bertolotti ◽  
Francesco Cerisoli ◽  
Anna L. Ferri ◽  
Elisa Brescia ◽  
...  

Abstract Several genes encoding transcription factors of different families have been implicated in the development and differentiation of multiple cell systems. The Sry-type high-mobility-group box 2 gene (Sox2) encodes a transcription factor that is expressed in very early cells such as embryonic stem cells and neural stem cells, where it plays important functional roles (Genes and Dev.17:126, 2003; Development131:3805, 2004). To investigate whether Sox2 plays a role also in blood cell production, we first analyzed its expression in murine hematopoietic cells. Results indicate that the gene is transcriptionally active at low levels in primitive progenitors. Furthermore, in order to address the functional implication of Sox2 in hematopoiesis we analyzed mature and precursor cells in mutant mice compound heterozygotes for a null Sox2 allele and for the deletion of a Sox2 5′ enhancer, as the complete inactivation of the gene in homozygosis is embryonic lethal. At the peripheral blood level we did not detect significant variations in the mutants. However analysis of bone marrow precursors in clonogenic assays showed that Sox2 knock-down mice exhibited a significant increase in the number of multipotent precursors, as compared to wild type animals. Moreover, bone marrow cells of wild type and mutant mice were analyzed for the expression of a panel of regulatory genes involved in the control of different somatic stem cells. Preliminary evidence suggests that some of these genes are modulated in the mutant cells. These observations support the view that Sox2 plays a role at early stages of blood cell production, providing further evidence that common molecular mechanisms may be involved in the regulation of several different types of multipotent cells.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1492-1492
Author(s):  
Akil Merchant ◽  
Anju Singh ◽  
Giselle Joseph ◽  
Qiuju Wang ◽  
Ping Zhang ◽  
...  

Abstract Abstract 1492 Poster Board I-515 Previous studies have established an important role for reactive oxygen species (ROS) in regulating the function and life-span of hematopoietic stem cells (HSC). Nuclear factor erythroid-2–related factor 2 (Nrf2) is a redox-sensitive transcription factor that regulates cellular responses to ROS and detoxification pathways implicated in chemoresistance, however, its role in normal stem cells is unknown. We analyzed Nrf2null mice and found increased total bone marrow cellularity, cKit+Sca1+Lin− (KSL) stem-progenitor cells, and long-term quiescent HSC (CD34−KSL) compared to wild type mice (p<0.05). Transplantation of equal numbers of KSL cells from Nrf2wt and Nrf2null resulted in a five-fold decrease in peripheral blood chimerism from Nrf2null derived cells at 16 weeks (15% wild type vs. 3% null, p<0.05). Unlike other models of deficiencies in genes associated with ROS handling, such as ATM or the FoxO family of transcription factors, basal ROS levels were not elevated in Nrf2null HSC. However, Nrf2null bone marrow cells demonstrated increased sensitivity to induced oxidative stress and in vitro treatment with H2O2 resulted in a 2 fold decrease in colony formation in methylcellulose. We also examined the in vivo sensitivity of Nrf2null cells to oxidative stress by irradiating (400 rads) stably chimeric mice 20 weeks following transplantation with either Nrf2wt or Nrf2null HSC. Mice receiving Nrf2null HSC demonstrated a 50% decrease in peripheral blood chimerism at 4 months following radiation compared to no change in Nrf2wt recipients (p<0.05) confirming that loss of Nrf2 leads to increased sensitivity to oxidative stress. Microarray gene expression analysis from Nrf2wt and Nrf2null mice revealed down regulation of the G-CSF cytokine receptor in Nrf2null HSC and suggested that defective cytokine signaling may contribute to the HSC dysfunction seen in Nrf2null bone marrow cells. To test this hypothesis, we attempted to rescue the function of Nrf2null HSC by treating mice with exogenous G-CSF. Nrf2wt and Nrf2null mice were treated with one week of daily G-CSF and then HSC were harvested and transplanted. In contrast to the defects in engraftment of untreated Nrf2null HSC, there was no significant difference in peripheral blood chimerism following transplantation of G-CSF treated Nrf2wt or Nrf2null HSC, thus demonstrating that G-CSF treatment could rescue the HSC defect in mutant mice. In conclusion, the Nrf2 transcription factor appears to be a novel and essential regulator of normal HSC function through the modulation of oxidative stress response and cytokine signaling. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 545-545
Author(s):  
Laura Belver ◽  
Alexander Y Yang ◽  
Daniel Herranz ◽  
Aidan Quinn ◽  
Francesco G Brundu ◽  
...  

Abstract Long range enhancers play critical roles in the control of gene expression during development and have emerged as key regulators of lineage commitment and oncogenic programs in hematopoiesis and leukemia. The MYC oncogene is dynamically regulated in the hematopoietic system under the control of a network of clustered distal enhancers, which provide modular regulation of MYC expression during lymphoid and myeloid development. In thymocyte development MYC transcription critically depends on the activity of N-Me, a distinct T-cell specific enhancer controlled by NOTCH1 signaling and located 1.4 Mb telomeric to the MYC transcription start site. Yet, the specific mechanisms governing N-Me enhancer activity and lineage specific control of MYC expression remain rudimentarily understood. Analysis of chromatin looping by 4C and chromatin accessibility by ATACseq revealed an unanticipated high density of chromatin contacts between N-Me and additional regulatory elements in the Myc locus and showed a distinct pattern of N-Me chromatin accessibility -opening as progenitors mature into T cell committed CD4 CD8 double negative (DN) 2b cells and returning to a closed configuration in CD4 CD8 double positive (DP) thymocytes-. To explore potential regulators of N-Me activity we performed Mass Spectrometry proteomic profiling of N-Me binding proteins and ChIPseq analyses identifying numerous factors involved in hematopoietic and lymphoid development (ERG, ETS1, GATA3, RUNX1, TCF3 and TCF12) and transcription factor oncogenes with prominent roles in the pathogenesis of T-ALL (HOXA9, MYB, MYC, LMO1, LMO2, TAL1 and TLX1). Moreover, phylogenetic footprinting analyses across vertebrate species identified two ultraconserved elements matching GATA factor binding motifs (GS1 and GS2). To test the functionality of these elements we introduced targeted mutations in the N-Me sequence at these sites using CRISPR/CAS9 directed mutagenesis. Mice homozygous for combined N-Me GS1 and GS2 mutations (GS1+2mut) revealed a marked defect in thymus cellularity with characteristic accumulation of DN and intermediate single positive (ISP) thymocytes and decreased numbers of more mature populations. Mechanistically, immunohistochemical, flow cytometry and single cell RNaseq analyses revealed decreased Myc protein levels in thymocyte poulations of GS1+2 mutant animals. In this context, we hypothesized that GATA3, a prominent N-Me binding transcription factor in our ChIP and proteomic analyses critically implicated in T-cell commitment, could play a major role in N-Me regulation via interaction with the GS1 and GS2 N-Me GATA sites. Consistent with this hypothesis analysis of Gata3 ChIPs from heterozygous GS1+2 mutant mice recovered only the N-Me wild type sequence, formally demonstrating the strict requirement of these sites for N-Me Gata3 binding. Mechanistically, ATACseq analysis revealed a marked reduction in chromatin accessibility and nucleosome invasion in thymocytes from GS1+2 mutant mice in support of a critical pioneering activity for GATA3 in the control of N-Me activity. Finally, given the important role of NOTCH1 induced MYC upregulation in the pathogenesis of T-ALL, we hypothesized that disruption of N-Me activity via targeted mutation of N-Me GATA sites could effectively impair the development of NOTCH1-driven T-ALL in N-Me GS1+2 mutant mice. To test this possibility we infected hematopoietic progenitors from N-Me wild type and N-Me GS1+2 homozygous mice with retroviruses driving the expression of an oncogenic constitutively active form of NOTCH1 (DE-NOTCH1) and transplanted them into sublethally irradiated recipients. In these experiments, mice transplanted with DE-NOTCH1 infected N-Me wild type cells developed overt T-ALL 6 weeks postransplant with 100% penetrance. In contrast, mice transplanted with DE-NOTCH1-expressing N-Me GS1+2 homozygous cells showed complete protection from NOTCH1 induced T-ALL (P <0.001). In all these results identify GATA3 binding to the N-Me enhancer as a critical driver of nucleosome eviction and enhancer activation strictly required for thymocyte development and NOTCH1-induced T-cell transformation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 642-642
Author(s):  
Minghui Tang ◽  
Zhenbiao Xia ◽  
Shubin Zhang ◽  
Shanshan Zhang ◽  
Xudong Wei ◽  
...  

Abstract TGFβ1-activated kinase 1 (TAK1), a member of the MAPKKK family, is a key mediator of stress and proinflammatory signals. TAK1 can be activated by inflammation-mediating cytokines, including tumor necrosis factor-α (TNF-α and interleukin-1b (IL-1β), as well as by T- and B- cell receptors (TCR/BCR), and Toll-like receptors (TLRs) signals. Activated TAK1 induces the nuclear localization of NF-kB and the activation of JNK/AP1 by stimulating IKKβ and MKK3/MKK6 phosphorylation respectively. TAK1 has been found to play an important role in inflammation, immunity, T- and B-cell activation, and epithelial cell survival. The TAK1−/ − phenotype is lethal in mice at the early embryonic stage. We found higher levels of TAK1 expression and activity in hematopoietic stem cells and progenitors (HSC/Ps), and reduced expression and activity in differentiated mature hematopoietic cells. To study the role of TAK1 in bone marrow hematopoiesis, we generated inducible-TAK1 knockout mice by crossing TAK1loxp mice with Mx1Cre mice, the latter being an interferon-inducible Cre mouse line. After injection of polyI:C to induce the knockout, we found that all the TAK1 knockout mice died within 8 to 10 days after the first polyI:C injection, showing severe hematopoietic and other defects; heterozygotes were phenotypically comparable to wild-type control animals. The TAK1 deletion in these mice resulted in ablation of bone marrow hematopoiesis due to the loss of C-Kit+ HSC/Ps. Annexin-V staining showed a 3-fold increase in apoptosis in the C-Kit+ HSC/Ps from TAK1 mutant mice compared to those from littermate control mice. Almost all of the mutant animals showed intestinal bleeding as well as other hemorrhaging due to the significant reductions in platelet counts. In reciprocal bone marrow transplantation experiments, we found that the TAK1-mutant bone marrow microenvironment was able to support the growth and function of wild-type HSC/Ps, while HSC/Ps from TAK1−/ − mice failed to grow within the wild-type bone marrow microenvironment. These observations suggest that the bone marrow ablation phenotype which develops in TAK1-mutant mice is the result of intrinsic defects in HSC/P’s. We propose that TAK1-mutant HSC/Ps might mediate a survival signal for HSC/Ps stimulated by hematopoietic growth factors and cytokines, such as stem cell factor (SCF). The details of possible mechanisms by which this phenomenon might occur is currently under active investigation by our group.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1649-1649 ◽  
Author(s):  
Ayana Kon ◽  
Satoshi Yamazaki ◽  
Yasunori Ota ◽  
Keisuke Kataoka ◽  
Yusuke Shiozawa ◽  
...  

Abstract Recent genetic studies have revealed frequent and specific pathway mutations involving multiple components of the RNA splicing machinery in myelodysplasia. Among these, SRSF2 mutations are more prevalent in CMML subtype and are associated with poor prognosis. Mutations showed a prominent hotspot involving proline 95, causing either P95H, P95L, or P95 conversion. Comprehensive analysis in our large cohort of MDS revealed that SRSF2 mutations showed a significant trend to coexist with TET2, STAG2, ASXL1 and RUNX1 mutations, while being mutually exclusive with EZH2 mutations. On the other hand, the molecular mechanism by which SRSF2 mutations lead to myelodysplasia remains largely unknown.  To elucidate the role of SRSF2 mutations in the development of myelodysplasia, we generated a heterozygous conditional knock-in mouse model of Srsf2 P95H mutation and crossed them with Vav1-Cre transgenic mice. Srsf2 P95H mutant mice exhibited macrocytic anemia, otherwise no significant changes in total peripheral blood (PB) cell counts compared to wild-type mice at 8-15 weeks after birth. There was no significant difference in lineage composition as well as blood cell morphology between wild-type and mutant mice in both bone marrow (BM) and PB. Flow cytometry of BM cells showed significant decrease of the number of hematopoietic stem cells (HSCs) and multipotent progenitor cells defined as Lin-Sca-1+Kit+ (LSK) fractions in Srsf2 P95H mice compared to wild-type mice. On the other hand, there were no significant differences in the number of more differentiated progenitor cells including common myeloid progenitors (CMPs), granulocyte/macrophage lineage-restricted progenitors (GMPs), megakaryocyte/erythrocyte lineage-restricted progenitors (MEPs), and common lymphoid progenitors (CLPs) between Srsf2 P95H and wild-type mice. These observations suggested that heterozygous Srsf2 mutation led to deregulation of hematopoietic stem cells, which however, is not sufficient for the development of MDS.  We next performed noncompetitive transplantation experiments to assess the cell intrinsic effects of Srsf2 P95H mutations. In PB, decreased white blood cell counts and progressive anemia were observed in mutant mice, which were evident as early as one month after transplantation. Cytological analysis of PB revealed morphological abnormalities in mice reconstituted with Srsf2 mutated cells, including hypersegmentation in neutrophils and dysplasia in the erythroid lineage. Srsf2 mutant-reconstituted mice showed normo-to-hypercellular marrow, where abnormal megakaryocyte distribution adjacent to trabecular bone and erythroid dysplasia was observed. Flow cytometrical analysis revealed decreased numbers of HSCs, LSK fractions and CMPs, whereas there was no significant change in the number of MEPs, GMPs and CLPs in BM. The BM erythroid progenitors were decreased in mutant-reconstituted mice, whereas the mutant mice showed splenic erythropoiesis with increased erythroid progenitors, suggesting the presence of extramedullary hematopoiesis, which was not seen in wild-type Srsf2 transduced mice. These observations suggested that the Srsf2 mutation led to ineffective hematopoiesis and morphological abnormalities, which seemed to recapitulate the phenotype of MDS.  Subsequently, we assessed the reconstitution capacity of whole BM cells from Srsf2 mutant mice in competitive transplantation experiments. The donor chimerism of Srsf2 P95H-derived cells in PB was significantly lower than that of wild-type cells. At 4 months post transplantation, the chimerism of Srsf2 P95H-derived cells was remarkably lower than that of wild-type cells in the fractions of HSCs, MPPs, CMPs, MEPs, GMPs and CLPs in BM. Furthermore, the reduced donor chimerism for Srsf2 P95H mutants was recapitulated in secondary transplantation experiments.  In summary, our results demonstrated that heterozygous P95H mutation of Srsf2 led to deregulation of hematopoietic stem cells that was evident from reduced competitive repopulation and impaired hematopoietic differentiation. Whereas mice reconstituted with Srsf2 mutant BM cells developed MDS-like phenotype in non-competitive transplantation setting, Srsf2 mutation by itself does not seem to be sufficient to develop MDS without transplantation, raising the possibility that an additional genetic and/or epigenetic events was required for overt MDS phenotype. Disclosures No relevant conflicts of interest to declare.


Development ◽  
1997 ◽  
Vol 124 (24) ◽  
pp. 5087-5096 ◽  
Author(s):  
D. Caric ◽  
D. Gooday ◽  
R.E. Hill ◽  
S.K. McConnell ◽  
D.J. Price

The cerebral cortex forms by the orderly migration and subsequent differentiation of neuronal precursors generated in the proliferative ventricular zone. We studied the role of the transcription factor Pax-6, which is expressed in the ventricular zone, in cortical development. Embryos homozygous for a mutation of Pax-6 (Small eye; Sey) had abnormalities suggesting defective migration of late-born cortical precursors. When late-born Sey/Sey precursors were transplanted into wild-type embryonic rat cortex, they showed similar integrative, migrational and differentiative abilities to those of transplanted wild-type mouse precursors. These results suggest that postmitotic cortical cells do not need Pax-6 to acquire the capacity to migrate and differentiate, but that Pax-6 generates a cortical environment that permits later-born precursors to express their full developmental potential.


2018 ◽  
Author(s):  
Pietro Mesirca ◽  
Daria Mamaeva ◽  
Isabelle Bidaud ◽  
Matthias Baudot ◽  
Romain Davaze ◽  
...  

AbstractCurrent treatments for heart automaticity disorders still lack a safe and efficient source of stem cells to restore normal biological pacemaking. Since adult Muscle-Derived Stem Cells (MDSC) show multi-lineage differentiation in vitro including into spontaneously beating cardiomyocytes, we questioned whether they could effectively differentiate into cardiac pacemakers, a specific population of cardiomyocytes producing electrical impulses in the sino-atrial node (SAN) of adult heart. We show here that beating cardiomyocytes, differentiated from MDSC in vitro, exhibit typical characteristics of cardiac pacemakers: expression of markers of the SAN lineage Hcn4, Tbx3 and Islet1, as well as spontaneous calcium transients and hyperpolarization-activated “funny” current and L-type Cav1.3 channels. Pacemaker-like myocytes differentiated in vitro from Cav1.3-deficient mouse stem cells produced slower rate of spontaneous Ca2+ transients, consistent with the reduced activity of native pacemakers in mutant mice. In vivo, undifferentiated wild type MDSC migrated and homed with increased engraftment to the SAN of bradycardic mutant Cav1.3-/- within 2-3 days after systemic I.P. injection. The increased homing of MDSCs corresponded to increased levels of the chemokine SDF1 and its receptor CXCR4 in mutant SAN tissue and was ensued by differentiation of MDSCs into Cav1.3-expressing pacemaker-like myocytes within 10 days and a significant improvement of the heart rate maintained for up to 40 days. Optical mapping and immunofluorescence analyses performed after 40 days on SAN tissue from transplanted wild type and mutant mice showed MDSCs integrated as pacemaking cells both electrically and functionally within recipient mouse SAN. These findings identify MDSCs as directly transplantable stem cells that efficiently home, differentiate and improve heart rhythm in mouse models of congenital bradycardia.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3080-3080
Author(s):  
Ayana Kon ◽  
Yasuhito Nannya ◽  
Masahiro Nakagawa ◽  
Keisuke Kataoka ◽  
Masashi Sanada ◽  
...  

Abstract Recent genetic studies have revealed frequent and specific pathway mutations involving multiple components of the RNA splicing machinery in myelodysplasia. Among these, U2AF1 mutations are more prevalent in MDS without increased ring sideroblasts and AML with myelodysplasia-related changes and are associated with a poor prognosis. Also found in approximately 4% of lung adenocarcinoma, U2AF1 mutations exclusively involved two highly conserved amino acid positions (S34 or Q157) within the amino- and the carboxyl-terminal zinc finger motifs flanking the U2AF homology motif (UHM) domain. Comprehensive analysis in a large cohort of MDS showed that U2AF1 mutations showed a significant trend to coexist with ASXL1. The molecular mechanism by which U2AF1 mutations lead to myelodysplasia have not fully been elucidated. To elucidate the role of U2AF1 mutations in the development of myelodysplasia, we generated heterozygous conditional knock-in mice for the U2af1 S34F mutation, which were crossed them with Vav1-Cre transgenic mice. Vav1-Cre mediated U2af1 S34F knock-in mice exhibited severe leukopenia and macrocytic anemia at 8-20 weeks after birth. Although there was no significant difference in blood cell morphology between wild-type and mutant mice in bone marrow (BM) and peripheral blood (PB) cells, there was strong myeloid skewing in lineage composition both in U2af1 mutant BM and PB cells compared to wild-type controls. Flow cytometry of U2af1 S34F BM cells showed a significant decrease in the number of hematopoietic stem cells (HSCs), common myeloid progenitors (CMPs) and megakaryocyte/erythrocyte lineage-restricted progenitors (MEPs), and an increase of the granulocyte/macrophage lineage-restricted progenitors (GMPs) compared to wild-type BM cells. These observations suggest that heterozygous U2af1 mutation leads to differentiation defects of HSCs, which however, is not sufficient for the development of MDS. We next assessed the phenotype of U2af1-mutated BM cells in transplantation settings to evaluate the effect of increasing replicative stress, which has been shown to substantially affect the behavior of normal and abnormal stem cells. In PB, progressive leukopenia, macrocytic anemia and decreased platelet counts were observed in mutant mice in transplantation settings. Surprisingly, all of the U2af1 mutant-transplanted mice died within two months after transplantation due to severe bone marrow failure. Cytological analysis of BM cells revealed morphological abnormalities in U2af1 mutant-transplanted mice, including hypersegmentation in neutrophils and erythroid dysplasia. Flow cytometrical analysis revealed decreased numbers of HSCs, CMPs and MEPs, and increased number of GMPs. These observations suggest that the U2af1 mutation leads to ineffective hematopoiesis and morphological abnormalities, which seems to recapitulate the phenotype of MDS in transplantation settings. Subsequently, we assessed the reconstitution capacity of whole BM cells from U2af1 mutant mice in competitive transplantation experiments. The donor chimerism of U2af1 S34F-derived cells in PB was remarkably reduced compared to that of wild-type cells. At four months post transplantation, the chimerism of U2af1 S34F-derived cells was markedly lower than that of wild-type cells in the fractions of HSCs, CMPs, MEPs, GMPs and common lymphoid progenitors (CLPs) in BM. RNA sequencing analysis of HSCs defined as Kit+Sca-1+Linlow (KSL) cells and CMPs from the mutant mice showed significant changes in alternative splicing and expression levels in many genes, including several potential targets implicated in the pathogenesis of hematopoietic malignancies. In summary, our results demonstrated that heterozygous U2af1 S34F mutation led to impaired HSC functions that was evident from reduced competitive repopulation and deregulated hematopoietic differentiation, which were augmented in transplantation settings. Our mice model provides a valuable tool to understand the molecular pathogenesis of U2af1-mutated myeloid neoplasms. Disclosures Nakagawa: Sumitomo Dainippon Pharma Co., Ltd.: Research Funding.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2463-2463
Author(s):  
Erica Bresciani ◽  
Blake Carrington ◽  
Erika M Kwon Kim ◽  
Kai Yu ◽  
Kevin Bishop ◽  
...  

The current notion about how hematopoietic stem cells (HSCs) are generated identifies the transcription factor RUNX1 as an essential factor for the emergence of definitive hematopoietic stem cells (HSCs) from the hemogenic endothelium. Consequently, Runx1knockout mice fail to develop definitive hematopoiesis and lack all definitive blood lineages and cannot survive past embryonic day 12. However, even though zebrafish with arunx1stop codon mutation (runx1W84X/W84X) presented defects in definitive hematopoiesis during embryogenesis, runx1W84X/W84Xembryos could develop to fertile adults with blood cells of multi-lineages, raising the possibility that HSCs can emerge without RUNX1. In order to determine if a RUNX1-independent mechanism can support the generation of HSCs we have generated three new zebrafish runx1-/- with engineered deletions of the runx1gene using TALEN and CRISPR-Cas9. Our analysis shows that all three mutants have identical phenotypei.e., failure to develop definitive hematopoiesis during early embryogenesis, with later reemergence of hematopoietic cells and survivalof therunx1 mutants to adulthood, further confirming the existence of a RUNX1-independent mechanism for the emergence of HSCs. In the absence of a functional runx1, a cd41-GFP+population of hematopoietic precursors can still be detected in the aorta-gonad-mesonephros (AGM) region and in the hematopoietic tissues of the mutant embryos. Single cell RNA sequencing of the wild type and mutant HSC/HSPC at embryonic and larval stages confirmed the presence of a population of runx1- /-cd41:GFPlow cells expressing HSC signature genes at 2.5 days post fertilization. At larval stages the runx1-/-HSCs maintain their ability to generate erythroid and myeloid lineage progenitors but they present a different expression profile compared to the wild type. In order to uncover the compensatory mechanism that drives the repopulation of the hematopoietic compartment in the absence of runx1we identified the molecular signatures that separate the runx1-/-HSC/HSPCs from the wild type and subsequently focused our attention on the transcription factors differentially expressed in the runx1-/-HSC/HSPCs. Our analysis shows that the master transcription factor gata2b is strongly upregulated in the runx1- /-HSCs during the recovery of hematopoiesis and it is also upregulated in the kidney marrow of the surviving runx1-/-adults. Given the key role of GATA2 in the HSC development and maintenance in both mouse and zebrafish, gata2b represented a strong candidate gene with the potential ability to drive the rescue of the runx1-/-phenotype. Indeed, a loss of function mutation or knock-down of gata2b can significantly reduce or abolish the survivability of the runx1-/-fish, indicating that gata2bis responsible for rescuing hematopoiesis in the runx1 mutant fish. Overall our results show that even though runx1 is necessary for the normal emergence of definitive HSCs in the embryos, in the absence of runx1the transcription factor gata2 is able to support definitive hematopoiesis that is sufficient for the embryos to develop to functional adults in the zebrafish. The current notion about how hematopoietic stem cells (HSCs) are generated identifies the transcription factor RUNX1 as an essential factor for the emergence of definitive hematopoietic stem cells (HSCs) from the hemogenic endothelium. Consequently, Runx1knockout mice fail to develop definitive hematopoiesis and lack all definitive blood lineages and cannot survive past embryonic day 12. However, even though zebrafish with arunx1stop codon mutation (runx1W84X/W84X) presented defects in definitive hematopoiesis during embryogenesis, runx1W84X/W84Xembryos could develop to fertile adults with blood cells of multi-lineages, raising the possibility that HSCs can emerge without RUNX1. In order to determine if a RUNX1-independent mechanism can support the generation of HSCs we have generated three new zebrafish runx1-/- with engineered deletions of the runx1gene using TALEN and CRISPR-Cas9. Our analysis shows that all three mutants have identical phenotypei.e., failure to develop definitive hematopoiesis during early embryogenesis, with later reemergence of hematopoietic cells and survivalof therunx1 mutants to adulthood, further confirming the existence of a RUNX1-independent mechanism for the emergence of HSCs. In the absence of a functional runx1, a cd41-GFP+population of hematopoietic precursors can still be detected in the aorta-gonad-mesonephros (AGM) region and in the hematopoietic tissues of the mutant embryos. Single cell RNA sequencing of the wild type and mutant HSC/HSPC at embryonic and larval stages confirmed the presence of a population of runx1- /-cd41:GFPlow cells expressing HSC signature genes at 2.5 days post fertilization. At larval stages the runx1-/-HSCs maintain their ability to generate erythroid and myeloid lineage progenitors but they present a different expression profile compared to the wild type. In order to uncover the compensatory mechanism that drives the repopulation of the hematopoietic compartment in the absence of runx1we identified the molecular signatures that separate the runx1-/-HSC/HSPCs from the wild type and subsequently focused our attention on the transcription factors differentially expressed in the runx1-/-HSC/HSPCs. Our analysis shows that the master transcription factor gata2b is strongly upregulated in the runx1- /-HSCs during the recovery of hematopoiesis and it is also upregulated in the kidney marrow of the surviving runx1-/-adults. Given the key role of GATA2 in the HSC development and maintenance in both mouse and zebrafish, gata2b represented a strong candidate gene with the potential ability to drive the rescue of the runx1-/-phenotype. Indeed, a loss of function mutation or knock-down of gata2b can significantly reduce or abolish the survivability of the runx1-/-fish, indicating that gata2bis responsible for rescuing hematopoiesis in the runx1 mutant fish. Overall our results show that even though runx1 is necessary for the normal emergence of definitive HSCs in the embryos, in the absence of runx1the transcription factor gata2 is able to support definitive hematopoiesis that is sufficient for the embryos to develop to functional adults in the zebrafish. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document