Requirement for Ssbp2 in Hematopoietic Stem Cell Maintenance and Stress Response June Li, Yasuhiro Kurasawa, Yang Wang, Karen Clise-Dwyer, Sherry A. Klumpp, Hong Liang, Ramesh C. This information is current as Tailor, Aaron C. Raymond, Zeev Estrov, Stephen J. Brandt, of September 26, 2021. Richard E. Davis, Patrick Zweidler-McKay, Hesham M. Amin and Lalitha Nagarajan J Immunol published online 19 September 2014

http://www.jimmunol.org/content/early/2014/09/19/jimmun Downloaded from ol.1300337

Supplementary http://www.jimmunol.org/content/suppl/2014/09/19/jimmunol.130033 Material 7.DCSupplemental http://www.jimmunol.org/

Why The JI? Submit online.

• Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists

by guest on September 26, 2021 • Fast Publication! 4 weeks from acceptance to publication

*average

Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2014 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published September 19, 2014, doi:10.4049/jimmunol.1300337 The Journal of Immunology

Requirement for Ssbp2 in Hematopoietic Stem Cell Maintenance and Stress Response

June Li,*,1 Yasuhiro Kurasawa,*,1 Yang Wang,* Karen Clise-Dwyer,† Sherry A. Klumpp,‡ Hong Liang,* Ramesh C. Tailor,x Aaron C. Raymond,*,{ Zeev Estrov,‖ Stephen J. Brandt,#,**,††,‡‡ Richard E. Davis,xx Patrick Zweidler-McKay,{{,2 Hesham M. Amin,‖‖,2 and Lalitha Nagarajan*,{,‖,##

Transcriptional mechanisms governing hematopoietic stem cell (HSC) quiescence, self-renewal, and differentiation are not fully understood. Sequence-specific ssDNA–binding 2 (SSBP2) is a candidate acute myelogenous leukemia (AML) suppressor located at 5q14. SSBP2 binds the transcriptional adaptor protein Lim domain–binding protein 1 (LDB1) and

enhances LDB1 stability to regulate gene expression. Notably, Ldb1 is essential for HSC specification during early development Downloaded from and maintenance in adults. We previously reported shortened lifespan and greater susceptibility to B cell lymphomas and carcinomas in Ssbp22/2 mice. However, whether Ssbp2 plays a regulatory role in normal HSC function and leukemogenesis is unknown. In this study, we provide several lines of evidence to demonstrate a requirement for Ssbp2 in the function and transcriptional program of hematopoietic stem and progenitor cells (HSPCs) in vivo. We found that hematopoietic tissues were hypoplastic in Ssbp22/2 mice, and the frequency of lymphoid-primed multipotent progenitor cells in bone marrow was reduced.

Other significant features of these mice were delayed recovery from 5-fluorouracil treatment and diminished multilineage recon- http://www.jimmunol.org/ stitution in lethally irradiated bone marrow recipients. Dramatic reduction of Notch1 transcripts and increased expression of transcripts encoding the transcription factor E2a and its downstream target Cdkn1a also distinguished Ssbp22/2 HSPCs from wild- type HSPCs. Finally, a tendency toward coordinated expression of SSBP2 and the AML suppressor NOTCH1 in a subset of the Cancer Genome Atlas AML cases suggested a role for SSBP2 in AML pathogenesis. Collectively, our results uncovered a critical regulatory function for SSBP2 in HSPC gene expression and function. The Journal of Immunology, 2014, 193: 000–000.

small pool of pluripotent stem cells capable of self- TAL1/SCL appears to be dispensable for hematopoietic mainte- renewal maintains normal cellular hematopoiesis and nance in adults because of functional redundancy with LYL1 (4). A recovery from cytotoxic stress (1, 2). The delicate bal- In erythroid and megakaryocytic progenitor cells, the non–DNA- by guest on September 26, 2021 ance between hematopoietic stem cell (HSC) self-renewal and dif- binding transcriptional adaptor LMO2 is thought to bridge TAL1/ ferentiation during these processes requires precise coordination SCL1 and the self-dimerizing cofactor LDB1 to nucleate the as- of gene expression with the abundance of their protein products. sembly of b-HLH-GATA factor– and LDB1-containing multi- The core multiprotein transcriptional complex that regulates he- subunit complexes (5, 6).This pathway raises the question of matopoietic and progenitor stem cell (HSPC) activity is composed whether a similar mechanism occurs in HSCs. Furthermore, unlike of the b-HLH protein TAL1/SCL, the Lim-only protein 2 (LMO2), TAL1/SCL1, LMO2 and LDB1 are continuously required for LIM domain–binding protein 1 (LDB1), and the zinc finger protein postnatal HSC maintenance (7–9). Elegant global chromatin GATA2. Initial gene-targeting studies demonstrated an absolute immunoprecipitation coupled with high-throughput sequenc- requirement for the HSC-expressed and lineage-restricted factor, ing (ChIP-Seq) studies have localized LDB1, TAL1/SCL1, and TAL1/SCL1, in embryonic HSC specification (3). In contrast, GATA2toconservedpromoterelementsontheTAL1/SCL1,

*Department of Genetics, University of Texas M.D. Anderson Cancer Center, Hous- Received for publication February 5, 2013. Accepted for publication August 11, ton, TX 77030; †Department of Stem Cell Transplantation, University of Texas M.D. 2014. Anderson Cancer Center, Houston, TX 77030; ‡Department of Veterinary Medicine This work was supported by HL744409, a developmental research project award from and Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030; x Specialized Program Of Research Excellence Grant P50-CA100632, and the Loper Department of Radiation Physics, University of Texas M.D. Anderson Cancer Center, { family research funds (to L.N.). Flow cytometry, genotyping, and veterinary services Houston, TX 77030; Graduate Program in and Development, University of ‖ were supported by National Cancer Institute Core Grant CA16672 to the University of Texas M.D. Anderson Cancer Center, Houston, TX 77030; Department of Leukemia, Texas M.D. Anderson Cancer Center. University of Texas M.D. Anderson Cancer Center, Houston, TX 77030; #Department of Medicine, Vanderbilt University, Nashville, TN 37232; **Department of Cell and Address correspondence and reprint requests to Dr. Lalitha Nagarajan, University of Developmental Biology, Vanderbilt University, Nashville, TN 37232; ††Department of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Box 45, Houston, Cancer Biology, Vanderbilt University, Nashville, TN 37232; ‡‡Vanderbilt-Ingram TX 77030. E-mail address: [email protected] Cancer Center, Vanderbilt University, Nashville, TN 37232; xxDepartment of Lym- The online version of this article contains supplemental material. phoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030; {{Division of Pediatrics, University of Texas M.D. Anderson Cancer Abbreviations used in this article: AML, acute myelogenous leukemia; BM, bone marrow; Center, Houston, TX 77030; ‖‖Department of Hematopathology, University of Texas 5-FU, 5-fluorouracil; HSC, hematopoietic stem cell; HSPC, hematopoietic stem and pro- M.D. Anderson Cancer Center, Houston, TX 77030; and ##Graduate Program in genitor cell; LDB1, Lim-domain binding protein 1; LMO2, Lim-only protein 2; LMPP, Human Molecular Genetics, Center for Stem Cell and Developmental Biology, and lymphoid-primed multipotent progenitor cell; LSK, Lin–Sca1+c-Kit+; MPP, multipotent Center for Cancer Genetics and Genomics, University of Texas M.D. Anderson Cancer progenitor cell; SSBP2, sequence-specific ssDNA-binding protein 2; WT, wild-type. Center, Houston, TX 77030 Ó 1J.L. and Y.K. contributed equally to this work. Copyright 2014 by The American Association of Immunologists, Inc. 0022-1767/14/$16.00 2P.Z.-M. and H.M.A. contributed equally to this work.

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1300337 2 ROLE OF Ssbp2 IN HSC ACTIVITY

Gata2, Runx1, Lmo2, E2a,andMyb genes. Furthermore, de- Materials and Methods creased expression of Tal1/Scl1, Gata2, Runx1, Lmo2,andMyb Mice 2/2 in Ldb1 mouse HSPCs has unequivocally demonstrated 2 2 Ssbp2 / mice were described previously (15). For the competi- these encoded to be positive regulators of HSPC- tive transplantation experiments, the mice were backcrossed to a specific gene expression (9). C57BL/6 (CD45.2) background for 10 generations. Mouse bleeding Human sequence specific ssDNA–binding protein 2 (SSBP2) and tissue harvests were according to Institutional Animal Care and was positionally cloned as a myeloid leukemia suppressor User Committee–approved protocols following standard operating from a chromosomal disruption within a critical region of loss in procedures. 5q13-14 in an acute myelogenous leukemia (AML) cell line (10). Immunofluorescence analysis SSBP2 and the related SSBP3 and SSBP4 were so designated be- cause of the in vitro ssDNA-binding activity of the founding BM mononuclear cells enriched by centrifugation or depleted of lineage- committed cells through magnetic-activated cell separation were stained member CSDP, the chicken ortholog of SSBP3 (11); however, the for SSBP2. Lin–Sca1+c-Kit+ (LSK) cells were purified by staining the Lin– significance of this putative ssDNA-binding activity in vivo is cells with anti–Sca1–PE and anti–c-Kit–FITC Abs (BD Pharmingen), unknown. All three proteins can bind LDB1 through a highly followed by cell sorting using a BD FACS Aria IIu sorter. Cells were conserved N-terminal domain; in turn, LDB1 binds the LIM centrifuged onto slides and fixed in 4% paraformaldehyde in PBS for 20 domains of LMO or LIM homeodomain proteins (LHX) through min, which was followed by permeabilization and then staining with rabbit polyclonal anti-SSBP2 Ab or mouse anti-LDB1 Ab (Molecular Probes) as an evolutionarily conserved COOH-terminal LIM-interacting do- detailed previously (26). main.

We and others have established that SSBPs enhance promoter Phenotypic analysis of HSPCs by flow cytometry Downloaded from occupancy and transcriptional activity of LMO2- and LHX- Seven-color FACS analysis (Pacific Blue, Alexa 488, allophycocyanin-Cy7, containing complexes by preventing the ubiquitylation and ulti- Alexa 647, PE, and PerCP-Cy5.5) was performed using a Fortessa cell mately the proteasomal degradation of LDB1 by the E3 ubiquitin analyzer (BD Biosciences), and data were analyzed with FlowJo software ligase RNF12/RLIM (12–15). Clearly, SSBPs have been selected (Tree Star). The Abs used against mouse Ags, including Pacific Blue for and evolutionarily maintained as key modulators of LDB1 lineage mixture (Pacific Blue anti-mCD3, -mLy-6G(Ly-6C), -mCD11b, -mCD45R(B220), and -mTer-119), Sca1–FITC, c-Kit–allophycocyanin- activity. In Caenorrhabditis elegans, for example, the sole SSBP Cy7, CD34–Alexa 647, Flt3–PE, and CD48–PerCP-Cy5.5 were from BD http://www.jimmunol.org/ ortholog SAM-10 coordinates with the LDB1 ortholog in a cell- Pharmingen, eBioscience, or BioLegend. BM mononuclear cells were autonomous fashion to regulate synaptic differentiation (16). stained with Pacific Blue–conjugated lineage mixture along with Sca1– + + Similarly, the expression of the Drosophila ortholog, SSDP, Alexa 488, c-Kit–allophycocyanin-Cy7, and Sca1 ,c-Kit cells were de- fined as LSK cells. Similar procedures were used to analyze Flt3–CD34– is a rate-limiting cofactor that regulates combinatorial tran- LSK cells (LT-HSCs), Flt3–CD34+ LSK cells (ST-HSCs), Flt3loCD34+ scriptional signals from CHIP (LDB1)-APTEROUS (LHX) or LSK cells (multipotent progenitor cells [MPPs]), Flt3hiCD34+ LSK cells CHIP (LDB1)-PANNIER (GATA) complexes (17, 18). Finally, in (lymphoid-primed multipotent progenitor cells [LMPPs]), and CD48+ or – zebrafish, SSDPs regulate neural patterning and sensory neuronal CD48 LSK cells. growth in part through LDB1 stabilization (19). Although SSBPs

Hematopoietic recovery from myelotoxic agents by guest on September 26, 2021 from all of these species binds and stabilizes LDB1, consistent with extensive evolutionary conservation of these members of this Mice were administered a single i.p. dose of 5-fluorouracil (5-FU) at 150 gene family (.99% identity), there also may be distinct function mg/kg body weight. Fifty-microliter aliquots of peripheral blood samples 2 2 collected from 5-FU–treated and control (untreated) mice were ana- for each member. Consistent with this notion, Ssbp2 / mice are 2/2 lyzed at 6, 12, and 18 d after 5-FU injection in a Siemens Advia 120 viable (15), whereas Ssbp3 mice die in utero (20, 21). In ad- auto analyzer for total and differential counts. Two complementary dition, a genome-wide in vivo screen by genetic cross of two approaches were taken to examine the role of Ssbp2 in HSPC recovery strains of mice that differ in hippocampal neurogenesis suggested from 5-FU. In the first set, wild-type (WT) BM was transplanted into Ssbp2 to be a quantitative trait regulating neuronal survival lethally irradiated Ssbp2 null mice, and the mice were challenged with 5-FU 4 mo after transplantation. In the second set, donor BM cells and regeneration (22). Finally, the highly penetrant autoimmune (CD45.2) were prepared from Ssbp22/2 or Ssbp2+/+ control mice and defects and enhanced predisposition to B cell lymphomas and mixed with congenic competitor BM cells (CD45.1) at a 10:1 ratio. The carcinomas of Ssbp22/2 mice suggest a unique role of Ssbp2 in cell mixture (5 3 106) was transplanted into the irradiated recipients + lymphoid differentiation and tumor suppression (15). (CD45.1 ) by tail vein injection, and the mice were challenged with 5-FU as detailed above. The pivotal role of Ldb1 in HSPC maintenance (9, 23) suggests that Ssbp2 plays a similarly important role. In this study, we report Competitive BM transplantation that Ssbp2 exerts a nonoverlapping regulatory function to maintain + murine HSPCs. In addition, reestablishment of homeostasis after Age-matched female congenic B6.SJL-PtrcaPep3b/BoyJ (B6.BoyJ;CD45.1 ) mice (The Jackson Laboratory) were used as transplant recipients. These elimination of cycling HSPCs by myeloablative treatment is im- mice were lethally irradiated (9.5 Gy). Donor BM cells (CD45.2) were 2/2 paired upon Ssbp2 ablation. Moreover, Ssbp2 bone marrow prepared from Ssbp22/2 or Ssbp2+/+ control mice and were mixed with (BM) competes poorly in multilineage reconstitution of lethally congenic competitor BM cells (CD45.1) at a 1:1 or 10:1 ratio. The mixed 3 6 + irradiated recipients. Furthermore, decreased expression of Notch1 cells (5 10 ) were transplanted into the irradiated recipients (CD45.1 ) by tail vein injection. Donor-derived engraftment and multilineage re- and elevated expression of E2a and its transcriptional target 2/2 constitution in BM and peripheral blood from recipients were assessed Cdkn1a in Ssbp2 HSPCs identify a role for Ssbp2 in the by flow cytometry using CD45.1–allophycocyanin or CD45.2–PE-Cy7 at HSPC-specific gene expression program. Finally, because ac- 4 and 16 wk posttransplantation to evaluate for short- and long-term cumulating evidence suggests a myeloid suppressor role for engraftment, respectively. NOTCH1 (24, 25), we evaluated the expression of NOTCH1 Real-time quantitative PCR and SSBP2 from data obtained from the Cancer Genome Atlas (TCGA). Interestingly, NOTCH1 expression was modestly corre- RNAwas isolated using the RNeasy mini kit (Qiagen). cDNAwas generated lated with SSBP2 expression in primary AML samples. Taken using SuperScript II reverse transcriptase (Invitrogen). Samples were normalized using 18S rRNA, and gene expression levels were determined together, these initial biological findings lay the foundation for using the relevant primers and probes on a TaqMan ABI7900HT instrument future mechanistic investigations on SSBP2-regulated transcrip- (Applied Biosystems). Each experiment was performed five times in tional networks in HSPCs. triplicate. Data are expressed as mean 6 SD. The Journal of Immunology 3

Analysis and data mining ciated with lymphoid priming and increased lymphoid gene ex- + hi TCGA AML database (27) was accessed, and the pertinent data were pression program. The frequency of CD34 FLT3 LMPPs was analyzed with cBioPortal for AML cases with mutations, copy number reduced by one-third. Although there was a trend toward fewer alterations, or an at least 2-fold change in expression level by RNA- LMPPs, the differences were not statistically significant in this sequence analysis (28, 29). small group of mice (WT, n = 4; null, n = 5). Furthermore, there + lo Statistical analysis was no compensatory increase in the frequency of CD34 FLT3 population, suggesting the decrease in FLT3hi expression in flow Data were analyzed for significance using two-tailed Student t test. A p cytometry was not due to absence of Flt3 upregulation by SSBP2 value , 0.05 was considered statistically significant. in the pre-LMPP cells. Likewise, the signaling lymphocyte acti- vation molecule marker CD150 distinguishes LT-HSCs in the Results larger the LSK population, whereas CD48 expression detects an Expression of Ssbp2 in HSPCs and hypoplasia of uncommitted, early progenitor population (32). Once again, the 2/2 hematopoietic organs in Ssbp2 mice frequency of CD48– cells representing LT-HSCs was increased in To determine whether Ssbp2 expression was regulated in a differ- the absence of Ssbp2 in contrast to the decreased frequency of entiation-specific manner, we examined BM cells enriched for CD48+ uncommitted progenitor cells with restricted amplifying HSPC subpopulations for transcript levels. Overall, Ssbp2 ex- potential (Fig. 3D, 3F). pression was higher in the long-term [Flt3–Lin–Sca+c-Kit+ (Flt3– In summary, the frequency of both CD34– cells and FLT3– cells LSK)] and short-term (Flt3+ LSK) HSPC populations than in com- within the LSK compartment increased and that of LSK cells mitted progenitor cells (LKs), and expression was lower in the expressing CD34 or FLT3 decreased (Supplemental Figs. 1, 2). Downloaded from lineage-positive (Lin+) cells than in the committed progenitor cells That these results would reflect a change in absolute numbers if (Fig. 1A). These findings suggested that Ssbp2 expression was a larger cohort of mice were used cannot be ruled out. Collec- repressed as the cells differentiated. Immunofluorescence staining tively, these data suggested a seminal role for Ssbp2 in main- of whole BM confirmed restricted expression, with expression in taining early HSPC homeostasis. ∼5% of all cells (Fig. 1B). Consistent with our previous charac- Delayed hematopoietic recovery of Ssbp22/2 mice after terization of the high specificity of anti-SSBP2 Abs (30), no http://www.jimmunol.org/ 2 2 myeloablative treatment specific signal was detected in Ssbp2 / BM. Flow cytometry– sorted populations revealed abundant levels of nuclear expression To assess whether loss of Ssbp2 affected the hematopoietic re- and colocalization with LDB1 in both Flt3+ LSKs and Flt3– LSKs sponse to the cytotoxic stress we observed, we challenged mice (Fig. 1C). These images reflected the relative change in Ssbp2 with a single dose of the myeloablative drug 5-FU, which 2 2 transcripts shown in Fig. 1A. The localization of SSBP2 to nuclear eliminates cycling HSPCs. Ssbp2 / mice displayed a markedly speckles in LK and Lin– cells was reminiscent of our previous delayed recovery as reflected in serial peripheral blood leukocyte findings with epithelial cells and fibroblasts in which SSBP2 lo- counts postdrug injection. On day 12, the total percentage of pe- calized to punctate nuclear structures (26, 31). Although the na- ripheral blood leukocytes was 75 6 10% in WT mice but only ture of these structures is unknown, our work in progress suggests 20 6 5% in null mice (p , 0.05) (Fig. 4A). The recovery of both by guest on September 26, 2021 that non–LDB1-interacting elements also may be present. myeloid cells and lymphoid cells was impaired; likewise, platelet To evaluate whether hematopoiesis was affected in the absence and RBC recovery were delayed (data not shown). The normal of Ssbp2, we first assessed mice 6–8 wk of age for the absolute BM cellularity of a WT mouse on day 12 was noticeably altered, number of total nucleated cells in the BM, spleen, and thymus. In with decreased BM cellularity observed in null mice (Fig. 4B). all three tissues, the mononuclear population was reduced by 30– Hypoplasia of both myeloid and erythroid lineages was evident in 2 2 40% (p , 0.05 for each tissue type), suggesting an overall re- Ssbp2 / BM (data not shown). duction in hematopoietic activity (Fig. 2A). The absolute number Weissman et al. (33) demonstrated that early stages of recovery of PBLs was twice as high in WT than in null mice (Fig. 2B, left from 5-FU treatment are characterized by a transient expansion panel). Although there was a tendency for an increase in gran- of Lin–Sca+ HSPCs. Therefore, we examined the BM of null and ulocytes, the values did not reach statistical significance (Fig. 2B, WT mice by flow cytometry at 48 and 96 h after 5-FU treatment. right panel). Likewise, the absolute number of RBCs and platelets Unlike the 50% expansion of Lin–Sca+ cells seen in the WT mice 2 2 in WT and Ssbp2-null mice were not significantly different (data (from 30 to 46%) between days 2 and 4, Ssbp2 / mice showed not shown). a more modest 12% expansion (from 33 to 36%) (Fig. 4C). These

2/2 findings suggested a diminished potential for stress-mediated HSPC homeostasis is impaired in Ssbp2 mice HSPC expansion in the absence of Ssbp2. The abundant expression of SSBP2 in HSPCs prompted us to These mice were homozygously deleted for Ssbp2 in all tissues, determine whether normal HSPC homeostasis was perturbed in the so it was important to examine cell intrinsic versus microenvi- absence of Ssbp2. Accordingly, flow cytometric analyses for stem ronmental mechanisms producing these results. To determine and early progenitor cell subpopulations were performed with whether the defects were intrinsic or extrinsic to HSCs, we null mice and their WT littermates. Characterization of the LSK transplanted isogenic (CD45.2) WT BM cells into lethally irra- population, which is composed of stem and early progenitor cells, diated congenic Ssbp22/2 mice, which had been lethally irradi- revealed no significant difference in frequencies (Fig. 3B). Within ated to ablate autologous hematopoiesis. The recipients also were the LSK compartment, the frequency of CD34– cells was signif- challenged with 5-FU after they had been fully engrafted (4 mo icantly lower in WT than null mice, suggesting an increase in the after the transplantation), and we found no significant difference ratio of early immature HSPCs (Supplemental Fig. 1B). CD34 and in the recovery rate between transplanted null mice and the control FLT3 expression were used to further subdivide the LSK sub- mice at that point. Regardless, the poor reconstituting potential of population (Fig. 3C). The CD34–Flt3– fraction, which corre- Ssbp22/2 BM prevented us from conducting the reverse experi- sponded to LT-HSCs, was twice as high in null than in control ments (i.e., testing the 5-FU response in WT mice reconstituted mice, whereas there was no significant difference in the ST-HSC with null mouse BM). In a complementary approach, Ssbp22/2 or or CD34+FLT3– population (Fig. 3E). FLT3 expression is asso- WT BM cells (CD45.2) were mixed with WT CD45.1 donor BM 4 ROLE OF Ssbp2 IN HSC ACTIVITY Downloaded from

FIGURE 1. Abundant Ssbp2 expression and colocalization with LDB1 in murine HSPCs. (A) Ssbp2 RNA expression pattern in subpopulations of mouse BM cells as detected by real-time PCR. Cells were sorted by flow cytometry based on their surface markers, including LT-HSCs (Lin–Sca1+c-Kit+Flt3– [Flt3– LSKs]), ST-HSCs (Lin–Sca1+c-Kit+Flt3+ [Flt3+ LSKs]), progenitor cells (Lin–c-Kit+ [LKs]), and Lin+ cells. Data represent mean 6 SD from three independent experiments. (B) SSBP2 expression is restricted to a few cells in normal BM. Mononuclear cells from whole BM were immunostained with anti-SSBP2 Abs. Nuclear DNA was stained with DAPI (blue). Representative fields illustrate Ab specificity. A few cells in WT BM were immunoreactive. http://www.jimmunol.org/ No signal was detected in null mice. (C) SSBP2 and LDB1 colocalize in the nucleus. FACS-sorted subpopulations of HSPCs were fixed, permeabilized, and stained with anti-SSBP2 and -LDB1 Abs. Nuclear DNA was stained with DAPI. cells at a ratio of 10:1 and transplanted into lethally irradiated WT Multilineage repopulating activity of Ssbp22/2 BM is severely CD45.1 mice (Fig. 4E) and the recipients challenged with 5-FU impaired after they had been fully engrafted (4 mo after the transplantation). Next, we tested the ability of Ssbp22/2 BM cells to competitively Surprisingly, we found no significant difference in the recovery reconstitute hematopoiesis in lethally irradiated mice. We assessed rate between mice transplanted with Ssbp2 2/2 or WT-enriched

their relative short- and long-term repopulating potential at 4 and by guest on September 26, 2021 BM (Fig. 4E). However, because of technical limitations in mon- 16 wk by transplanting BM cell mixtures containing different ratios itoring CD45.1 or CD45.2 alleles during recovery, these findings of Ssbp22/2 donor to WT competitor cells into lethally irradiated should be interpreted with caution. Overall, these findings are recipients. Initial transplantations at a ratio of 1:1 donor to com- consistent with a contribution from nonhematopoietic cells of the petitor revealed that WT cells predominated in both BM (7 versus BM niche to the recovery Ssbp22/2 HSPCs from cytotoxic stress. 1%; p , 0.05) and peripheral blood (13 versus 2%; p , 0.05) at 4 wk

FIGURE 2. Hypoplasia of hematopoietic tissues in Ssbp22/2 mice. (A) Reduced cellularity of BM, spleen, and thymus in Ssbp2-null mice. Cells from single-cell suspensions prepared from BM, spleen, and thymus from Ssbp22/2 mice and their WT littermates were counted. (B) PBLs were decreased in the absence of Ssbp2. Absolute numbers of PBLs and granulocytes are shown. Data represent mean 6 SD from six age- and gender-matched pairs of WT and null mice. *p , 0.05 versus WT as determined with unpaired two-tailed t test. The Journal of Immunology 5 Downloaded from http://www.jimmunol.org/ by guest on September 26, 2021

FIGURE 3. Ssbp2 deletion affects the frequency of HSPC subpopulations. (A) Frequency of Lin– cells is unaffected by Ssbp22/2 BM. A representative image of gating within live population for lineage-negative cells. (B) Frequency of LSK cells was unaffected by Ssbp22/2 BM. A representative image of c-Kit, Sca1 gating in Lin– cells. (C and E) Frequency of LMPPs (Flt3hiCD34+ LSKs) is decreased in Ssbp22/2 BM. (C) Representative flow cytometry analysis showing LT-HSCs (Flt3–CD34– LSKs), ST-HSCs (Flt3–CD34+ LSKs), MPPs (Flt3loCD34+ LSKs), and LMPPs (Flt3hiCD34+ LSKs) within LSKs in Ssbp2-null mice and their WT littermates. (E) Frequency of LT-HSCs, ST-HSCs, MPPs, and LMPPs within the LSK population in WT (n = 5) and null mice (n = 5). (D, F, and G) Frequency of CD482 cells is increased in Ssbp22/2 BM. (D) Representative flow cytometry analysis showing CD48+ cells within LSKs in Ssbp2-null mice and their WT littermates. (F) Frequency of CD48+ and CD48– population within the LSK compartment in WT (n = 5) and null mice (n = 5). (G) Representative histogram for anti-CD48 staining. Note the increase in frequency in CD48– cells in null mice. Data in (E) and (F) represent mean 6 SD from age-matched WT (n = 5) and null mice ([n =5]*p , 0.05) as determined by unpaired two-tailed t test between WT and Ssbp22/2 mice. posttransplantation (Fig. 5A). However, when the ratio was in- (CD45.1). At 24 and 48 h posttransplantation, there was no sig- creased to 10:1, Ssbp22/2 cells could be detected in ∼50–70% of nificant difference between the percentage of CD45.2–-expressing the recipients (Fig. 5A). Despite the transplantation of a much WT and Ssbp22/2 cells in homing to the BM (24 h: 1.2 versus higher number of Ssbp22/2 than WT cells, the frequency of donor 1.0%; 48 h: 2.7 versus 2.8%) (Supplemental Fig. 2A, 2B). In cells in the Ssbp22/2 recipients was still significantly lower than addition, we sorted the transplanted cells for Flt3 expression 18 h that of WT recipients at 4 wk (72.2 6 6.7 versus 89.6 6 6.1%; after transplantation and found no significant difference between p , 0.05) and at 16 wk (55 6 7.5 versus 96 6 12.2%; p , 0.05), WT and null donors (32 versus 29%, Supplemental Fig. 2C). suggesting a diminished potential for short- or long-term recon- Taken together, these data demonstrated reduced multilineage stitution (Fig. 5B). repopulating activity in Ssbp22/2 HSPCs. We next examined the peripheral blood contribution of Ssbp22/2 cells to specific hematopoietic lineages. Consistent with the de- Abundant expression of b-HLH factor E2a and loss of Notch1 creased bone contribution to BM homeostasis, the peripheral expression in the absence of Ssbp2 blood cell types, including granulocytes, macrophages, and B To assess whether the impaired HSC activity in Ssbp22/2 mice and T lymphocytes, was severely compromised (Fig. 5C). correlates with an altered gene expression pattern, we evaluated To rule out the possibility that the reduced repopulating capacity transcript levels of 16 genes encoding known regulators of qui- of Ssbp22/2 HSCs was due to impaired homing of these cells to escence and self-renewal in purified HSPCs: Trp53, Myc, BM, we compared the ability of Ssbp22/2 cells (CD45.2) and p27Kip1, Cdkn1a (p21), Gfi1, Ldb1, Runx1, E2a, HoxB4, Bmi1, their WT counterparts to move into the BM of irradiated recipients Klf10, Ski, Sox4, Trim27, p18Ink4c,andNotch1). Of these, E2a 6 ROLE OF Ssbp2 IN HSC ACTIVITY Downloaded from http://www.jimmunol.org/

FIGURE 4. Delayed hematopoietic recovery from cytotoxic stress in Ssbp22/2 mice. (A) Peripheral blood hematopoietic recovery is delayed in Ssbp22/2 mice. Hematopoietic reconstitution was monitored by serial peripheral blood counts in mice injected with a single dose of 5-FU (150 mg/kg, i.p.). Total WBC counts are shown as a percentage of the initial values for each group of mice. (B) Hypoplastic BM on day 12 post–5-FU treatment in Ssbp22/2 mice. H&E staining of BM from Ssbp2-null mice and their WT littermates at day 12 post–5-FU injection. (C) Early post–5-FU HSPC recovery is compromised in Ssbp22/2 BM. A representative flow cytometric analysis showing Lin2Sca1+ cells in BM from Ssbp22/2 mice and their WT littermates 2 and 4 d after 5-FU injection. (D) WT BM rescues Ssbp22/2 mice from delayed post–5-FU recovery. Lethally irradiated WT and Ssbp22/2 cells were reconstituted with

WT BM. Four months after transplantation, recipients were challenged with a single dose of 5-FU, and hematopoietic recovery was followed with serial by guest on September 26, 2021 peripheral blood counts. WBC counts are shown as a percentage of the initial values for each group of mice. *p , 0.05 as determined by unpaired two- tailed t test between WT and Ssbp22/2 mice. Each experiment was conducted twice with a group of six age-matched mice per genotype. (E) WT mice reconstituted with Ssbp22/2-enriched marrow show normal hematopoietic recovery after 5-FU treatment. and Cdkn1A (p21) were expressed at a significantly higher level address the consequences of disrupting a critical transcriptional and Gfi1 at a significantly lower level in null HSPCs than in WT pathway in HSPC homeostasis because SSBPs directly interacting HSPCs (p , 0.05 for each gene). Although high expression levels with LDB1 and are present in all of the same transcriptional of E2a-andCdkn1a-positive regulators of quiescence and a complexes examined to date. Earlier ChIP-Seq studies established modest decrease in Gfi1 expression a negative regulator of HSC the colocalization of LDB1, SCL1, and GATA2 on the Tal1/SCL1, quiescence appear contradictory, the net outcome reflected im- Gata2, Runx1, Lmo2, E2a, and Myb gene promoters in HSPCs. paired stress recovery and engraftment defects and suggested More important, a feed-forward autoregulatory loop may exist perturbation in the transcriptional network overall. because the expression all these genes except E2A was decreased Accumulating evidence has suggested a myeloid leukemia in the absence of LDB1 (9). suppressor role for NOTCH1 (24, 25). Therefore, we queried TCGA AML dataset for SSBP2 and NOTCH1 expression as de- Abundant SSBP2 expression and colocalization with LDB1 in termined by RNA-Seq studies. As shown in Fig. 6C, 21 of 166 HSPCs identified cases (12.6%) had loss of SSBP2 expression and 27 High expression of Ssbp2 in undifferentiated stem and progenitor cases (16.2%) had increased expression. Twenty-four percent of cells, mirroring Ldb1 expression (9), is indicative of a role for the patients with low SSBP2 expression also had decreased Ssbp2 in normal hematopoiesis (Fig. 1A). These findings are in NOTCH1 expression, whereas 9 of the 27 cases (37% of total) agreement with global expression profiling studies. SSBP2 is with higher SSBP2 expression also had elevated NOTCH1 ex- frequently on the list of genes preferentially expressed in HSPCs pression. The coordinated expression of both genes was mod- (34–37). Because nuclear localization of Drosophila orthologs erately significant (p = 0.046). Considering the genetic and (SSDPs) depends on the nuclear localization signal of LDB1 (38), phenotypic heterogeneity in AML (27), these results, taken to- the colocalization with LDB1 raises the possibility of an LDB1- 2/2 gether with the absence of Notch1 expression in Ssbp2 HSPCs, dependent role for SSBP2 in HSPCs (Fig. 1C). Such an interac- suggest that this axis is preserved in a subset of AMLs. tion suggests that some of the phenotypic consequences of Ssbp2 loss may be Ldb1 dependent. Moreover, the unexpected locali- Discussion zation of SSBP2 to nuclear speckles in differentiated cells sug- Our studies revealed Ssbp2 as a novel positive regulator of HSC gests that SSBP2–LDB1 interactions may be less crucial in homeostasis and recovery from cytotoxic stress. These findings mature cells. As we reported previously, in most epithelial cells The Journal of Immunology 7

FIGURE 5. Long-term and short-term repopulating ability is decreased in the absence of Ssbp2.(A) Ssbp22/2 BM cells do not reconstitute at a 1:1 donor- to-competitor ratio. Short-term engraftment (4 wk) results as determined with CD45.2-expressing cells in BM and peripheral blood are depicted. Data represent mean 6 SD from five recipient mice. (B) Reduced frequency of Ssbp22/2 BM donor cells in peripheral blood of recipients transplanted at a donor-to-compet- itor ratio of 10:1. Engraftment analyses were performed at 4 wk (short term) and 16 wk (long term) after transplantation. Data represent mean 6 SD from one of two experiments each with five mice per genotype. *p , 0.05 as determined by unpaired two-tailed t test between WT and Ssbp22/2 mice. (C) Contribution to all

the peripheral blood lineages is compromised in the Downloaded from absence of Ssbp2. Representative analysis showing percent CD45.1 and CD45.2 populations within gran- ulocyte(Gr1+), macrophage (Mac1+), B lymphocyte (B220+), and T lymphocyte (CD3+) in the peripheral blood from Ssbp22/2 BM recipients 16 wk after transplantation. http://www.jimmunol.org/

(MCF7, HeLa, and A549) and in fibroblasts (IMR90), SSBP2 Impaired HSPC homeostasis and stress response in Ssbp22/2 localizes to punctate structures in both the nucleus and nucleolus mice (26, 31). In some of these cells, SSBP2 colocalized with the DNA/ The biological consequences of Ssbp2 ablation, reduced cellularity by guest on September 26, 2021 RNA-binding protein families of ILF2 and EWS (H. Liang et al., in multiple hematopoietic tissues (Fig. 2), increased frequency of manuscript in preparation). Future studies should shed light on the LT-HSCs, and reduced frequency of LMPPs, underscore the sig- role of LDB1-dependent and -independent SSBP2 pathways in nificance of high Ssbp2 expression in HSPCs (Fig. 3). Although hematopoiesis. the absolute number of HSPC subpopulations was not significantly

FIGURE 6. Gene expression alterations in Ssbp22/2 HSPCs. (A and B) E2a and Cdkn1a transcript levels are increased and Notch1 transcript levels are decreased in the absence of Ssbp2. Quantitative RT-PCR results illustrate differences between Ssbp22/2 and WT counterparts in the expression of HSC quiescence (A) and self-renewal regulators (B) normalized to 18sRNA control. Representative results of triplicates from one of two separate cDNA pools from a group of five mice. *p , 0.05 between paired samples. (C) Tendency toward coordinated SSBP2 and NOTCH1 expression in primary AMLs. The cBIOPortal database was queried for at least .2-fold alteration in expression as determined by RNA-Seq; 166 AML cases were identified. Each column represents a patient. p value at 95% confidence interval is 0.047. 8 ROLE OF Ssbp2 IN HSC ACTIVITY altered, the increased frequency of LT-HSCs and decreased fre- ChIP studies will further elucidate the mechanistic underpinnings quency of LMPPs pointed to impediments to normal HSPC dif- of SSBP2-regulated transcriptional network. Regardless, the pre- ferentiation and stress response in the absence of Ssbp2. A larger liminary findings shown in Fig. 6 identify Ssbp22/2 mice as cohort of mice may better detect a small but biologically signifi- a novel tool with which to examine the complex E2A-NOTCH1 cant change in the absolute number of cells. axis in regulating HSPC integrity. When cycling HSPCs are eliminated by 5-FU treatment or other Human SSBP2 was positionally cloned as a candidate mye- hematopoietic stress, quiescent HSCs capable of self-renewal enter lodysplasia/AML suppressor gene from chromosome 5q14 (10). the cell cycle rapidly and re-establish the stem cell pool before Although most patients harbor large deletions, the smallest re- returning to dormancy (33, 39, 40). The initial proliferative burst gion of overlap is thought to map to 5q31. However, a recent of HSCs, which expands the Lin–Sca+ population, was severely study with high-density single nucleotide polymorphism arrays compromised in the absence of Ssbp2 (Fig. 4). Whether the HSPC demonstrated that larger deletions are associated with poorer defects are HSC intrinsic or due to a niche contribution or some outcome and shortened disease-free survival (48). In addition, combination of the two cannot be readily answered by the present SSBP2 expression is decreased in primary AML stem cells (49) studies. These findings, regardless, raise the possibility of a role and cell lines (30) in contrast to normal HSCs. Unlike murine for Ssbp2 in an interplay between HSC and BM niche because HSCs, which acquire self-renewal abilities without transforma- reconstitution with either WT marrow or the microenvironment tion, with HoxB4 overexpression in two primates induced AML in rescued the delayed recovery from cytotoxic stress. Finally, the both (50). One of these monkeys harbored retroviral integration in possibility that Ssbp2 may be a strain-specific quantitative trait the SSBP2 gene and an associated reduction in expression by 70%. locus in hematopoiesis analogous to neuronal survival (22) cannot Moreover, inducible expression of SSBP2 in the myelomonocytic Downloaded from be ruled out. cell line U937 resulted in growth arrest and differentiation, al- The overall decrease in the short- and long-term reconstituting though the mechanisms are unknown (30). Although these find- potential of BM from null mice likewise implicates Ssbp2 in ings suggest a role for AML suppressor, direct evidence for HSPC expansion (Fig. 5). The subtle but significant differences SSBP2-mediated myeloid leukemia suppression is lacking (50). in the LT-HSC and LMPP frequency and pronounced defects in AML is genetically diverse (27, 51, 52). The role of NOTCH1

myeloregenerative response, and competitive repopulating activity in myeloid differentiation and transformation is controversial http://www.jimmunol.org/ of knockout BM cells underscore a requirement for Ssbp2 in and may reflect loss and gain of function roles. Overexpression of maintaining normal HSPC integrity. HES1, a NOTCH1 downstream target, exacerbates chronic mye- logenous leukemia blast crisis in BCR-ABL–induced murine Aberrant expression of regulatory genes and potential role in models (53). Furthermore, inactivating NOTCH1 mutations are AML found in chronic myelomonocytic leukemia. Taken together, loss Our characterization of aberrant E2a and Notch1 expression in the of SSBP2 and altered NOTCH1 expression may identify a distinct absence of Ssbp2 raises a number of questions. Both E2a and subset of leukemia initiating cells originating HSCs or MPPs, Notch1 have critical regulatory roles and potentially important whereas enhanced SSBP2 and NOTCH1 expression may define roles in HSPC maintenance, and our findings implicated SSBP2 LMPPs as a target of transformation. The present report is an by guest on September 26, 2021 directly or indirectly in modulating E2a and Notch1 transcription. early step in connecting these intricate networks with HSPC ac- E2a expression, which is first detectable in LT-HSCs, increases tivity and, potentially, AML pathogenesis. steadily as those cells develop into MPPs and LMPPs, then rises dramatically in common lymphoid progenitor cells (41). The en- Acknowledgments 2/2 hanced LT-HSC cycling and stem cell exhaustion seen in E2a We thank Dr. Margaret Goodell (Baylor College of Medicine, Houston, TX) mice has uncovered a role for E2A in HSC cycling through for helpful discussions and critical review of the results and Verlene Henry transcriptional upregulation of CDK inhibitors (42, 43). Elegant and Adrienne Ferguson (M.D. Anderson Cancer Center) for assistance with serial adoptive transfer studies with compound heterozygotes for tail vein injections and bleeding. We also thank Hank Adams (Department E2a and Cdkn1a suggest an absolute requirement for this path- of Genetics, M.D. Anderson Cancer Center) for assistance with microscopy. way in maintaining LT-HSCs (44). Furthermore, a global genetic screen identified E2a promoted Cdkn1a expression as a regulator Disclosures of cell cycle arrest in the absence of cell death in several tumor The authors have no financial conflicts of interest. cell lines (45). Although high expression levels of E2a and Cdkn1a potential positive regulators of quiescence and the modest References decrease in Gfi1 expression a negative regulator of HSC quies- 1. Wilson, A., E. Laurenti, and A. Trumpp. 2009. Balancing dormant and self- cence, appear contradictory, the net outcome of Ssbp2 loss was renewing hematopoietic stem cells. Curr. Opin. Genet. Dev. 19: 461–468. impaired stress recovery and engraftment defects. 2. Trumpp, A., M. Essers, and A. Wilson. 2010. Awakening dormant haemato- In normal BM, Notch1 expression increases modestly from LT- poietic stem cells. Nat. Rev. Immunol. 10: 201–209. 3. Shivdasani, R. A., E. L. Mayer, and S. H. Orkin. 1995. Absence of blood for- HSC to ST-HSCs. Similar to E2a expression, Notch1 expression mation in mice lacking the T-cell leukaemia oncoprotein tal-1/SCL. Nature 373: increases progressively from LT-HSC to MPPs to LMPPs before 432–434. 4. Souroullas, G. P., J. M. Salmon, F. Sablitzky, D. J. Curtis, and M. A. Goodell. rising dramatically in common lymphoid progenitor cells (46). 2009. Adult hematopoietic stem and progenitor cells require either Lyl1 or Scl 2/2 The reduced frequency of LMPPs in Ssbp2 BM could poten- for survival. Cell Stem Cell 4: 180–186. tially be a direct consequence of increased E2a expression because 5. Meier, N., S. Krpic, P. Rodriguez, J. Strouboulis, M. Monti, J. Krijgsveld, M. Gering, R. Patient, A. Hostert, and F. Grosveld. 2006. Novel binding partners HSPCs and downstream progeny are exquisitely sensitive to E2a of Ldb1 are required for haematopoietic development. Development 133: 4913– dosage (42) and to loss of Notch1 expression, which is critical for 4923. LMPP expansion. 6. Le´cuyer, E., S. Larivie`re, M. C. Sincennes, A. Haman, R. Lahlil, M. Todorova, M. Tremblay, B. C. Wilkes, and T. Hoang. 2007. Protein stability and tran- Clearly, the resolution of HSPC analyses and gene expression scription factor complex assembly determined by the SCL-LMO2 interaction. J. changes in subpopulations presented in this report can be enhanced Biol. Chem. 282: 33649–33658. 7. Warren, A. J., W. H. Colledge, M. B. Carlton, M. J. Evans, A. J. Smith, and considerably with the use of larger cohorts of mice and more T. H. Rabbitts. 1994. The oncogenic cysteine-rich LIM domain protein rbtn2 is sensitive signaling lymphocyte activation molecule markers (47). essential for erythroid development. Cell 78: 45–57. The Journal of Immunology 9

8. Mukhopadhyay, M., A. Teufel, T. Yamashita, A. D. Agulnick, L. Chen, 31. Fleisig, H. B., H. Liang, and L. Nagarajan. 2010. Adenoviral oncoprotein K. M. Downs, A. Schindler, A. Grinberg, S. P. Huang, D. Dorward, and E1B55K mediates colocalization of SSBP2 and PML in response to stress. J. H. Westphal. 2003. Functional ablation of the mouse Ldb1 gene results in severe Mol. Signal. 5: 6. patterning defects during gastrulation. Development 130: 495–505. 32. Kiel, M. J., O. H. Yilmaz, T. Iwashita, O. H. Yilmaz, C. Terhorst, and 9. Li, L., R. Jothi, K. Cui, J. Y. Lee, T. Cohen, M. Gorivodsky, I. Tzchori, Y. Zhao, S. J. Morrison. 2005. SLAM family receptors distinguish hematopoietic stem S. M. Hayes, E. H. Bresnick, et al. 2011. Nuclear adaptor Ldb1 regulates and progenitor cells and reveal endothelial niches for stem cells. Cell 121: 1109– a transcriptional program essential for the maintenance of hematopoietic stem 1121. cells. Nat. Immunol. 12: 129–136. 33. Randall, T. D., and I. L. Weissman. 1997. Phenotypic and functional changes 10. Castro, P., H. Liang, J. C. Liang, and L. Nagarajan. 2002. A novel, evolutionarily induced at the clonal level in hematopoietic stem cells after 5-fluorouracil conserved gene family with putative sequence-specific single-stranded DNA- treatment. Blood 89: 3596–3606. binding activity. Genomics 80: 78–85. 34. Eckfeldt, C. E., E. M. Mendenhall, C. M. Flynn, T. F. Wang, M. A. Pickart, 11. Bayarsaihan, D., and L. N. Lukens. 1996. Single-strand-DNA-binding factors S. M. Grindle, S. C. Ekker, and C. M. Verfaillie. 2005. Functional analysis of specifically recognize the pyrimidine element in the chick a2(I) collagen gene human hematopoietic stem cell gene expression using zebrafish. PLoS Biol. 3: promoter. Biochem. J. 314: 293–296. e254. 12. Cai, Y., Z. Xu, L. Nagarajan, and S. J. Brandt. 2008. Single-stranded DNA- 35. He, X., V. Gonzalez, A. Tsang, J. Thompson, T. C. Tsang, and D. T. Harris. binding proteins regulate the abundance and function of the LIM- 2005. Differential gene expression profiling of CD34+CD133+ umbilical cord homeodomain transcription factor LHX2 in pituitary cells. Biochem. Biophys. blood hematopoietic stem progenitor cells. Stem Cells Dev. 14: 188–198. Res. Commun. 373: 303–308. 36. Wagner, W., A. Ansorge, U. Wirkner, V. Eckstein, C. Schwager, J. Blake, 13. Gungo€ ¨r, C., N. Taniguchi-Ishigaki, H. Ma, A. Drung, B. Tursun, K. Miesala, J. Selig, R. Saffrich, W. Ansorge, and A. D. Ho. 2004. Molecular H. P. Ostendorff, M. Bossenz, C. G. Becker, T. Becker, and I. Bach. 2007. evidence for stem cell function of the slow-dividing fraction among human Proteasomal selection of multiprotein complexes recruited by LIM homeo- hematopoietic progenitor cells by genome-wide analysis. Blood 104: 675–686. domain transcription factors. Proc. Natl. Acad. Sci. USA 104: 15000–15005. 37. Laurenti, E., S. Doulatov, S. Zandi, I. Plumb, J. Chen, C. April, J. B. Fan, and 14. Xu, Z., X. Meng, Y. Cai, H. Liang, L. Nagarajan, and S. J. Brandt. 2007. Single- J. E. Dick. 2013. The transcriptional architecture of early human hematopoiesis stranded DNA-binding proteins regulate the abundance of LIM domain and LIM identifies multilevel control of lymphoid commitment. Nat. Immunol. 14: 756–763. domain-binding proteins. Genes Dev. 21: 942–955. 38. van Meyel, D. J., J. B. Thomas, and A. D. Agulnick. 2003. Ssdp proteins bind to 15. Wang, Y., S. Klumpp, H. M. Amin, H. Liang, J. Li, Z. Estrov, P. Zweidler- LIM-interacting co-factors and regulate the activity of LIM-homeodomain Downloaded from McKay, S. J. Brandt, A. Agulnick, and L. Nagarajan. 2010. SSBP2 is an in vivo protein complexes in vivo. Development 130: 1915–1925. tumor suppressor and regulator of LDB1 stability. Oncogene 29: 3044–3053. 39. Wilson, A., E. Laurenti, G. Oser, R. C. van der Wath, W. Blanco-Bose, 16. Zheng, Q., A. M. Schaefer, and M. L. Nonet. 2011. Regulation of C. elegans M. Jaworski, S. Offner, C. F. Dunant, L. Eshkind, E. Bockamp, et al. 2008. presynaptic differentiation and neurite branching via a novel signaling pathway Hematopoietic stem cells reversibly switch from dormancy to self-renewal initiated by SAM-10. Development 138: 87–96. during homeostasis and repair. Cell 135: 1118–1129. 17. Bronstein, R., L. Levkovitz, N. Yosef, M. Yanku, E. Ruppin, R. Sharan, 40. Venezia, T. A., A. A. Merchant, C. A. Ramos, N. L. Whitehouse, A. S. Young, H. Westphal, B. Oliver, and D. Segal. 2010. Transcriptional regulation by CHIP/ C. A. Shaw, and M. A. Goodell. 2004. Molecular signatures of proliferation and

LDB complexes. PLoS Genet. 6: e1001063. quiescence in hematopoietic stem cells. PLoS Biol. 2: e301. http://www.jimmunol.org/ 18. Bronstein, R., and D. Segal. 2011. Modularity of CHIP/LDB transcription 41. Dias, S., R. Ma˚nsson, S. Gurbuxani, M. Sigvardsson, and B. L. Kee. 2008. E2A complexes regulates cell differentiation. Fly (Austin) 5: 200–205. proteins promote development of lymphoid-primed multipotent progenitors. 19. Zhong, Z., H. Ma, N. Taniguchi-Ishigaki, L. Nagarajan, C. G. Becker, I. Bach, Immunity 29: 217–227. and T. Becker. 2011. SSDP cofactors regulate neural patterning and differenti- 42. Semerad, C. L., E. M. Mercer, M. A. Inlay, I. L. Weissman, and C. Murre. 2009. ation of specific axonal projections. Dev. Biol. 349: 213–224. E2A proteins maintain the hematopoietic stem cell pool and promote the mat- 20. Nishioka, N., S. Nagano, R. Nakayama, H. Kiyonari, T. Ijiri, K. Taniguchi, uration of myelolymphoid and myeloerythroid progenitors. Proc. Natl. Acad. W. Shawlot, Y. Hayashizaki, H. Westphal, R. R. Behringer, et al. 2005. Ssdp1 Sci. USA 106: 1930–1935. regulates head morphogenesis of mouse embryos by activating the Lim1-Ldb1 43. Yang, Q., L. Kardava, A. St Leger, K. Martincic, B. Varnum-Finney, complex. Development 132: 2535–2546. I. D. Bernstein, C. Milcarek, and L. Borghesi. 2008. E47 controls the develop- 21. Enkhmandakh, B., A. V. Makeyev, and D. Bayarsaihan. 2006. The role of the mental integrity and cell cycle quiescence of multipotential hematopoietic pro- proline-rich domain of Ssdp1 in the modular architecture of the vertebrate head genitors. J. Immunol. 181: 5885–5894.

organizer. Proc. Natl. Acad. Sci. USA 103: 11631–11636. 44. Santos, P. M., Y. Ding, and L. Borghesi. 2014. Cell-intrinsic in vivo requirement by guest on September 26, 2021 22. Kempermann, G., E. J. Chesler, L. Lu, R. W. Williams, and F. H. Gage. 2006. for the E47–p21 pathway in long-term hematopoietic stem cells. J. Immunol. Natural variation and genetic covariance in adult hippocampal neurogenesis. 192: 160–168. Proc. Natl. Acad. Sci. USA 103: 780–785. 45. Andrysik, Z., J. Kim, A. C. Tan, and J. M. Espinosa. 2013. A genetic screen 23. Li, L., J. Freudenberg, K. Cui, R. Dale, S. H. Song, A. Dean, K. Zhao, R. Jothi, identifies TCF3/E2A and TRIAP1 as pathway-specific regulators of the cellular and P. E. Love. 2013. Ldb1-nucleated transcription complexes function as pri- response to p53 activation. Cell Reports 3: 1346–1354. mary mediators of global erythroid gene activation. Blood 121: 4575–4585. 46. Oh, P., C. Lobry, J. Gao, A. Tikhonova, E. Loizou, J. Manent, B. van Handel, 24. Klinakis, A., C. Lobry, O. Abdel-Wahab, P. Oh, H. Haeno, S. Buonamici, I. van S. Ibrahim, J. Greve, H. Mikkola, et al. 2013. In vivo mapping of notch pathway De Walle, S. Cathelin, T. Trimarchi, E. Araldi, et al. 2011. A novel tumour- activity in normal and stress hematopoiesis. Cell Stem Cell 13: 190–204. suppressor function for the Notch pathway in myeloid leukaemia. Nature 473: 47. Oguro, H., L. Ding, and S. J. Morrison. 2013. SLAM family markers resolve 230–233. functionally distinct subpopulations of hematopoietic stem cells and multipotent 25. Kannan, S., R. M. Sutphin, M. G. Hall, L. S. Golfman, W. Fang, R. M. Nolo, progenitors. Cell Stem Cell 13: 102–116. L. J. Akers, R. A. Hammitt, J. S. McMurray, S. M. Kornblau, A. M. Melnick, 48. Jerez, A., L. P. Gondek, A. M. Jankowska, H. Makishima, B. Przychodzen, M. E. Figueroa, and P. A. Zweidler-McKay. 2013. Notch activation inhibits R. V. Tiu, C. L. O’Keefe, A. M. Mohamedali, D. Batista, M. A. Sekeres, et al. AML growth and survival: a potential therapeutic approach. J Exp Med 210: 2012. Topography, clinical, and genomic correlates of 5q myeloid malignancies 321–337. revisited. J. Clin. Oncol. 30: 1343–1349. 26. Fleisig, H. B., N. I. Orazio, H. Liang, A. F. Tyler, H. P. Adams, M. D. Weitzman, 49. Qian, Z., A. A. Fernald, L. A. Godley, R. A. Larson, and M. M. Le Beau. 2002. and L. Nagarajan. 2007. Adenoviral E1B55K oncoprotein sequesters candidate Expression profiling of CD34+ hematopoietic stem/ progenitor cells reveals leukemia suppressor sequence-specific single-stranded DNA-binding protein 2 distinct subtypes of therapy-related acute myeloid leukemia. Proc. Natl. Acad. into aggresomes. Oncogene 26: 4797–4805. Sci. USA 99: 14925–14930. 27. Cancer Genome Atlas Research Network. 2013. Genomic and epigenomic 50. Zhang, X. B., B. C. Beard, G. D. Trobridge, B. L. Wood, G. E. Sale, R. Sud, landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368: R. K. Humphries, and H. P. Kiem. 2008. High incidence of leukemia in large 2059–2074. animals after stem cell gene therapy with a HOXB4-expressing retroviral vector. 28. Cerami, E., J. Gao, U. Dogrusoz, B. E. Gross, S. O. Sumer, B. A. Aksoy, J. Clin. Invest. 118: 1502–1510. A. Jacobsen, C. J. Byrne, M. L. Heuer, E. Larsson, Y. Antipin, B. Reva, 51. Majeti, R., M. W. Becker, Q. Tian, T. L. Lee, X. Yan, R. Liu, J. H. Chiang, A. P. Goldberg, C. Sander, and N. Schultz. 2012. The cBio cancer genomics L. Hood, M. F. Clarke, and I. L. Weissman. 2009. Dysregulated gene expression portal: an open platform for exploring multidimensional cancer genomics data. networks in human acute myelogenous leukemia stem cells. Proc. Natl. Acad. Cancer Discovery 2: 401–404. Sci. USA 106: 3396–3401. 29. Gao, J., B. A. Aksoy, U. Dogrusoz, G. Dresdner, B. Gross, S. O. Sumer, Y. Sun, 52. Chen, S. J., Y. Shen, and Z. Chen. 2013. A panoramic view of acute myeloid A. Jacobsen, R. Sinha, E. Larsson, et al. 2013. Integrative analysis of complex leukemia. Nat. Genet. 45: 586–587. cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6: pl1. 53. Nakahara, F., M. Sakata-Yanagimoto, Y. Komeno, N. Kato, T. Uchida, 30. Liang, H., S. Samanta, and L. Nagarajan. 2005. SSBP2, a candidate tumor K. Haraguchi, K. Kumano, Y. Harada, H. Harada, J. Kitaura, et al. 2010. Hes1 suppressor gene, induces growth arrest and differentiation of myeloid leukemia immortalizes committed progenitors and plays a role in blast crisis transition in cells. Oncogene 24: 2625–2634. chronic myelogenous leukemia. Blood 115: 2872–2881.