Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

Research Article

Phf6 Loss Enhances HSC Self-Renewal Driving Tumor Initiation and Activity in T-ALL

Agnieszka A. Wendorff1, S. Aidan Quinn1,2, Marissa Rashkovan1, Chioma J. Madubata3, Alberto Ambesi-Impiombato1, Mark R. Litzow4, Martin S. Tallman5, Elisabeth Paietta6, Maddalena Paganin7, Giuseppe Basso7,8, Julie M. Gastier-Foster9,10,11,12, Mignon L. Loh13,14, Raul Rabadan3,15, Pieter Van Vlierberghe16,17, and Adolfo A. Ferrando1,2,3,18

abstract The plant homeodomain 6 (PHF6) is frequently mutated in human T-cell acute lymphoblastic leukemia (T-ALL); however, its specific functional role in leukemia development remains to be established. Here, we show that loss of PHF6 is an early mutational event in leukemia transformation. Mechanistically, genetic inactivation of Phf6 in the hematopoietic system enhances hematopoietic stem cell (HSC) long-term self-renewal and hematopoietic recovery after chemotherapy by rendering Phf6 knockout HSCs more quiescent and less prone to stress-induced activation. Consistent with a leukemia-initiating tumor suppressor role, inactivation of Phf6 in hemat- opoietic progenitors lowers the threshold for the development of NOTCH1-induced T-ALL. Moreover, loss of Phf6 in leukemia lymphoblasts activates a leukemia stem cell transcriptional program and drives enhanced T-ALL leukemia-initiating cell activity. These results implicate Phf6 in the control of HSC homeostasis and long-term self-renewal and support a role for PHF6 loss as a driver of leukemia- initiating cell activity in T-ALL.

SIGNIFICANCE: Phf6 controls HSC homeostasis, leukemia initiation, and T-ALL leukemia-initiating cell self-renewal. These results substantiate a role for PHF6 mutations as early events and drivers of leukemia stem cell activity in the pathogenesis of T-ALL.

1Institute for Cancer Genetics, Columbia University, New York, New York. 15Department of Biomedical Informatics, Columbia University, New York, 2Department of Pediatrics, Columbia University Medical Center, New New York. 16Center for Medical Genetics Ghent, Ghent University, Ghent, York, New York. 3Department of Systems Biology, Columbia University, Belgium. 17Cancer Research Institute Ghent, Ghent, Belgium. 18Depart- New York, New York. 4Division of Hematology, Mayo Clinic, Rochester, ment of Pathology and Cell Biology, Columbia University Medical Center, Minnesota. 5Department of Hematologic Oncology, Memorial Sloan Ket- New York, New York. 6 tering Cancer Center, New York, New York. Department of Medicine, Note: Supplementary data for this article are available at Cancer Discovery 7 Albert Einstein College of Medicine, Bronx, New York. Onco-Hematology Online (http://cancerdiscovery.aacrjournals.org/). Division, Department, Salute della Donna e del Bambino (SDB), University of Padua, Padua, Italy. 8Italian Institute for Genomic Medicine (HMG), Current address for A. Ambesi-Impiombato: PsychoGenics, Paramus, New Turin, Italy. 9Department of Pathology and Laboratory Medicine, Nation- Jersey. wide Children’s Hospital, Columbus, Ohio. 10Department of Pathology, Corresponding Author: Adolfo A. Ferrando, Columbia University Medical Ohio State University School of Medicine, Columbus, Ohio. 11Depart- Center, 1130 St. Nicholas Avenue, ICRC 402A, New York, NY 10032. Phone: ment of Pediatrics, Ohio State University School of Medicine, Columbus, 212-851-4611; Fax: 212-851-5256; E-mail: [email protected] 12 13 Ohio. Children’s Oncology Group, Arcadia, California. Department of doi: 10.1158/2159-8290.CD-18-1005 Pediatrics, University of California, San Francisco, California. 14Helen Diller Family Comprehensive Cancer Center, San Francisco, California. ©2018 American Association for Cancer Research.

OF1 | CANCER DISCOVERY March 2019 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

INTRODUCTION PHF6 mutations seem restricted to hematologic tumors, are most frequently found in tumors from male patients Somatic loss-of-function mutations in the plant homeo- (1, 2), and are typically nonsense and frameshift truncating domain factor 6 (PHF6) are highly prevalent in T-cell acute alleles resulting in complete loss of expression (1–3, lymphoblastic leukemia (T-ALL; ref. 1) and are also present in 10, 11). In all, genetic loss of PHF6 as a result of deletions or (AML; refs. 2, 3). In addition, PHF6 is mutations is present in about 20% of T-ALLs, in 20% to 25% the causative gene mutated in Börjeson–Forssman–Lehman of mixed phenotype acute (MPAL) with early T-cell syndrome (BFLS; MIM#301900), a rare X-linked Mendelian precursor (ETP) and T/myeloid characteristics, and in 3% of disease characterized by mental retardation, obesity, hypo­ AML cases (1–3, 10, 11). Interestingly, the development of gonadism, gynecomastia, digit abnormalities, large ears, and pediatric T-ALL in a male patient with BFLS harboring a ger- coarse and characteristic facial features (4). mline PHF6 nonsense mutation (12) and the presence of PHF6 Isolation of protein complexes has shown the interac- mutations in preleukemic clonal hematopoiesis (13, 14) sup- tion of PHF6 with the nucleosome remodeling deacetylase port a role for this tumor suppressor in leukemia initiation (NuRD) complex, a major chromatin regulator controlling and hematopoietic stem cell (HSC) self-renewal, respectively. nucleosome positioning and transcription with important roles in development, genome integrity, and cell-cycle pro- gression (5, 6). In addition, PHF6 localizes to the nucleolus RESULTS and interacts with the PAF1 transcription elongation com- plex (7) implicated in the control of RNA polymerase I activ- PHF6 Mutations Are Early Events in Leukemia ity and ribosomal DNA (rDNA) transcription (8), and with Transformation and Drive Enhanced HSC UBF (7, 9), a transcriptional activator in the RNA Pol I preini- Self-Renewal tiation complex, supporting a role for PHF6 in the control of To evaluate the potential role of PHF6 loss as a leukemia- ribosome biogenesis. initiating event, we analyzed the timing of somatically

March 2019 CANCER DISCOVERY | OF2

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

RESEARCH ARTICLE Wendorff et al. acquired mutations in T-ALL using Integrated Sequential demonstrating preservation of lymphoid-lineage differentia- Network (ISN; ref. 15) analysis of clonal evolution and muta- tion potential (Supplementary Fig. S3A and S3C). tion dynamics using whole-exome sequencing data from Most somatically acquired mutations in T-ALL presum- diagnostic and relapse leukemias. This analysis revealed that ably occur postnatally in the adult hematopoietic system. somatic mutations in PHF6 occur as early lesions in the To specifically evaluate the effects ofPhf6 deletion in adult natural history of T-ALL (P = 0.03; Fig. 1A), prompting us HSC function, we analyzed the hematopoietic system of to evaluate a mechanistic link between the loss of Phf6, HSC recipient mice engrafted with bone marrow cells from con- function, and transformation leading to leukemia initiation. trol wild-type (tamoxifen-treated Phf6f/Y Rosa26+/+; referred Toward this goal, we generated conditional Phf6 knockout to simply as Phf6f/Y Rosa26+/+) and adult Phf6 knockout mice mice (Supplementary Fig. S1A–S1C) and crossed them with (tamoxifen-treated Phf6−/Y Rosa26+/Cre-ERT2; hereafter referred a Vav-iCre line to inactivate Phf6 in fetal HSCs. Analysis of to as Phf6–/Y Rosa26+/Cre-ERT2). In this setting, loss of Phf6 8-week-old Phf6−/YVav-iCre animals revealed an expansion of in adult HSCs resulted in expansion of immature hemat- total immature hematopoietic LSK progenitors (Fig. 1B–D) opoietic LSK progenitors, resulting from increased numbers resulting from increased numbers of multipotent MPP2 and of MPP2 and MPP3 multipotent progenitors, as well as MPP3 populations compared with controls (Phf6+/YVav-iCre; lymphoid-committed MPP4 and CLP populations (Fig. 2A Fig. 1B–G) and no differences in more mature myeloid (MEP, and B; Supplementary Fig. S4A and B). Furthermore, we CMP, and GMP) and uncommitted lymphoid (CLP) bone also observed a modest increase in CMP-committed myeloid marrow progenitors (Supplementary Fig. S1D–S1G). Phf6 bone marrow progenitors, with no changes in MEP and knockout mice showed no significant differences in bone GMP populations (Supplementary Fig. S4C and S4D). We marrow B-cell precursors (Supplementary Fig. S2A and S2B), observed no differences in cell-cycle distribution between and analysis of thymic populations revealed only a modest wild-type and Phf6 knockout–derived HSCs (LSK CD48−) and but significant reduction of double-negative DN2 and DN3 MPP cells (LSK CD48+; Supplementary Fig. S4E and S4F). thymic progenitors (Supplementary Fig. S2C–S2G). Animals transplanted with Phf6−/Y Rosa26+/Cre-ERT2 bone mar- The observed expansion of LSK cells in hematopoietic-spe- row showed largely preserved B-cell development, with only cific Phf6 knockout mice further supports a potential role for a modest decrease in pre-B and immature B-precursor cell PHF6 in the control of HSC activity. To test this possibility, we populations (Supplementary Fig. S5A and S5B). In addition, evaluated HSC function in mixed bone marrow chimera serial and as noted in Phf6−/Y Vav-iCre knockout mice, analysis of transplantation assays. Primary transplants of Phf6 knockout the thymus in mice reconstituted with Phf6−/Y Rosa26+/Cre-ERT2 (Phf6−/YVav-iCre) cells showed improved early hematopoietic cells revealed a modest but significant reduction in DN2 and reconstitution 5 and 10 weeks after transplant, yet equivalent DN3 thymic progenitor cells (Supplementary Fig. S5C–S5G). long-term total lymphoid and myeloid regeneration capacity Consistently, mixed bone marrow chimera serial transplanta- 15 weeks after transplant compared with Phf6+/YVav-iCre wild- tion assays showed that Phf6−/Y Rosa26+/Cre-ERT2 adult HSCs type controls (Fig. 1H; Supplementary Fig. S3A and S3B). display improved competitive multilineage reconstitution, In secondary transplants, Phf6 knockout (Phf6−/YVav-iCre) increased serial repopulation capacity (Fig. 2C), and succes- cells showed increased total hematopoietic reconstitution sively enhanced retention of lymphoid potential in secondary and pronounced improvement in both lymphoid and mye- and tertiary transplants compared with Phf6f/Y Rosa26+/+ con- loid-lineage repopulation compared with controls (Fig. 1H; trols (Supplementary Fig. S6A–S6C). Moreover, as a result of Supplementary Fig. S3A). This phenotype was further accen- exhaustion of HSC function in these experiments, we noted tuated in tertiary transplants, in which Phf6 knockout cells a progressive decrease in the number of LSK cells with each markedly outcompeted their Phf6 wild-type counterparts consecutive transplantation of Phf6f/Y Rosa26+/+ wild-type bone (Fig. 1H). In these experiments, Phf6 wild-type serially trans- marrow (Fig. 2D), which was accompanied by an increase in planted HSCs showed a myeloid bias and decreased ability to the relative frequency of long term HSCs (LT-HSC) as a result generate B cells (Supplementary Fig. S3A and S3C), features of decreased hematopoietic output (Fig. 2E–H). characteristically associated with HSC aging and functional In comparison, secondary and tertiary recipients of Phf6−/Y exhaustion (16, 17). In contrast, HSCs from hematopoietic- Rosa26+/Cre-ERT2 knockout cells had a less severe and delayed specific Phf6 knockout mice effectively contributed to both decline in LSK numbers (Fig. 2D), showed preservation of lymphoid and myeloid reconstitution in tertiary transplants, hematopoietic function, improved MPP output, and lacked

Figure 1. PHF6 mutations are early events in T-ALL, and loss of Phf6 expands the hematopoietic stem compartment. A, Integrated Sequential Network (ISN) illustrating the sequential order of mutations (nodes) in diagnosis and relapse ALL samples (n = 37) by pooling evolutionary paths (arrows) across patients. B, FACS plots at the top show representative analysis of total myeloid progenitor cells (MyP: Lin− CD117+ Sca1−) and total hematopoietic stem and progenitor cells (LSK: Lin− CD117+ Sca1+) from Phf6 wild-type (Phf6+/YVav-iCre) and Phf6 knockout (Phf6−/YVav-iCre) littermates at 8 weeks of age. FACS plots at the bottom show representative analysis of LSK subpopulations: long-term HSCs (LT-HSC; Lin− CD117+ Sca1+ CD150+ CD48−), short- term HSCs (ST-HSC; Lin− CD117+ Sca1+ CD150− CD48−), MPP2 (Lin− CD117+ Sca1+ CD150+ CD48+), and MPP3 (Lin− CD117+ Sca1+ CD150− CD48+). For each representative population, frequencies reflect the mean percentage gated on total live cells. C, Frequency of LSK cells derived from Phf6 wild-type (n = 5) and Phf6 knockout (n = 4) littermates at 8 weeks of age. D, Quantification of total LSK cell numbers of populations depicted inB and C. E, The frequency of LT-HSCs, ST-HSCs, MPP2, MPP3, and lymphoid-restricted MPP4 (Lin− CD117+ Sca1+ CD135+ CD150−) progenitors derived from Phf6 wild-type (n = 5) and Phf6 knockout (n = 4) littermates. F, Absolute number of LT-HSCs and ST-HSCs as in B and E. G, Quantification of total cell numbers in multipotent (MPP2 and MPP3) and lymphoid-restricted (MPP4) progenitor cell populations as in B and E. H, Total donor-derived cell frequencies in peripheral blood after primary, secondary, and tertiary competitive transplantation of bone marrow cells from Phf6 wild-type (Phf6+/YVav-iCre) or Phf6 knockout (Phf6−/YVav-iCre) control mice (n = 6–7 recipients per group). Plots of frequency and absolute cell number show individual mice; bars, mean ± standard deviation. P values were calculated using the two-tailed Student t test.

OF3 | CANCER DISCOVERY March 2019 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

PHF6 in Hematopoietic Stem Cells and T-ALL RESEARCH ARTICLE

PHF6 A BCL11B Early KRAS DNM2 P value FBXW7 EPHA3 less than HTR3A 0.04 SHROOM3 TP53 0.02 0.0001 USP9X SYNE1 NR3C1 WT1 Late STAT5B NOTCH1 11 NRAS cases CACNA1H

6 RYR1

1 JAK3 CREBBP NT5C2

B Phf6 +/Y Phf6 −/Y CD 1.0 0.001 50 0.01 Phf6 +/Y ) 0.5 0.75 4 Vav-iCre 0.8 40 1.8 1.9 Phf6 −/Y

CD117 0.6 30 Vav-iCre Sca-1 0.04 0.06 0.03 0.4 20

0.1 Frequency

0.2 10 Absolute cell number ( × 10

CD150 0.03 0.17 0.03 0.3 0 0 LSK LSK CD48

E FG 0.4 0.9 0.450.01 0.001 0.008 25 0.8 0.045 20 0.007 0.002 0.085 ) ) 3 4 20 0.3 15

15 0.2 10 10 Frequency

0.1 5 5 Absolute cell number ( × 10 Absolute cell number ( × 10

0 0 0 LT- ST- MPP2 MPP3 MPP4 LT- ST- MPP2 MPP3 MPP4 HSC HSC HSC HSC H 1ry transplant 2ry transplant 3ry transplant 100 100 100 0.002 0.03 0.3 Phf6 +/Y 80 80 80 Vav-iCre 60 60 60 Phf6 −/Y 40 40 40 Vav-iCre 20 20 20 Donor cells (%) P < 0.0001 P < 0.0001 0 0 0 51015 5101520 51018 Weeks after transplantation

March 2019 CANCER DISCOVERY | OF4

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

RESEARCH ARTICLE Wendorff et al.

A Phf 6 f/Y Phf 6−/Y B 1.5 0.003 0.04 0.002 0.006 ) 5 1.5 1.8 0.2 0.4 1.0 CD117 Sca-1 cell number ( × 10

+ 0.5 0.03 0.07 CD45.2

0.004 0.01 0 LT- HSC 0.08 0.2 MPP2 MPP3 MPP4 CD150 CD48 Phf6 f/Y Phf6 −/Y Rosa26 +/+ Rosa26 +/Cre-ERT2

C 1ry transplant 2ry transplant 3ry transplant 100 P < 0.0001 100 P < 0.005 100 P < 0.001 80 80 80 60 60 60 40 40 40 20 Phf6 +/Y 20 20 Donor cells (%) Phf6 −/Y 0 0 0 4812 16 20 4812 16 20 4812 16 20 Weeks after transplantation

D 7.5 E 1ry transplant 2ry transplant 3ry transplant

) 0.1 0.0002 0.02 5 17 17 29 17 40 22 + /Y 5.0 Phf6 39 47 37

13

LSK number ( × 10 14 912 4 15

+ 2.5 − /Y Phf6 CD45.2 40 75 57 0 CD150

1ry 2ry 3ry CD48

F G H LT-HSC MPP MPP4 60 0.1 0.02 0.0001 1.0 0.6 0.5 0.1 0.8 0.03 15 0.0005 0.04 ) *** ) 4 100 4 50 7.5 LSK (%) LSK (%) + + 40 75 10

30 5.0 50 cell number ( × 10 cell number ( × 10 + 20 + 5 2.5 25 10 CD45.2 CD45.2 Frequency of CD45.2 Frequency of CD45.2 0 0 0 0 1ry 2ry 3ry 1ry 2ry 3ry 1ry 2ry 3ry 1ry 3ry

OF5 | CANCER DISCOVERY March 2019 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

PHF6 in Hematopoietic Stem Cells and T-ALL RESEARCH ARTICLE the characteristic increase in the relative frequency of LT- out versus wild-type LT-HSCs. Of these significantly differen- HSCs observed in mice serially transplanted with wild-type tially accessible peaks, 2,334 (86%) became more accessible in cells (Fig. 2E–H; Supplementary Fig. S6D and S6E). In addi- Phf6 knockout LT-HSCs and 373 (14%) became less accessible, tion, in-depth analysis of bone marrow MPP progenitor indicating a global increase in chromatin accessibility in populations revealed preserved numbers of Phf6−/Y knockout- the Phf6 knockout setting (Fig. 3E). Notably, although only derived lymphoid-restricted MPP4 cells in tertiary relative to 10% of the total ATAC-seq peaks identified in LT-HSCs fell primary recipients of adult-deleted Phf6 knockout bone mar- within proximal promoter regions, 32% of the significantly row chimeras, a finding consistent with their distinctive long- differentially accessible peaks in Phf6 knockout LT-HSCs were term retention of lymphoid-lineage differentiation potential located at the proximal promoter, indicating a preferential (Fig. 2G and H; Supplementary Fig. S6). chromatin opening around transcription start sites (TSS; Next, to further and directly test the role of Phf6 in HSC Fig. 3F and G). Functional enrichment analysis of promoter/ self-renewal, we compared the reconstitution capacity of puri- TSS peaks with increased accessibility in Phf6 knockout LT- fied LT-HSCs fromPhf6 −/Y Rosa26+/Cre-ERT2 and wild-type Phf6f/Y HSCs revealed significant enrichment of GO Term categories Rosa26+/+ control mice after serial transplantation into iso- potentially related to HSC homeostasis, including cell cycle genic recipients. In this experiment, primary recipient mice (GO:0007049, P = 9.67 × 10−12); translation (GO:0006412, engrafted with Phf6-null LT-HSCs showed accelerated early- P = 4.75 × 10−12) and ribosome biogenesis (GO:0042254, P = stage hematopoietic reconstitution at 5 weeks after trans- 2.49 × 10−7); targets (MSIGDB:M15774, P = 5.3 × 10−9; plantation. Increased long-term donor-derived reconstitution MSIGDB:M17753, P = 1.807 × 10−10), and cellular response derived directly from Phf6-null LT-HSCs was maintained at to stress (GO:0033554, P = 2.28 × 10−7). Consistently, we 15 weeks after transplantation (Fig. 3A). Of note, equal num- observed increased promoter accessibility at MYC target gene bers of primary transplanted Phf6-null and wild-type LT-HSCs promoters in Phf6 knockout LT-HSCs (Fig. 3G). In addition, gave rise to equivalent numbers of LT-HSCs in the bone mar- gene-expression profiling by RNA sequencing (RNA-seq) of row of recipient mice 16 weeks after transplant (Fig. 3B). Strik- sorted Phf6 wild-type and knockout LT-HSCs revealed modest ingly, Phf6-null LT-HSCs gave rise to increased numbers of changes in with 306 differentially expressed

Phf6-null–derived downstream progenitor cells compared with (P < 0.01; log2 fold change > 0.3), of which 150 were Phf6 wild-type LT-HSCs; increased numbers of Phf6-null donor- upregulated and 156 were downregulated in Phf6 knockout derived cells included ST-HSCs, MPP2-4 progenitor cells, and LT-HSCs, respectively (Fig. 3H). Notably, this transcriptional myeloid-committed MEP, CMP, and GMP progenitors (Fig. 3A program was significantly enriched in genes controlled by and B). This phenotype was further augmented in recipients JAK1 in HSCs (ref. 18; Fig. 3I), supporting a potential conver- of secondary transplants performed with sorted LT-HSCs iso- gent transcriptional mechanism between genetic inactivation lated 16 weeks after primary transplantation (Fig. 3C). Of note, of Phf6 and JAK–STAT signaling in HSC self-renewal. and as observed in competitive bone marrow chimeras, Phf6 adult-deleted knockout LT-HSCs showed improved retention Loss of Phf6 Increases HSC Reconstitution of B lymphoid–cell reconstitution potential compared with Capacity and Quiescence after Insult wild-type controls, which showed preferential myeloid-lineage Under homeostatic conditions, LT-HSCs are predominantly differentiation (Fig. 3D). In all, these experiments indicate quiescent, a property characteristically required to maintain that loss of Phf6 results in increased LT-HSC self-renewal and their self-renewal capacity and functionally protective under hematopoietic reconstitution capacity with maintenance of stress (19). However, following hematopoietic challenges such long-term lymphoid-lineage commitment. as genotoxic insult by radiation or chemotherapy, HSCs tran- To explore the mechanisms mediating the effects of Phf6 siently proliferate to ensure efficient replenishment of blood in LT-HSCs, we performed genome-wide chromatin acces- and immune cellularity (19). Similarly, inflammatory antiviral sibility profiling by Assay for Transposase-Accessible Chro- responses and interferon signaling also induce the activation matin using sequencing (ATAC-seq). This analysis identified and proliferation of LT-HSCs, and their return to quiescence is 140,485 transposase-accessible peaks in mouse LT-HSCs, essential to avoid accumulation of DNA damage and apoptosis of which 2,707 were significantly differentially accessible (20). Loss of homeostatic equilibrium in the hematopoietic

(adjusted P value: Padj < 0.01; fold change > 4) in Phf6 knock- system in the context of chemotherapy or immune challenge

Figure 2. Phf6 deletion in adult hematopoiesis increases HSC self-renewal following long-term competitive serial transplantation. A, Representa- tive FACS plots of total donor-derived myeloid progenitor cells (MyP: Lin− CD117+ Sca1−) and total hematopoietic stem and progenitor cells (LSK: Lin− CD117+ Sca1+) 8–10 weeks after transplantation of lineage-negative cells isolated from Phf6 wild-type (Phf6f/YRosa26+/+) or Phf6 knockout (Phf6−/Y Rosa26+/Cre-ERT2) mice. For each representative population, frequencies reflect the mean percentage gated on total live cells n( = 5 per group). B, Absolute number of donor-derived LT-HSC, MPP2, MPP3, and MPP4 cells in recipients of Phf6 wild-type or Phf6 knockout donor bone marrow cells. C, Total donor-derived cell reconstitution in peripheral blood after primary, secondary, and tertiary competitive transplantation of bone marrow cells from Phf6 wild-type and Phf6 knockout mice (n = 5–7 recipients per group). D, Total number of donor-derived wild-type (Phf6f/YRosa26+/+) and knockout (Phf6−/Y Rosa26+/Cre-ERT2) LSK cells in the bone marrow of recipient mice 24 weeks after primary (n = 6 per group), secondary (n = 5 and 4, respectively), and ter- tiary (n = 5 per group) competitive mixed bone marrow transplantation. E, Representative FACS plots of total donor-derived stem and progenitor cells in mice after primary, secondary, and tertiary competitive mixed bone marrow transplantation with Phf6 wild-type or Phf6 knockout bone marrow cells. For each population, the mean percentage gated on total LSK cells is indicated. F, Frequency and absolute cell number of LT-HSCs in primary, secondary, and tertiary competitive mixed bone marrow recipients as in D and E. G, Frequency of total multipotent progenitor cells (MPP) in primary, secondary, and ter- tiary competitive mixed bone marrow recipients as in D and E. H, Absolute numbers of lymphoid-restricted MPP4 progenitor cells in primary and tertiary competitive mixed bone marrow recipients as in D and E. Plots of both frequency and absolute cell number show individual mice; bars, mean ± standard deviation. P values were calculated using two-tailed Student t test. ***, P < 0.0001.

March 2019 CANCER DISCOVERY | OF6

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

RESEARCH ARTICLE Wendorff et al.

A B C D f/Y −/Y

100 0.0002 0.001 0.007 12.5 80 0.01 0.01 0.0003 100 0.0001 Phf6 Phf6 0.9 0.0002 0.007 0.00030.01 *** *** 100 -HSCs (%)

(%) + + 80 10.0 80 60 80 0.00016

60 7.5 60 60 40

40 )/500 input LT 5.0 40

4 40 20 20 2.5 20 Donor derived (%) 20 0.00017 NS Frequency of CD45.2 Frequency of CD45.2 0 0 0 0 0 51015 Output ( × 10 51510 CD11b+ MEP + Weeks MPP2MPP3MPP4 CMPGMP Weeks CD3 LT-HSCST-HSC B220+

E 19 1 G 18 0MB 0MB 70MB 70MB 140MB All CDS gene promoters 0MB 0MB 1770MB 200 70MB f/Y 0MB 2 Phf6

16 140MB −/Y 70MB 150 Phf6

0MB 0MB 100 15 70MB 70MB 3 (RPGC) 0MB 140MB 50

0MB

14 70MB 0 Mean normalized signal

70MB

0MB −2.0 Kb TSS 2.0 Kb Phf6 f/Y Rosa26+/+ 4

70MB 13 Phf6 −/Y Rosa26+/Cre-ERT2

140MB 0MB MYC target gene promoters

0MB 70MB 250 Phf6 f/Y 12 70MB 5 Phf6 −/Y 0MB 200 140MB

70MB 0MB 150 11 70MB 0MB 6 100 (RPGC) 140MB 70MB 0MB 50 10 0MB 70MB 70MB 140MB 7 0MB 70MB 0MB 0 9 8 Mean normalized signal −2.0 Kb TSS 2.0 Kb

5’UTR 3’UTR F 1% H TTS 1% 2% Exon Promoter/TSS 3%

10% Ctsw Ifit3 Ifi44 Fndc4 Phf11d Mum1l1 Cxcl10 Cd14 H2−Ab1 Ccdc60 Ppm1e Fjx1 Irf7 Tubb1 Itm2a Slc1a3 Rab27b Mt1 Msmo1 Slc6a15 Paqr5 Ifit1 Adcy4 Cryz Dhx58 Myof Il2rg Gm16702 Mmp2 Tgm2 Gas6 Akr1c12 Prss57 Plk2 Foxa3 Smim6 Gimap3 Nectin2 Sell Dok2 Serpinf1 Igfbp4 Tnip3 Mpzl1 Hmox1 Gstm2 Sdsl Sult1a1 Bcam Phf6 − /Y Intergenic

40% Phf6

Intron f/Y 43% All peaks Phf 6 Row min Row 5’UTR 3’UTR 1% TTS 1% Exon I 2% Up in JAK1-KO 2% Down in JAK1-KO 0.2 NES = −1.61 0.5 0.1 P = 0.031 0.4 NES = 2.45 0.0 Intergenic 0.3 P < 0.0001 −0.1 32% 30% 0.2 −0.2 Promoter/TSS 0.1 −0.3 Intron 0.0 −0.4 32% Enrichment score (ES)

Differentially Enrichment score (ES) accessible Phf6 KO Phf6 WT Phf6 KO Phf6 WT peaks

Figure 3. Phf6 deletion in adult LT-HSCs increases hematopoietic repopulation capacity associated with increased chromatin accessibility and transcriptional upregulation of JAK–STAT signature genes. A, Frequency of total donor-derived cells in peripheral blood 5, 10, and 15 weeks after transplantation of 500 LT-HSCs sorted from Phf6 wild-type (Phf6f/YRosa26+/+) or Phf6 knockout (Phf6−/YRosa26+/Cre-ERT2) donor mice (n = 10 recipients per group). B, The absolute number of hematopoietic stem and progenitor cells (LT- and ST-HSCs, MPP2-4) and myeloid-restricted progenitor cells (MEP, CMP, and GMP) derived from 500 Phf6 wild-type or Phf6 knockout sorted LT-HSCs in the bone marrow of recipient mice 16 weeks after transplantations (n = 5 per group), as in A. C, Frequency of total donor-derived reconstitution in peripheral blood after secondary transplantation of 1,000 re-sorted Phf6 wild-type or Phf6 knockout LT-HSCs (n = 10 per group). D, Relative distribution of the B-cell (B220+), T-cell (CD3+), and myeloid (CD11b+) populations in peripheral blood 15 weeks after transplant, as in C. Values are shown for individual mice. Graphs represent mean ± standard deviation. P values were calculated using the two-tailed Student t test. ***, P < 0.0001. E, Genome-wide distribution of normalized chromatin accessibility in Phf6 knockout LT- HSCs (red track) and wild-type LT-HSCs (blue track) shown separately and in overlay (outer track). F, Distribution of ATAC-seq peaks, and of significantly differentially accessible peaks in Phf6 knockout compared with wild-type LT-HSCs. G, Normalized average ATAC-seq signal across all protein-coding transcript promoters (top) and MYC target genes (DANG_MYC_TARGETS_UP, C2 MSIG M6506) centered on the transcription start site (TSS). H, Heat map representation of top differentially expressed genes in Phf6 knockout compared with Phf6 wild-type LT-HSCs. I, Gene set enrichment plots from gene set enrichment analysis (GSEA) of a JAK1-loss signature comprised by genes differentially expressed in LT-HSCs of Jak1 knockout mice tested in Phf6 knockout LT-HSCs. Genes that display decreased expression in Jak1 knockout LT-HSCs were tested separately (left) from those that show increased expression (right). Normalized enrichment score (NES) and the FEWER P value are shown.

OF7 | CANCER DISCOVERY March 2019 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

PHF6 in Hematopoietic Stem Cells and T-ALL RESEARCH ARTICLE has been implicated in the expansion of HSCs harboring clonal circulating cells in peripheral blood 3 weeks after transplant hematopoiesis-associated mutations and in leukemia develop- (Fig. 5A). Nonetheless, analysis of the thymic compartment ment (21). In this context, we explored the capacity of Phf6-null in this setting revealed increased numbers of GFP+ cells in HSCs to withstand stress-induced activation by examining the mice transplanted with Phf6 knockout NOTCH1-L1601P- kinetics of recovery after myeloablative treatment with 5-fluo- ΔPEST–transduced bone marrow progenitor cells (Fig. 5B). rouracil (5-FU) compared with wild-type controls (22). In these Moreover, analysis of circulating tumor cells 5 and 11 weeks experiments, mice reconstituted with Phf6−/Y Rosa26+/Cre-ERT2 after transplant revealed accelerated leukemia development knockout cells showed accelerated and improved expansion in mice transplanted with Phf6 knockout NOTCH1-L1601P- of LSK cells and total myeloid progenitors (MyP; Lin− CD117+ ΔPEST–expressing progenitors (Fig. 5C), which ultimately Sca1−) on days 9 and 12 after treatment compared with wild- translated to increased leukemia penetrance and markedly type Phf6f/Y Rosa26+/+ controls (Fig. 4A and B; Supplementary accelerated leukemia-associated mortality in this group (Phf6 Fig. S7). In addition, recipients of Phf6 knockout bone marrow wild-type NOTCH1-L1601P-ΔPEST median survival = 23.6 showed improved recovery of total bone marrow cellularity, weeks; Phf6 knockout NOTCH1-L1601P-ΔPEST median sur- yet lower levels of activation-induced expansion in the HSC vival = 13.3 weeks; log-rank test P = 0.042; Fig. 5D). Inter- compartment (LSK CD48−), following genotoxic ablation of estingly, flow cytometry analysis of hematopoietic tissues hematopoietic progenitors with cyclophosphamide and rescue at endpoint showed increased GFP+ leukemia lymphoblast with G-CSF cytokine stimulation (ref. 23; Fig. 4C–E). To more infiltration in the thymus of animals bearingPhf6 knockout directly explore a potential cell-intrinsic role of Phf6 loss in HSC leukemia, despite nearly equivalent tumor burden in the bone quiescence, we tested the effects of interferon stimulation on marrow and spleen (Fig. 5E and F). Moreover, analysis of HSC activation by polyinosinic:polycytidylic acid [poly (I:C)] leukemia lymphoblasts recovered from leukemia-infiltrated treatment of mice reconstituted with control Phf6 wild-type tissues showed expression of T-cell surface markers with (Phf6f/Y Rosa26+/+) and Phf6 knockout (Phf6−/Y Rosa26+/Cre-ERT2) increased prevalence of a CD4+ CD8+ double-positive immu- bone marrow 8 weeks after transplant, a time frame in which nophenotype in Phf6 knockout tumors compared with Phf6 the hematopoietic system has returned to homeostatic equi- wild-type controls (Fig. 5G). Western blot analysis confirmed librium. Consistently, wild-type and Phf6 knockout–derived complete loss of PHF6 expression in Phf6 knockout tumor HSCs (LSK CD48−) and MPP cells (LSK CD48+) showed cells (Fig. 5H). no differences in cell-cycle distribution in basal conditions (Supplementary Fig. S4E and S4F). In contrast, and most Phf6 Loss Drives Leukemia-Initiating notably, 24 hours after poly (I:C) administration, Phf6 knock- Cell Activity in T-ALL out HSCs (LSK CD48−) showed a more limited proliferative To better explore the specific role ofPhf6 in the pathogenesis response and improved retention of the quiescent G0 cell- of T-ALL without the confounding effect of genetic and immu- cycle state compared with wild-type controls (Fig. 4F and nophenotypic heterogeneity of this model, we developed iso- G). Conversely, unrestrained cell-cycle reentry was observed genic tumors with loss of Phf6. Toward this goal, we generated in Phf6 knockout multipotent progenitor cells (LSK CD48+; NOTCH1-L1601P-ΔPEST–induced leukemias as described Fig. 4F and G). Taken together, these results support that above using hematopoietic progenitors from tamoxifen- the loss of Phf6 limits the proliferative response of LT-HSCs inducible conditional Phf6 knockout (Phf6f/YRosa26+/CreERT2) in reaction to genotoxic stress and facilitates hematopoietic mice. The resulting Phf6 conditional knockout leukemias reconstitution after injury. were transplanted into secondary recipients and treated with vehicle only or tamoxifen to generate matched Phf6 wild- type and knockout tumors, respectively (Supplementary Fig. Deletion of Phf6 Sensitizes Hematopoietic S8A–S8C). When transplanted into isogenic recipients, Phf6 Progenitors to Oncogenic NOTCH1-Induced knockout leukemia cells showed modestly accelerated tumor Transformation progression compared with Phf6 wild-type isogenic controls To formally and directly test the role of Phf6 loss as a leu- as documented by increased GFP+ cell counts in peripheral kemia-initiating event in T-ALL transformation, we infected blood (Fig. 6A) and accelerated mortality (log-rank test P = hematopoietic progenitor cells from Phf6 knockout (Phf6−/Y 0.013; Fig. 6B). However, and most notably, limiting dilution Vav-iCre) and wild-type (Phf6+/YVav-iCre) controls with bicis- transplantation assays demonstrated a marked increase in tronic GFP retroviruses driving expression of a constitu- leukemia-initiating cell activity in Phf6 knockout tumors com- tively active T-ALL–mutant form of the NOTCH1 pared with wild-type isogenic controls, supporting a role for (NOTCH1-L1601P-ΔPEST; refs. 24, 25). Of note, activating loss of Phf6 in promoting leukemia blast self-renewal (Fig. 6C mutations in NOTCH1 are highly prevalent in T-ALL (24) and D; Supplementary Fig. S8D). and frequently co-occur with mutational loss of PHF6 (1). Analysis of transcriptional programs associated with Phf6 In this model, forced expression of mutant NOTCH1 typi- loss in matched isogenic Phf6 wild-type and Phf6-null leu- cally results in a wave of extrathymic T-cell lymphopoiesis 3 kemia samples revealed a distinct gene-expression signature weeks after transplantation, which manifests as a transient dominated by upregulated transcripts (608 genes upregu- appearance of GFP+ preleukemic CD4+ CD8+ double-positive lated and 67 genes downregulated; P < 0.001, fold change > 2; (DP) T cells in peripheral blood, and is followed by full T-ALL Fig. 6E). Genes upregulated in Phf6-null leukemia lympho- leukemia transformation 10 to 15 weeks later (25). Mice blasts included the early hematopoietic marker Cd34, as well transplanted with Phf6 knockout NOTCH1-L1601P-ΔPEST– as genes associated with B-cell (Cd19, Pax5) and myeloid (Kit, expressing progenitors showed a consistent wave of GFP+ Cd14, Cd38, Flt3, Csf3r, Fcgr1) differentiation, even though

March 2019 CANCER DISCOVERY | OF8

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

RESEARCH ARTICLE Wendorff et al.

A B D9 D12 LSK MyP 0.04 0.4 3.2 0.4 2.0 0.03 0.0015 106 0.045 0.001 f/Y

) Phf6 5 +/+ f/Y 5 Rosa26

6 10 1.5 −/Y Phf Phf6 104 Rosa26+/Cre 1.0 0.4 1.2 9.2 1 103

0.5 Absolute cell number − /Y 2

6 10 Absolute cell number ( × 10 Phf 0 101 CD117 D9 D12 D9 D12 Sca-1

C D E D2 LSK CD48− LSK CD48+ 60 0.4 0.0006 80 0.004 0.02 0.2 80 0.16 0.20.3 Ph f6 )

6 50 60 60 f/Y 40 40 40 30 Percent (%) Percent Percent (%) Percent 20 20 20 Ph f6

− /Y 0 0 10 Absolute cell number ( × 10 G0 G1 S/G2/M G0 G1 S/G2/M

f/Y f/Y 0 CD117 Phf6 Phf6 D2 D4 Sca-1 Phf6 −/Y Phf6 −/Y

F G LSK CD48− LSK CD48+ LSK CD48− LSK CD48+ 16 8 34 20 100 0.0035 0.02 0.003 100 0.10.3 0.013 Ph f6 80 80 f/Y 60 60 75 42

rcent (%) 40 rcent (%) 40

7 3 17 13 Pe Pe

Ph f6 20 20

− /Y 0 0 G0 G1 S/G2/M G0 G1 S/G2/M 90 70 Ki67 Phf6f/Y Phf6f/Y Hoechst Phf6 −/Y Phf6 −/Y

Figure 4. Phf6 loss facilitates hematopoietic recovery following genotoxic stress and enhances retention of cell quiescence after DNA damage and interferon challenge. A, Representative FACS plots of CD117 and Sca-1 expression in lineage-negative bone marrow cells of Phf6 wild-type (Phf6f/Y Rosa26+/+) and Phf6 knockout (Phf6−/YRosa26+/Cre-ERT2) mice 9 days (n = 4 per group) and 12 days (n = 5 per group) after 5-FU treatment. Average frequen- cies of LSK (Lin− CD117+ Sca-1+) and total myeloid progenitor cells (MyP: Lin− CD117+ Sca-1−) are indicated. B, Absolute quantification of LSK and MyP populations as in A. Graphs show values for individual mice; bars, mean and standard deviation. C, Absolute bone marrow cell numbers of Phf6 wild-type (Rosa26+/+Phf6f/Y) and Phf6 knockout (Rosa26+/Cre-ERT2Phf6−/Y) mice following cyclophosphamide treatment (Cy) and 2 (D2) or 4 (D4) consecutive doses of G-CSF. D, Representative FACS plots of CD117 and Sca-1 expression in lineage-negative bone marrow cells of Phf6 wild-type (Rosa26+/+Phf6f/Y) and Phf6 knockout (Rosa26+/Cre-ERT2Phf6−/Y) mice following cyclophosphamide treatment (Cy) and 2 (D2) consecutive doses of G-CSF. E, Cell-cycle distribution of Phf6 wild-type (n = 5) and Phf6 knockout (n = 5) CD48− LSK and CD48+ LSK cells following cyclophosphamide treatment (Cy) and 2 (D2) consecutive doses of G-CSF. F, Representative FACS plots of Ki67 expression and DNA content (Hoechst) in bone marrow LSK CD48− stem and LSK CD48+ progenitor cells of Phf6 wild-type (Rosa26+/+Phf6f/Y) and Phf6 knockout (Rosa26+/Cre-ERT2Phf6−/Y) mice 24 hours after p(I:C) treatment. G, Cell-cycle distribution in bone mar- row LSK CD48− stem and LSK CD48+ progenitor populations of Phf6 wild-type (n = 5) and Phf6 knockout (n = 4) mice 24 hours after p(I:C) treatment. P values were calculated using the two-tailed Student t test. Bar graphs show values for individual mice ± standard deviation.

OF9 | CANCER DISCOVERY March 2019 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

PHF6 in Hematopoietic Stem Cells and T-ALL RESEARCH ARTICLE

A BC

100 0.2 6 0.007 100 0.01 100 0.04 /Y

) + ) ) Phf6 ) + + + 7 P P 80 5 P 80 80 Vav-iCre × 10 GF GF GF

( −/Y 4 Phf6 60 60 60 Vav-iCre 3 40 Thymus 40 40

cell number 2

+ P 20 1 20 20 GF Peripheral blood (% Peripheral blood (% Peripheral blood (% 0 0 0 0 3 weeks 3 weeks 5 weeks 11 weeks

D F Spleen Liver 100 Phf6 +/Y 0.04 Phf6 −/Y + /Y 75 Phf6

50 − /Y Percent survival 25 Phf6

0 0 10 20 30 G Weeks after transplantation Phf6 +/Y Phf6 −/Y

10%

12.5% 20% E 25% 62.5% 10 0.1 10 0.5 15.0 0.025 60% 10% ) ) ) 7 8 7 8 8 12.5 1.0 DN DP DP+CD8 6 6 ISP-CD8 Mixed 7.5 4 4 5.0 cell number ( × 10 cell number ( × 10 cell number ( × 10 + + + H Phf6 +/Y Phf6 −/Y 2 2 2.5 GFP GFP GFP PHF6 0 0 0.0 Bone marrow Spleen Thymus GAPDH

Figure 5. Loss of Phf6 enhances NOTCH1-induced T-ALL development. A, Peripheral blood quantification of GFP+ cells in mice transplanted with NOTCH1-L1601P-ΔPEST–transduced Phf6 wild-type (Phf6+/YVav-iCre; n = 20) and Phf6 knockout (Phf6−/YVav-iCre; n = 19) bone marrow progenitors showing transient NOTCH1-driven preleukemic T-cell expansion 3 weeks after transplant. B, Quantification of GFP+ preleukemia cell infiltration in the thymus of mice 3 weeks after transplantation with NOTCH1 L1601P-ΔPEST–transduced Phf6 wild-type and Phf6 knockout (n = 5 per group) bone marrow progenitors. C, Peripheral blood analysis of early (5 weeks) and late (11 weeks) leukemia development detected by circulating GFP+ leukemia cells in mice transplanted with NOTCH1-L1601P-ΔPEST–transduced Phf6 wild-type and Phf6 knockout bone marrow progenitors. D, Kaplan–Meier survival curves of mice harboring Phf6 wild-type (Phf6+/YVav-iCre; n = 14) and Phf6 knockout (Phf6−/YVav-iCre; n = 14) NOTCH1-induced leukemias, as in C. E, Flow cytom- etry quantification of leukemia infiltration in bone marrow, spleen, and thymus ofPhf6 wild-type and Phf6 knockout NOTCH1-induced leukemia-bearing mice at endpoint. F, Representative micrographs of histology analysis of Phf6 wild-type and Phf6 knockout NOTCH1-induced leukemias. G, Phenotypic categorization of Phf6 wild-type and Phf6 knockout NOTCH1-induced leukemias based on CD4 and CD8 surface expression analyzed by flow cytometry. H, Western blot analysis of PHF6 expression in Phf6 wild-type and Phf6 knockout NOTCH1-induced leukemia lymphoblasts. P values in A–C and E were calculated using the two-tailed Student t test. P values in D were calculated using the log-rank test. Scale bars in F, 50 μm.

March 2019 CANCER DISCOVERY | OF10

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

RESEARCH ARTICLE Wendorff et al.

Phf6 wild-type and Phf6 knockout isogenic tumors showed ers of leukemia-initiating cell activity (21, 34). In this context, no apparent differences in their immunophenotype (Fig. 6F). DNMT3A and TET2, two epigenetic factors governing HSC Notably, suppression of Phf6 in mouse B-cell lymphoma cells self-renewal, have emerged as prominent tumor suppressors has been described to increase lineage plasticity, in this case frequently mutated in clonal hematopoiesis and as early resulting in decreased expression of B cell–lineage genes and events in the pathogenesis of AML and myelodysplastic syn- upregulation of T-cell markers (26), and PHF6 mutations dromes (21, 34). As in the case of DNMT3A and TET2, PHF6 are prevalent in ETP ALLs and myeloid/T MPALs, which are mutations can be found in clonal hematopoiesis associ- characterized by mixed expression programs and lineage plas- ated with aging (14) and following recovery from aplastic ticity (1–3, 10, 11). In addition, genes upregulated upon Phf6 anemia (13), and Phf6 inactivation increases the long-term knockout in our NOTCH1 leukemia model include bona fide self-renewal capacity of HSCs. However, although DNMT3A T-ALL oncogenes such as Lmo2, Mef2c, Zeb2, and Hex (ref. 27; and TET2 both regulate DNA methylation, PHF6 is a nucleo- Fig. 6F), which are characteristically expressed at higher levels lar protein implicated in transcriptional control, ribosome in early immature ETP ALL leukemias (28, 29). In addition, biogenesis, and the regulation of nucleosome positioning and in agreement with an increase in leukemia-initiating via interaction with the NuRD nucleosome remodeling com- cell activity, gene set enrichment analysis showed preferen- plex (35). Notably, analysis of Phf6 wild-type and knockout tial activation of a gene-expression program associated with mice showed increased chromatin accessibility in Phf6-null human leukemia stem cell activity (30) in Phf6 knockout LT-HSCs preferentially involving MYC targets and genes NOTCH1-induced T-ALL cells compared with isogenic wild- implicated in cell cycle, translation, and response to stress, in type controls (Fig. 6G). Finally, and most notably, analysis support of a chromatin remodeling function of PHF6 in HSC of this leukemia-initiating cell signature in human T-ALLs homeostasis. In addition, gene-expression profiling showed validated and extended these results, showing a marked inverse effects of Phf6 inactivation and Jak1 knockout in enrichment of leukemia stem cell genes (31) in human PHF6- LT-HSCs, potentially indicating a convergent mechanism of mutant leukemias compared with matched PHF6 wild-type action between PHF6 inactivation and JAK–STAT signaling controls (Fig. 6H). in promoting HSC self-renewal. PHF6 in Leukemia Development DISCUSSION Clonal evolution mapping analysis supports that PHF6 Chemoresistant leukemia-initiating cells with expanded mutations may occur early in T-ALL transformation, and we self-renewal potential are major drivers of leukemia relapse demonstrate experimentally that Phf6 loss can enhance the and are associated with poor prognosis (32). Self-renewal oncogenic activity of constitutively active mutant NOTCH1. potential and intrinsic resistance to therapeutic agents are Increased penetrance of NOTCH1-induced leukemia from characteristic properties of HSCs, and transcriptional profil- Phf6-null cells may result from a priming effect resulting ing of human leukemia-initiating cell populations has estab- from increased chromatin accessibility in MYC target genes lished a link between the transcriptional programs operating and in genes involved in cell growth and protein biosynthesis, in HSCs and those present in leukemia stem cells (33). These programs that are tightly linked to T-ALL transformation observations are in agreement with data that support HSCs and directly controlled by NOTCH1 (25, 36, 37). In addition, representing the cell of origin of many human leukemias and and most notably, secondary deletion of Phf6 in NOTCH1- further support an overlap between the circuitries governing induced leukemia cells increased leukemia-initiating cell HSC self-renewal and those operating in leukemia-initiating activity, a phenotype linked to the effects of PHF6 inactiva- cell populations. tion in human leukemia by the enrichment in both mouse Phf6-null tumors and in PHF6-mutant human T-ALLs of a PHF6 Regulation of HSC Activity human leukemia-initiating cell transcriptional program. On Here, we identify PHF6 as a new leukemia-initiating tumor the basis of these observations, we propose that loss of PHF6 suppressor gene implicated in the control of long-term self- favors leukemia initiation by enhancing the self-renewal renewal in HSCs and as a driver of leukemia-initiating cell capacity of HSCs and lowering the threshold for oncogenic activity in T-ALL. Mutations that increase the self-renewal transformation in hematopoietic progenitors, and postulate capacity of hematopoietic progenitors are postulated to play a role for PHF6 mutations as early drivers of leukemia-initiat- important roles in leukemia initiation and as potential driv- ing cell activity in T-ALL.

Figure 6. Loss of Phf6 activates a leukemia-initiating cell (LIC) program and increases LIC frequency in T-ALL. A, Quantification of GFP+ leukemia cells in the peripheral blood of mice engrafted with Phf6f/YRosa26+/Cre-ERT2 wild-type and isogenic Phf6−/YRosa26+/Cre-ERT2 knockout leukemia cells on day 15 after transplantation (n = 15 per group). B, Kaplan–Meier survival curves of mice harboring Phf6 wild-type and isogenic Phf6 knockout NOTCH1-induced leukemias (n = 10 per group), as in A. C, LIC analysis in mice transplanted with Phf6f/YRosa26+/Cre-ERT2 wild-type and isogenic Phf6−/YRosa26+/Cre-ERT2 knockout leukemia cells (n = 6 per group). D, Confidence intervals showing 1/(stem cell frequency) based onC . E, Heat map representation of relative gene expression for top 50 ranking genes differentially expressed between Phf6f/YRosa26+/Cre-ERT2 wild-type and isogenic Phf6−/YRosa26+/Cre-ERT2 knockout NOTCH1-induced leukemia cells. F, Normalized gene expression of selected genes differentially expressed between isogenic wild-type and Phf6 knockout NOTCH1-induced leukemia cells. G, GSEA depicting enrichment of leukemia stem cell (LSC) signature genes in the expression signature associated with Phf6−/YRosa26+/Cre-ERT2 knockout NOTCH1-induced leukemia lymphoblast cells compared with Phf6f/YRosa26+/Cre-ERT2 wild-type isogenic controls. H, GSEA depicting enrichment of LSC signature genes in the expression signature associated with human PHF6-mutant T-ALL compared with matched PHF6 wild-type controls. Graphs in A depict measurements from individual mice with mean and standard deviation. P values were calculated using the two-tailed Student t test in A, the log-rank test in C, and the χ2 test in D.

OF11 | CANCER DISCOVERY March 2019 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

PHF6 in Hematopoietic Stem Cells and T-ALL RESEARCH ARTICLE

A B ) + 60 0.0003 P 100 f/Y 21 ± 5 GF 50 Phf6 −/Y 40 75 Phf6 30 50 20 31 ± 7 10 25

Percent survival Percent P = 0.013

Cell number 0 Peripheral blood (% 0 GFP f/Y /Y − 0 10 20 30 40 Phf6Phf6 Days after transplantation CD 0.0 P = 0.000558 Phf6 f/Y /Y −0.5 Phf6 − Confidence intervals for 1/(stem cell frequency) −1.0 Group Lower Estimate Upper −1.5 Phf6 f/Y R26+/Cre-ERT2 7,530 2,7561,009 2.0 − Phf6 −/Y R26+/Cre-ERT2 1,003 412 170 −2.5

Log fraction nonresponding 0.1 246810 Dose (number of cells ×104)

E f/Y −/Y F −08 −04 −06 −06 −05 −07 −07 80 150 300 300 1,500 6.9e Phf6 Phf6 5.7e 3.6e8.3e −05 −10 −06 −03 −08 6.3e6.7e4.4e Vopp1 250 250 1,250 Msantd3 60 4.9e8.0e 5.1e1.2e2.2e Amotl1 100 200 200 1,000 Csf2rb Clec12a Igf2bp3 40 150 150 750 Slc6a7 Flnc 50 100 100 500 Ccr9 20 App

Normalized counts 50 50 250 Fmnl2 Sorl1 Kif26b 0 0 0 0 0 Lmo2 Ptgs1 Cd34 Kit Cd300lf Flt3 Hex Pax5Cd19 FcYr1 Cd38 Csf3r Mef2cZeb2 Hif3a Serpine2 Syt14 Ankrd29 Tspan9 G Lbp 0.6 LSC signature Fes 0.5 Sort1 NES = 1.8 Nfix 0.4 P = < 1 × 10−4 Add3 0.3 B430306N03Rik Mtus1 0.2 Xist 0.1 Map3k9 Ptch2 0 Cpne2 −0.1

Napsa Enrichment score (ES) Dok3 Il18rap Phf6 KO Phf6 WT Hk3 Muc13 H Nsg1 LSC signature Fcgr2b 0.4 Gm30292 Rab44 0.3 NES = 1.6 Myo1f 0.2 P = 0.0005 C3 Efs 0.1 Abcd2 0 Igsf6 Hr −0.1 Phf6 −0.2 Crhbp Rpgr Enrichment score (ES) Oprk1 Phf6 KO Phf6 WT

Row minRow max

March 2019 CANCER DISCOVERY | OF12

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

RESEARCH ARTICLE Wendorff et al.

Open Questions Male CD45.1 mice (B6.SJL-Ptprca/BoyAiTac) were purchased from Taconic Farms (cat. #4007) and were used as recipients in non- Although the specific molecular functions of the PHF6 competitive transplantation experiments and as both bone mar- protein remain to be fully established (35), the association row donors and congenic recipients in competitive transplantation of PHF6 loss with increased chromatin accessibility warrants experiments. further evaluation of the role of PHF6 regulation of NuRD activity in HSCs and leukemia. Further studies will also char- Bone Marrow Reconstitution Experiments acterize the interaction of Phf6 inactivation with leukemia In noncompetitive bone marrow transplantation experiments, oncogenes other than NOTCH1 and evaluate the role and 0.5 million lineage-negative bone marrow cells isolated from Phf6−/Y mechanisms of Phf6 mutations in myeloid and mixed-lineage Rosa26+/Cre-ERT2 and Phf6+/YRosa26+/Cre-ERT2 bone marrow donor mice leukemias. Finally, analysis of Phf6 loss in the hematopoi- (CD45.2; 6–8 weeks old) 2 weeks after tamoxifen treatment were etic system of female mice will help clarify the mechanisms transplanted into the retro-orbital sinus of lethally irradiated (990 responsible for the observed male predominance of PHF6 Rad) congenic CD45.1 recipients. All experimental procedures were mutations in human leukemia. performed 8 to 10 weeks after bone marrow transplantation. Com- petitive transplants were performed using 0.5 million lineage- negative Phf6−/YRosa26+/Cre-ERT2 and Phf6+/YRosa26+/Cre-ERT2–derived METHODS bone marrow cells 2 weeks after tamoxifen treatment (as above) Patient Samples combined with 0.5 million lineage-negative bone marrow cells DNA from leukemic ALL blasts at diagnosis and relapse and derived from age-matched CD45.1 congenic mice. Lethally irradi- matched remission samples were provided by the Hemato-Oncol- ated (990 Rad) 7- to 8-week-old congenic CD45.1 male mice served ogy Laboratory at University of Padua, Italy, the Children’s Oncol- as primary mixed bone marrow recipients. Serial transplantation ogy Group, and the Eastern Cooperative Oncology Group (ECOG). was performed after 24 weeks by transplanting 3 million total Samples were collected under the supervision of local Institutional bone marrow cells from primary mixed bone marrow chimeras Review Boards for participating institutions and analyzed under the into secondary recipients, and from secondary mixed bone marrow supervision of the Columbia University Medical Center Institutional chimeras into tertiary recipients. Donor frequency and lineage com- Review Board (protocol number: IRB-AAAB3250). All samples were mitment were assessed every 4 weeks by flow-cytometric analysis of obtained with written informed consent at study entry. peripheral blood.

Mice and Animal Procedures Generation of NOTCH1-Induced Leukemia All animals were housed in specific pathogen-free facilities at the To generate NOTCH1-induced T-ALL tumors in mice, we per- Irving Cancer Research Center at the Columbia University Medical formed retroviral transduction of lineage negative–enriched bone Center in accordance with NIH guidelines for the Care and Use of marrow cells (Lineage Cell Depletion kit, Miltenyi Biotec; cat. #130- Laboratory Animals. All animal procedures were approved by the 090-858) with activated forms of the NOTCH1 oncogene (NOTCH1- Columbia University Institutional Animal Care and Use Committee. L1601P-ΔPEST) tagged with GFP and transplanted them into the To generate Phf6 conditional knockout mice, we used homologous lethally irradiated (990 Rad) isogenic recipients (40). T-ALL tumor + + + recombination in C57BL/6 embryonic stem cells to introduce a development was assessed by monitoring CD4 CD8 GFP cells in LoxP site upstream of exon 4 in introns 3 to 4, and the LoxP/FRT- peripheral blood by flow cytometry. flanked Neo selection cassette downstream of exon 5 in introns 5 To generate isogenic Phf6 wild-type and Phf6 knockout tumors, to 6. We generated chimeras in C57BL/6 albino blastocysts using we generated NOTCH1-L1601P-ΔPEST–induced leukemia using f/Y +/CreERT2 three independent targeted embryonic stem cell clones identified Phf6 Rosa26 donor cells as before (40). In all subsequent f/Y +/Cre-ERT2 by PCR analysis and verified by southern blot. We verified germline in vivo studies, 1 million Phf6 Rosa26 T-ALL tumor cells were transmission in the offspring of highly chimeric male mice crossed transplanted into primary sublethally irradiated (450 Rad) congenic with C57BL/6 females. To remove the neomycin selection cassette, we C57BL/6 recipients by retro-orbital injection. After 48 hours, mice crossed mice harboring the targeting construct with an Flp germline received either 3 mg tamoxifen or an equivalent volume of corn oil f/Y deleter line [Gt(ROSA)26Sortm1(FLP1)Dym/RainJ; The Jackson Laboratory] by intraperitoneal injection. Resulting isogenic Phf6 wild-type (Phf6 +/Cre-ERT2 −/Y +/Cre-ERT2 and crossed the resulting mice with C57BL/6 to breed out the Flp Rosa26 ) and Phf6 knockout (Phf6 Rosa26 ) tumor allele. To generate conditional Phf6 knockout mice, we bred animals cells recovered from primary recipient mice were transplanted via harboring the conditional targeted allele with Vav-iCre+/− animals (B6. retro-orbital injection into isogenic secondary recipients for analysis Cg-Commd10Tg(Vav1-icre)A2Kio/J; The Jackson Laboratory; ref. 38) expressing of tumor progression (10,000 tumor cells/recipient; n = 10–15 ani- the Cre recombinase under control of the pan-hematopoietic Vav1 onco- mals per group) and leukemia-initiating cell activity (10,000–1 tumor gene promoter. To generate conditional inducible Phf6 knockout mice, cells/recipient; n = 6 animals per group). we bred animals with Rosa26+/Cre-ERT2 mice [Gt(ROSA)26Sortm1(cre/ERT2)Thl], which express a tamoxifen-inducible form of the Cre recombinase Drug Treatments from the ubiquitous Rosa26 locus (39). Rosa26+/Cre-ERT2 mice were a Tamoxifen (cat. #T5648) and 5-FU (cat. #F6627) were purchased kind gift from T. Ludwig. Primers used for genotyping are provided from Sigma-Aldrich. Tamoxifen was dissolved in corn oil at 55°C in the Supplementary Methods section (Supplementary Table S1). to a stock concentration of 30 mg/mL and stored at −20°C. In all Phf6−/YVav-iCre and Phf6+/YVav-iCre male mice were used in experi- leukemia model experiments, a single 3-mg dose of tamoxifen in ments at 7 to 9 weeks of age. Phf6 inactivation in age-matched Phf6f/Y 100 μL was administered by intraperitoneal injection. Vehicle-treated Rosa26+/Cre-ERT2 and Phf6f/YRosa26+/+ control mice was induced at 5 animals received 100 μL of corn oil. 5-FU was dissolved in PBS at to 6 weeks of age by intraperitoneal administration of 1 mg/day 65°C to a final concentration of 15 mg/mL and administered by tamoxifen for 3 consecutive days. Mice were used in experiments a intraperitoneal injection at a dose of 150 mg/kg. A 1 mg/mL work- minimum of 8 weeks after tamoxifen treatment. ing solution of polyinosine-polycytidylic acid high-molecular-weight Male C57BL/6J mice (6 weeks old) were purchased from The Jackson [poly (I:C); Invivogen; cat. #11C21-MM] was prepared in physiologic Laboratory (cat. #000664) and used as recipients in all mouse leuke- saline solution (NaCL 0.9%) and was administered at a dose of 5 μg/g mia model experiments. body weight by intraperitoneal injection.

OF13 | CANCER DISCOVERY March 2019 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

PHF6 in Hematopoietic Stem Cells and T-ALL RESEARCH ARTICLE

Western Blotting 75-nucleotide-long reads per sample on an Illumina NextSeq 500. Western blot analysis was performed using standard protocols. BCL files were demultiplexed, and FASTQ files were generated on the Rabbit antibody against PHF6 (Sigma-Aldrich; cat. #HPA001023) BaseSpace platform (Illumina). Reads were trimmed of contaminat- was used at a 1:1,000 dilution, and rabbit anti-GAPDH antibody (Cell ing adapter sequences using cutadapt and aligned to the GRCM38- Signaling, cat. #2118) was used at a 1:10,000 dilution. build mouse genome using Bowtie2 in very sensitive mode. Peaks of transposase-accessible chromatin were called using MACS2 v2.1.1 Cell Isolation and Flow Cytometry Analysis (42) in SPMR mode on each sample independently and were subse- quently centered by symmetrically extending peak summits in the 5′ Single-cell suspensions of total bone marrow cells were prepared by and 3′ direction by 200 nucleotides. Overlapping peaks were merged crushing the long bones and passing cells through a 40-μm filter. For into single regions, and the set of all nonoverlapping peaks was used experiments involving HSC sorting, bone marrow cells were addition- for further analysis. We used the featureCounts method from the ally isolated from the illia and spinal vertebrae. Single-cell suspen- Rsubread v1.32.1 package to quantify the accessibility of each peak sions of soft tissues were obtained by pressing the tissue through a in all samples. Statistical analysis of differentially accessible regions 40-μm filter. All cell suspensions were prepared in PBS supplemented was performed using total library size normalization and a negative with 2% FCS, 1 mmol/L Hepes, and 0.5 μmol/L EDTA. Erythrocytes binomial model in R using the DESeq2 1.22.1 package (43). We anno- were eliminated using ACK lysing buffer. Absolute quantification of tated ATAC-seq accessible peaks according to their nearest genomic cell number was performed using CountBright absolute Counting feature using the annotatePeaks script from the HOMER package Beads (Invitrogen; cat. #MP36950). B-cell progenitor populations v4.10. analysis was also performed for all regions + − − + were defined as follows: pre–pro-B (B220 IgD CD19 ), pro-B (B220 with significantly increased accessibility using HOMER. Enrichment − − − − + − − − + IgD CD19 IgM CD25 ), pre-B (B220 IgD CD19 IgM CD25 ), of gene signatures was calculated using the hypergeometric distri- + − + + − immature B (B220 IgD CD19 IgM CD25 ), and total B cells bution. Genome-wide distribution of chromatin accessibility was + − (B220 IgD ). A full list of fluorochrome-conjugated anti-mouse plotted using the Circlize v0.4.4 package in R. Global proximal antibodies is included in the Supplementary Methods section (Sup- promoter accessibility was plotted with deeptools, 2 Kb 5′ and 3′ to plementary Table S2). (and centered at) the TSS of all protein-coding transcripts annotated Flow cytometry analysis was performed on a FACSCantoII, LSRII, using the RPGC normalized signal for all protein-coding genes in the and FortessaLSRII (BD Biosciences) and analyzed using FlowJo Tree- GRCm38.p6 comprehensive gene annotation from Gencode. star software (FlowJo LLC). Fluorescence-activated cell sorting was performed on a BD Influx cell sorter. RNA-seq and Gene-Expression Profiling

Cell-Cycle Analysis For RNA-seq analysis of isogenic Phf6 wild-type and knockout mouse T-ALL cells, RNA was extracted from 106 cells (n = 3 per After staining of surface antigens, bone marrow cells were fixed group) using the RNeasy Mini Kit (Qiagen; cat. #74106). cDNA using the Cytofix Fixation/Permeabilization Kit (BD Biosciences; cat. synthesis and subsequent library synthesis were performed using #554714) according to the manufacturer’s instructions. Cells were the NEBNext Ultra RNA Library Prep Kit for Illumina (New Eng- stained with anti-Ki67 antibody (1:30 dilution) overnight on ice. One land Biolabs; cat. #E7530). For RNA-seq analysis of Phf6 wild-type hour prior to analysis, cells were incubated with 1.62 μmol/L Hoechst and knockout LT-HSCs, cells were sorted directly into buffer RLT 33342 (Invitrogen; cat. #H3570) at room temperature. Acquisition was Plus, and total RNA was isolated using the RNeasy Plus Micro Kit performed using a BD LSRII (BD Biosciences) flow cytometry analyzer. (Qiagen; cat. #74034). First-strand cDNA synthesis and full-length ds cDNA amplification were performed using the SMART-Seq v4 Mobilization Protocol Ultra Low Input RNA Kit for Sequencing (Clontech Laboratories; Mice were injected intraperitoneally with 200 mg/kg of cyclo- cat. #634888). Sequencing-ready libraries were generated using the phosphamide (Cy; Calbiochem; cat. #239785) and then on succeed- Low Input Library Prep Kit v2 (Clontech Laboratories; cat. #634899). ing days with 5 μg/day recombinant human G-SCF (Amgen; cat. The Agencourt AMPure XP kit was used to purify cDNA (Beckman #55513-924-10) administered by subcutaneous injection. The day of Coulter; cat. #A63880). Libraries were quantified using the Qubit cyclophosphamide treatment was considered day −1 and the first day dsDNA HS Assay Kit (Invitrogen; cat. #Q32854) and by quantitative of G-CSF treatment was counted as day 0 (e.g., mice sacrificed on day PCR using KAPA Library Quantification Kit (Kapa Biosystems; cat. 4 of the mobilization protocol were sacrificed on the day after the #KR0405). Libraries were pooled in equimolar concentrations and fourth G-CSF injection, designated as D4). sequenced on an Illumina NextSeq 500 to a total target depth of 10 million 150-bp paired-end reads per sample. All protocols were per- Cell Sorting formed following the manufacturer’s instructions. Raw reads were demultiplexed, and FASTQ files were generated LT-HSCs (defined here as Lin− Sca1+ CD117+ CD135− CD150+ CD48−) using bcl2fastq v2.20.0.422 (Illumina). Reads were aligned to a hybrid were sorted from lineage-depleted bone marrow cells isolated from mouse (GRCm38.p6)-ERCC genome using STAR v2.3.5.3a, and gene- wild-type (Phf6f/YRosa26+/+) and Phf6 knockout (Phf6−/YRosa26+/Cre-ERT2) count matrices were computed using featureCounts v1.6.0 (Rsub- mice 8 weeks after tamoxifen treatment. Cell sorting was performed read). The removal of unwanted variance (RUV) method (44) was using a BD Influx cell sorter (BD Biosciences). used to normalize based on the synthetic spike-in controls (Ambion; ATAC Sequencing cat. #4456740) in RUVseq v1.14.0. Differential expression analysis was performed using the negative binomial distribution to estimate ATAC sequencing was performed as previously described (41) the variance-mean dependence and to test for statistical significance in 20,000 LT-HSCs sorted from three individual knockout Phf6 in DESeq2 v1.20.0 (43). (Phf6−/Y Rosa26+/Cre-ERT2) and Phf6 wild-type (Phf6f/Y Rosa26+/+) mice 8 weeks after tamoxifen treatment. Nuclei were isolated and tag- mented using Nextera Tn5 Transposase with single indexed, library- Gene Set Enrichment Analysis specific sequencing adapters. ATAC-seq libraries were quantified Gene set enrichment analysis (45) was performed using regularized based on the average fragment size according to Bioanalyzer results, log-transformed normalized count data in the javaGSEA program pooled at equimolar concentrations, and sequenced as a pool to (Broad Institute). We used the Jak1 mouse knockout hematopoietic a total read-depth target of approximately 7 × 107, paired-end, stem cell signature gene set from ref. 18 and the human 128-gene

March 2019 CANCER DISCOVERY | OF14

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

RESEARCH ARTICLE Wendorff et al. leukemia-initiating cell signature gene set from ref. 31. For analysis of Administrative, technical, or material support (i.e., reporting mouse tumors, human gene identifiers were converted to mouse in R or organizing data, constructing databases): A.A. Wendorff, using the biomaRt bioconductor package. Analysis of human T-ALL S.A. Quinn, A. Ambesi-Impiombato, E. Paietta tested the enrichment of the leukemia-initiating signature in PHF6 Study supervision: R. Rabadan, A.A. Ferrando wild-type (n = 5) versus PHF6-mutant (n = 5) leukemias (46). Analysis was restricted to tumors in the TLX1 and TLX3 group to avoid the Acknowledgments confounding effect of the association of TLX1/TLX3 translocation and This work was supported by the NIH grants R35 CA210065 (A.A. overexpression with PHF6 mutations (1). Enrichment scores were com- Ferrando), R01 CA155743 (A.A. Ferrando), CA180827 (E. Paietta), puted, and 10,000 gene permutations were used to estimate P values. CA196172 (E. Paietta), U54 CA193313 (R. Rabadan), R01 CA185486 (R. Rabadan), U54 CA209997 (R. Rabadan), CA180820 (ECOG-ACRIN), Data Availability CA189859 (ECOG-ACRIN), CA14958 (ECOG-ACRIN), CA180791 RNA-seq and ATAC-seq data sets generated during this study, (ECOG-ACRIN), CA17145 (ECOG-ACRIN), U10 CA180827 (ECOG- cited RNA-seq data sets, and microarray data sets are available in the ACRIN), U10 CA98543 (J.M. Gastier-Foster and M.L. Loh); the Human Gene Expression Omnibus under accession numbers GSE117165, Specimen Banking Grant U24 CA114766 (J.M. Gastier-Foster), and P30 GSE85745, and GSE42328, respectively. CA013696 (in support of the Herbert Irving Comprehensive Cancer Center Genomics, Flow Cytometry and Transgenic Mouse Shared ISN of T-ALL Resources). A.A. Wendorff was supported by a Rally Foundation fel- lowship. P. Van Vlierberghe was supported by the Fund for Scientific We illustrated the sequential order of somatic mutations in T-ALL Research (FWO) Flanders (postdoctoral fellowship and Odysseus type using the ISN, which pools evolutionary paths across all patients. We 2 grant). M. Rashkovan was supported by a Damon-Runyon Sohn selected recurrently mutated genes that were previously defined as Pediatric Cancer fellowship. We are grateful to T. Ludwig (The Ohio drivers of pediatric T-ALL (27) and relapse-specific genes. Synonymous State University Comprehensive Cancer Center) for the Rosa26+/Cre-ERT2 single-nucleotide variants were not included in the analysis. For each mouse. We thank E. Passegué for insightful suggestions in the experi- patient, we generated a sequential network that defined early events mental design and interpretation of our results. as mutations observed in both primary tumor and relapsed tumor. Each node represents a gene, and each arrow points from a gene The costs of publication of this article were defrayed in part by with an early event to a gene with a late event. The ISN then pooled the payment of page charges. This article must therefore be hereby sequential networks across all patients. To test whether a gene within marked advertisement in accordance with 18 U.S.C. Section 1734 the ISN was significantly early or late, we used the binomial test based solely to indicate this fact. on in-degree and out-degree of each node. Specifically, thePHF6 P = 0.03 corresponds to the binomial cumulative distribution function for Received August 29, 2018; revised November 29, 2018; accepted x = 0 arrows going into PHF6, n = 5 total trials, and probability P = 0.5 December 13, 2018; published first December 19, 2018. that any arrow will point toward PHF6 [binocdf(x = 0, N = 4, P = 0.5)].

Statistical Analyses References Statistical analysis was performed using GraphPad Prism software . 1 Van Vlierberghe P, Palomero T, Khiabanian H, Van der Meulen J, v5.0 (GraphPad Software). We assumed normality and equal distribu- Castillo M, Van Roy N, et al. PHF6 mutations in T-cell acute lympho- tion of variance between groups, and we considered results with Stu- blastic leukemia. Nat Genet 2010;42:338–42. dent t test P < 0.05 as statistically significant. Kaplan–Meier curves 2. Van Vlierberghe P, Patel J, Abdel-Wahab O, Lobry C, Hedvat CV, were used to represent survival, where significance was calculated Balbin M, et al. PHF6 mutations in adult acute myeloid leukemia. with the log-rank test. We analyzed serial limited dilution leukemia- Leukemia 2011;25:130–4. initiating cell data using the ELDA online resource software (47). 3. Patel JP, Gonen M, Figueroa ME, Fernandez H, Sun Z, Racevskis J, et al. Prognostic relevance of integrated genetic profiling in acute Disclosure of Potential Conflicts of Interest myeloid leukemia. N Engl J Med 2012;366:1079–89. 4. Lower KM, Turner G, Kerr BA, Mathews KD, Shaw MA, Gedeon AK, J.M. Gastier-Foster reports receiving commercial research grants et al. Mutations in PHF6 are associated with Borjeson-Forssman- from Bristol-Myers Squibb and Incyte Corporation. M.L. Loh is Lehmann syndrome. Nat Genet 2002;32:661–5. a consultant/advisory board member for Medisix Therapeutics. 5. Todd MA, Picketts DJ. PHF6 interacts with the nucleosome remod- R. Rabadan is a consultant/advisory board member for CNIO. No eling and deacetylation (NuRD) complex. J Proteome Res 2012;11: potential conflicts of interest were disclosed by the other authors. 4326–37. 6. Torchy MP, Hamiche A, Klaholz BP. Structure and function insights Authors’ Contributions into the NuRD chromatin remodeling complex. Cell Mol Life Sci Conception and design: A.A. Wendorff, A.A. Ferrando 2015;72:2491–507. Development of methodology: A.A. Wendorff, A. Ambesi-Impiombato, 7. Zhang C, Mejia LA, Huang J, Valnegri P, Bennett EJ, Anckar J, et al. R. Rabadan The X-linked intellectual disability protein PHF6 associates with the PAF1 complex and regulates neuronal migration in the mammalian Acquisition of data (provided animals, acquired and managed brain. Neuron 2013;78:986–93. patients, provided facilities, etc.): A.A. Wendorff, A. Ambesi-Impi- 8. Zhang Y, Smith ADt, Renfrow MB, Schneider DA. The RNA polymer- ombato, M.S. Tallman, E. Paietta, M. Paganin, G. Basso, J.M. Gastier- ase-associated factor 1 complex (Paf1C) directly increases the elonga- Foster, M.L. Loh, P. Van Vlierberghe tion rate of RNA polymerase I and is required for efficient regulation Analysis and interpretation of data (e.g., statistical analysis, of rRNA synthesis. J Biol Chem 2010;285:14152–9. biostatistics, computational analysis): A.A. Wendorff, S.A. Quinn, 9. Wang J, Leung JW, Gong Z, Feng L, Shi X, Chen J. PHF6 regulates M. Rashkovan, C.J. Madubata, A. Ambesi-Impiombato, M.R. Litzow, cell cycle progression by suppressing ribosomal RNA synthesis. J Biol R. Rabadan Chem 2013;288:3174–83. Writing, review, and/or revision of the manuscript: A.A. Wen- 10. Alexander TB, Gu Z, Iacobucci I, Dickerson K, Choi JK, Xu B, et al. dorff, S.A. Quinn, M.R. Litzow, M.S. Tallman, J.M. Gastier-Foster, The genetic basis and cell of origin of mixed phenotype acute leukae- M.L. Loh, A.A. Ferrando mia. Nature 2018;562:373–9.

OF15 | CANCER DISCOVERY March 2019 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

PHF6 in Hematopoietic Stem Cells and T-ALL RESEARCH ARTICLE

.11 Zhang J, Ding L, Holmfeldt L, Wu G, Heatley SL, Payne-Turner D, 30. Ng SW, Mitchell A, Kennedy JA, Chen WC, McLeod J, Ibrahimova et al. The genetic basis of early T-cell precursor acute lymphoblastic N, et al. A 17-gene stemness score for rapid determination of risk in leukaemia. Nature 2012;481:157–63. acute leukaemia. Nature 2016;540:433–7. 12. Chao MM, Todd MA, Kontny U, Neas K, Sullivan MJ, Hunter 31. Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van AG, et al. T-cell acute lymphoblastic leukemia in association with Galen P, et al. Stem cell gene expression programs influence clinical Borjeson-Forssman-Lehmann syndrome due to a mutation in PHF6. outcome in human leukemia. Nat Med 2011;17:1086–93. Pediatr Blood Cancer 2010;55:722–4. 32. Wang JC, Dick JE. Cancer stem cells: lessons from leukemia. Trends 13. Yoshizato T, Dumitriu B, Hosokawa K, Makishima H, Yoshida K, Cell Biol 2005;15:494–501. Townsley D, et al. Somatic mutations and clonal hematopoiesis in 33. Vedi A, Santoro A, Dunant CF, Dick JE, Laurenti E. Molecular land- aplastic anemia. N Engl J Med 2015;373:35–47. scapes of human hematopoietic stem cells in health and leukemia. 14. Abelson S, Collord G, Ng SWK, Weissbrod O, Mendelson Cohen Ann N Y Acad Sci 2016;1370:5–14. N, Niemeyer E, et al. Prediction of acute myeloid leukaemia risk in 34. Bowman RL, Busque L, Levine RL. Clonal hematopoiesis and evolu- healthy individuals. Nature 2018;559:400–4. tion to hematopoietic malignancies. Cell Stem Cell 2018;22:157–70. 15. Landau DA, Clement K, Ziller MJ, Boyle P, Fan J, Gu H, et al. Locally 35. Todd MA, Ivanochko D, Picketts DJ. PHF6 degrees of separation: disordered methylation forms the basis of intratumor methylome var- the multifaceted roles of a chromatin adaptor protein. Genes 2015;6: iation in chronic lymphocytic leukemia. Cancer Cell 2014;26:813–25. 325–52. 16. Pang WW, Schrier SL, Weissman IL. Age-associated changes in human 36. Palomero T, Lim WK, Odom DT, Sulis ML, Real PJ, Margolin A, et al. hematopoietic stem cells. Semin Hematol 2017;54:39–42. NOTCH1 directly regulates c-MYC and activates a feed-forward-loop 17. Elias HK, Bryder D, Park CY. Molecular mechanisms underlying line- transcriptional network promoting leukemic cell growth. Proc Natl age bias in aging hematopoiesis. Semin Hematol 2017;54:4–11. Acad Sci U S A 2006;103:18261–6. 18. Kleppe M, Spitzer MH, Li S, Hill CE, Dong L, Papalexi E, et al. Jak1 37. Margolin AA, Palomero T, Sumazin P, Califano A, Ferrando AA, integrates cytokine sensing to regulate hematopoietic stem cell func- Stolovitzky G. ChIP-on-chip significance analysis reveals large-scale tion and stress hematopoiesis. Cell Stem Cell 2018;22:277. binding and regulation by human oncogenes. 19. Walter D, Lier A, Geiselhart A, Thalheimer FB, Huntscha S, Sobotta Proc Natl Acad Sci U S A 2009;106:244–9. MC, et al. Exit from dormancy provokes DNA-damage-induced attri- 38. de Boer J, Williams A, Skavdis G, Harker N, Coles M, Tolaini M, et al. tion in haematopoietic stem cells. Nature 2015;520:549–52. Transgenic mice with hematopoietic and lymphoid specific expres- 20. Pietras EM, Lakshminarasimhan R, Techner JM, Fong S, Flach J, sion of Cre. Eur J Immunol 2003;33:314–25. Binnewies M, et al. Re-entry into quiescence protects hematopoietic 39. Guo K, McMinn JE, Ludwig T, Yu YH, Yang G, Chen L, et al. Dis- stem cells from the killing effect of chronic exposure to type I inter- ruption of peripheral leptin signaling in mice results in hyperlep- ferons. J Exp Med 2014;211:245–62. tinemia without associated metabolic abnormalities. Endocrinology 21. Ferrando AA, Lopez-Otin C. Clonal evolution in leukemia. Nat Med 2007;148:3987–97. 2017;23:1135–45. 40. Herranz D, Ambesi-Impiombato A, Sudderth J, Sanchez-Martin M, 22. Randall TD, Weissman IL. Phenotypic and functional changes Belver L, Tosello V, et al. Metabolic reprogramming induces resistance induced at the clonal level in hematopoietic stem cells after 5-fluoro- to anti-NOTCH1 therapies in T cell acute lymphoblastic leukemia. uracil treatment. Blood 1997;89:3596–606. Nat Med 2015;21:1182–9. 23. Morrison SJ, Wright DE, Weissman IL. Cyclophosphamide/granulo- 41. Buenrostro JD, Wu B, Chang HY, Greenleaf WJ. ATAC-seq: a method cyte colony-stimulating factor induces hematopoietic stem cells to for assaying chromatin accessibility genome-wide. Curr Protoc Mol proliferate prior to mobilization. Proc Natl Acad Sci U S A 1997;94: Biol 2015;109:21.9.1–9. 1908–13. 42. Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein 24. Weng AP, Ferrando AA, Lee W, Morris JPt, Silverman LB, Sanchez- BE, et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol Irizarry C, et al. Activating mutations of NOTCH1 in human T cell 2008;9:R137. acute lymphoblastic leukemia. Science 2004;306:269–71. 43. Love MI, Huber W, Anders S. Moderated estimation of fold change 25. Herranz D, Ambesi-Impiombato A, Palomero T, Schnell SA, Belver and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15: L, Wendorff AA, et al. A NOTCH1-driven MYC enhancer promotes 550. T cell development, transformation and acute lymphoblastic leuke- 44. Risso D, Ngai J, Speed TP, Dudoit S. Normalization of RNA-seq data mia. Nat Med 2014;20:1130–7. using factor analysis of control genes or samples. Nat Biotechnol 26. Soto-Feliciano YM, Bartlebaugh JME, Liu Y, Sanchez-Rivera FJ, 2014;32:896–902. Bhutkar A, Weintraub AS, et al. PHF6 regulates phenotypic plasticity 45. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, through chromatin organization within lineage-specific genes. Genes Gillette MA, et al. Gene set enrichment analysis: a knowledge-based Dev 2017;31:973–89. approach for interpreting genome-wide expression profiles. Proc Natl 27. Belver L, Ferrando A. The genetics and mechanisms of T cell acute Acad Sci U S A 2005;102:15545–50. lymphoblastic leukaemia. Nat Rev Cancer 2016;16:494–507. 46. Van Vlierberghe P, Ambesi-Impiombato A, De Keersmaecker K, 28. Coustan-Smith E, Mullighan CG, Onciu M, Behm FG, Raimondi SC, Hadler M, Paietta E, Tallman MS, et al. Prognostic relevance of inte- Pei D, et al. Early T-cell precursor leukaemia: a subtype of very high- grated genetic profiling in adult T-cell acute lymphoblastic leukemia. risk acute lymphoblastic leukaemia. Lancet Oncol 2009;10:147–56. Blood 2013;122:74–82. 29. Van Vlierberghe P, Ambesi-Impiombato A, Perez-Garcia A, Haydu JE, 47. Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for com- Rigo I, Hadler M, et al. ETV6 mutations in early immature human paring depleted and enriched populations in stem cell and other T cell leukemias. J Exp Med 2011;208:2571–9. assays. J Immunol Methods 2009;347:70–8.

March 2019 CANCER DISCOVERY | OF16

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst December 19, 2018; DOI: 10.1158/2159-8290.CD-18-1005

Phf6 Loss Enhances HSC Self-Renewal Driving Tumor Initiation and Leukemia Stem Cell Activity in T-ALL

Agnieszka A. Wendorff, S. Aidan Quinn, Marissa Rashkovan, et al.

Cancer Discov Published OnlineFirst December 19, 2018.

Updated version Access the most recent version of this article at: doi:10.1158/2159-8290.CD-18-1005

Supplementary Access the most recent supplemental material at: Material http://cancerdiscovery.aacrjournals.org/content/suppl/2018/12/19/2159-8290.CD-18-1005.DC1

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerdiscovery.aacrjournals.org/content/early/2019/02/12/2159-8290.CD-18-1005. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerdiscovery.aacrjournals.org on October 3, 2021. © 2018 American Association for Cancer Research.