Many of the clinical manifestations of leukemia, including infections, anemia and hemorrhage, reflect a progressive disruption of normal blood cell development. While chronic lymphoid leukemia impairs the function of hematopoietic stem cells (HSC),1 acute myeloid leukemia2 and T-lineage acute lymphoblastic leukemia (T-ALL)3 primarily target lineage-restricted progenitor compartments. Data obtained from a xenograft model of B-lineage acute lymphoblastic leukemia (B-ALL) suggested that leukemia expansion results in a reduced expression of the chemokine (C-X-C motif) ligand 12 (CXCL12), causing defective HSC homing and reduction of CD34+ cell populations in the bone marrow (BM).4 However, single-cell RNA sequencing from a mouse B-ALL model indicated a substantial presence of cells with the RNA expression pattern of HSC in the BM even at advanced stages of leukemia.5 Thus, the impact of B-ALL on HSC function and residency in the BM remains unclear.
We have developed a murine B-ALL model based on transplantation of primary tumor cells from mice carrying combined heterozygote mutations in the Pax5 and Ebf1 genes.6 Pax5 and Ebf1 encode two transcription factors frequently targeted in human B-ALL and a significant proportion of the trans-heterozygote mice develop monoclonal or oligoclonal B-ALL before 40 weeks of age.6 To determine the impact of B-ALL progression on hematopoiesis, we transplanted 150,000 primary mouse tumor cells from the lymph node of a leukemic Cd45.2+/+Ebf1+/–Pax5+/– mouse to wildtype CD45.1 mice by tail vein injection (Figure 1A). The transplants were performed without pre-conditioning allowing us to determine the impact of B-ALL on steady-state hematopoiesis (Figure 1B). Disease was manifested by palpable accessory axillary or subiliac lymph nodes 3-4 weeks after transplantation. At this time the tumor burden in the BM of the transplanted mice was over 90% (Figure 1C) and the absolute numbers of CD45.1 cells in the BM reduced (Online Supplementary Figure S1A). To determine the composition of the remaining BM population, the lineage negative (Lin–)CD45.1+KIT+ host populations were divided into lineage-restricted Lin–KIT+SCA1–, (LK) progenitors and Lin–KIT+SCA1+ (LSK) multipotent progenitors including HSC (Figure 1D). Sub-fractionation of the LK compartment7 (Figure 1D) revealed 10- to 100-fold reductions in both relative and absolute numbers of myeloid lineage-restricted progenitors in tumor transplanted mice as compared to the numbers in sham transplanted control mice (Figure 1E).
This included the pre-granulocyte/monocytes, granulocyte/monocyte progenitors, pre-megakaryocyte/erythrocytes, megakaryocyte progenitors, pre-colony-forming unit erythrocytes and colony-forming unit erythrocytes (Figure 1E). The multipotent LSK population was fractionated into CD150– short-term and CD150+ long-term HSC8 (Figure 1D, E). The frequency as well as absolute numbers of LSK were reduced in leukemic mice (Figure 1E). However, even though the absolute numbers of CD150+ HSC were reduced 1.6fold, the relative frequency of the most primitive progenitors was increased. Transplantation of two other independently generated pro-B-ALL tumors generated similar results as we detected reduced frequency of lineage restricted progenitors but no reduced frequency of CD150+LSK in mice transplanted with tumor (Online Supplementary Figure S1B-D).
To further analyze the cellular composition of the leukemic BM we performed single-cell RNA-sequencing analysis of Lin–KIT+CD45.1+ cells in control and leukemic mice (GSE207819). The data were processed using Seurat whereby the control and leukemic samples were integrated using Harmony prior to clustering. The cells were assigned an identity using a previously generated data set from hematopoietic progenitors9 (Online Supplementary Figure S2A, B) employing SingleR. This revealed an 18-fold enrichment of HSC and a 5-fold reduction in megakaryocyte/erythroid progenitors in the BM from tumor-exposed mice as compared to the numbers in control mice (Online Supplementary Figure S2C). Even though the single-cell RNA-sequencing analysis did not quite recapitulate the dramatic loss of linage-restricted progenitors detected by fluorescence activated cell sorting analysis, the data support the idea that the HSC compartment is relatively well conserved while the frequencies of lineage-restricted progenitors are reduced in B-ALL.
To determine the status of the progenitor compartments in B-ALL patients, we analyzed flow cytometry data from the BM of 19 patients diagnosed with B-ALL (median age, 6.5; range, 0-59 years).10 Cells were gated on high expression of CD34, a marker for early progenitor cells,11 and lack of CD19, a defining B-ALL marker (Figure 2A). The CD34+ population was gated negative for CD20 and CD66 (Online Supplementary Figure S3) and was highly heterogeneous for expression of CD38 (Figure 2A). This allowed us to investigate leukemia-associated changes in the ratios between CD38high cells, reported to be largely lineage-restricted progenitors, and CD38low/– progenitors, enriched for multipotent HSC.11 While the relative frequency of CD34+ cells was reduced in the patients’ BM as compared to control samples (Figure 2B), the frequency of CD34+CD38low/– cells was not significantly different. Re-analysis of singlecell RNA-sequencing data from leukemia patients12 using SingleR as above provided additional support for the fact that the frequency of HSC was not reduced in leukemia patients (Figure 2C). Analyzing BM samples from B-ALL patients at diagnosis and at days 15, 29 and 78-79 after initiation of treatment, according to the NOPHO2008 protocol,13 revealed a rapid reduction in the frequency of CD19+ cells (Figure 2D), and increased abundance of CD34+CD38+ lineage-restricted progenitors (Figure 2E). The frequency of CD34+CD38–/low cells did not change to a significant degree throughout the treatment (Figure 2F). These data reveal that the earliest progenitor compartments are well preserved even in advanced stages of disease and the rapid recovery after initiation of treatment suggests that they retain a substantial functional capacity for blood cell production.
To determine the functional capacity of leukemia-exposed HSC, we sorted CD150+LSK from leukemic and control CD45.1 mice and transplanted 250 cells into lethally irradi ated (9 Gy) CD45.2 mice together with 200,000 CD45.2 BM support cells (Figure 3A). Analysis of peripheral blood 4 and 8 weeks after transplantation identified a comparable fraction of CD45.1+ cells generated from both control and leukemia-exposed HSC (Figure 3B). We were unable to detect any significant lineage bias as both groups of mice presented comparable ratios of CD19+, CD3+ and GR1/MAC1+ cells (Figure 3B). Seventeen weeks after transplantation, we noted a tendency towards a reduced level of reconstitution in mice transplanted with B-ALL-exposed HSC (Figure 3B). To determine whether the phenotypic stem cells retained long-term reconstitution potential, we performed serial transplantation of total BM from the mice reconstituted with control or leukemia-exposed HSC into lethally irradiated CD45.2 mice and followed the generation of CD45.1+ cells (Figure 3C). Even though the lineage distribution was comparable between the two groups of serially transplanted mice, the reconstitution level was significantly reduced, already 4 weeks after transplantation, in mice that obtained BM containing tumor-exposed hematopoietic progenitors (Figure 3C). This effect became even more prominent at 16 weeks after the secondary transplantation and upon analysis of CD45.1+ progenitors in the BM we detected a relative reduction in frequency of all progenitor populations, including the CD150+LSK compartment, in mice transplanted with BM containing leukemia-exposed HSC (Figure 3D). Hence, even though exposure to leukemia does not have a major impact on the presence of HSC in the BM, the remaining stem cells display a defective ability for long-term reconstitution.
To identify B-ALL-induced changes in gene expression patterns, we compared RNA levels in cells defined as part of a given population in the SingleR analysis (Online Supplementary Figure S2) in mice injected with phosphate-buffered saline or leukemia (Figure 1A). These changes were mined for significantly enriched gene ontology terms using ToppCluster (https://toppcluster.cchmc.org/) and displayed in a heatmap. A major part of the gene ontology terms were related to mitochondrial function and electron transport (Online Supplementary Figure S2D, E). To determine the total mitochondria content in progenitor cells we stained HSC from control and leukemic mice (Figure 1A) with Mitotracker green. While CD150+LSK contained a slightly higher mitochondria mass in leukemic as compared to control mice, no such difference was found in the CD150–LSK population (Figure 3E). To compare mitochondrial membrane potential in control and leukemia exposed cells, we stained the LSK cells with Mitrotracker CMXRos (MTR). The retention of MTR was reduced in both the CD150+ and CD150– cells from the leukemic BM as compared to control cells (Figure 3F) indicating that the B-ALL-exposed cells harbor a deficiency in mitochondria function.
The impact of leukemia on normal blood cell development is reported to involve displacement as well as changes in the microenvironment causing disruptions in stem and progenitor cell function.1-4 Our data show that even though the phenotypic HSC are well preserved in the BM in B-ALL, the cells display a defect limiting their ability for long-term reconstitution. As we were unable to detect pathological expansion of CD45.2+CD19+ cells or symptoms of leukemia in the mice transplanted with stem cells, we believe that the functional defect is HSC autonomous and not a consequence of contaminating leukemia cells. Furthermore, in contrast to our observation in leukemic mice (Figure 1E), the stem cell-transplanted animals show a clear reduction in the frequency of HSC (Figure 3D). The functional impairment was associated with defective mitochondria function in a manner resembling that observed in aged HSC14 likely arising from cellular stress in the leukemic BM. Even if such a defect would not have an impact on the initial recovery of blood cell production after treatment of a leukemia patient, it could potentially contribute to late effects such as clonal hematopoiesis linked to primary hematologic malignancies.15 We believe that our findings open an interesting path for further studies of the underlying causes of secondary cancers and long-term medical conditions in leukemia patients.
Footnotes
- Received November 21, 2022
- Accepted February 22, 2023
Correspondence
Disclosures
No conflicts of interest to disclose.
Contributions
CJ, JÅ, AP, JTG, RS, JU, and MS designed, conducted and analyzed the experiments. SS, SL, and CJ performed bio-informatics analysis of data. All authors contributed to writing the manuscript.
Data-sharing statement
The RNA-sequencing data generated are deposited in GEO (GSE207819). Excel files with FACS data from mouse experiments will be shared upon request. Detailed materials and methods will be provided upon request.
Funding
Acknowledgments
We thank Maria Malmberg, Liselotte Lenner and Linda Bergström, the CTG and the SCC FACS core facility at Lund University for expert technical assistance. We also thank Dr David Bryder for his critical reading of the manuscript.
References
- Kikushige Y, Ishikawa F, Miyamoto T. Self-renewing hematopoietic stem cell is the primary target in pathogenesis of human chronic lymphocytic leukemia. Cancer Cell. 2011; 20(2):246-259. https://doi.org/10.1016/j.ccr.2011.06.029PubMedGoogle Scholar
- Miraki-Moud F, Anjos-Afonso F, Hodby KA. Acute myeloid leukemia does not deplete normal hematopoietic stem cells but induces cytopenias by impeding their differentiation. Proc Natl Acad Sci U S A. 2013; 110(33):13576-135681. https://doi.org/10.1073/pnas.1301891110PubMedPubMed CentralGoogle Scholar
- Hu X, Shen H, Tian C. Kinetics of normal hematopoietic stem and progenitor cells in a Notch1-induced leukemia model. Blood. 2009; 114(18):3783-3792. https://doi.org/10.1182/blood-2009-06-227843PubMedPubMed CentralGoogle Scholar
- Colmone A, Amorim M, Pontier AL, Wang S, Jablonski E, Sipkins DA. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science. 2008; 322(5909):1861-1865. https://doi.org/10.1126/science.1164390PubMedGoogle Scholar
- Anderson D, Skut P, Hughes AM. The bone marrow microenvironment of pre-B acute lymphoblastic leukemia at single-cell resolution. Sci Rep. 2020; 10(1):19173. https://doi.org/10.1038/s41598-020-76157-4PubMedPubMed CentralGoogle Scholar
- Prasad MA, Ungerback J, Ahsberg J. Ebf1 heterozygosity results in increased DNA damage in pro-B cells and their synergistic transformation by Pax5 haploinsufficiency. Blood. 2015; 125(26):4052-4059. https://doi.org/10.1182/blood-2014-12-617282PubMedPubMed CentralGoogle Scholar
- Pronk C, Rossi D, Månsson R. Elucidation of the phenotype, functional, and molecular topography of a myeloerythroid progenitor cell hierarchy. Cell Stem Cell. 2007; 1(4):428-442. https://doi.org/10.1016/j.stem.2007.07.005PubMedGoogle Scholar
- Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005; 121(7):1109-1121. https://doi.org/10.1016/j.cell.2005.05.026PubMedGoogle Scholar
- Nestorowa S, Hamey FK, Pijuan Sala B. A single-cell resolution map of mouse hematopoietic stem and progenitor cell differentiation. Blood. 2016; 128(8):20-31. https://doi.org/10.1182/blood-2016-05-716480PubMedPubMed CentralGoogle Scholar
- Modvig S, Hallbook H, Madsen HO. Value of flow cytometry for MRD-based relapse prediction in B-cell precursor ALL in a multicenter setting. Leukemia. 2021; 35(7):1894-1906. https://doi.org/10.1038/s41375-020-01100-5PubMedPubMed CentralGoogle Scholar
- Notta F, Zandi S, Takayama N. Distinct routes of lineage development reshape the human blood hierarchy across ontogeny. Science. 2016; 351(6269):aab2116. https://doi.org/10.1126/science.aab2116PubMedPubMed CentralGoogle Scholar
- Witkowski MT, Dolgalev I, Evensen NA. Extensive remodeling of the immune microenvironment in B cell acute lymphoblastic leukemia. Cancer Cell. 2020; 37(6):867-882. https://doi.org/10.1016/j.ccell.2020.04.015PubMedPubMed CentralGoogle Scholar
- Toft N, Birgens H, Abrahamsson J. Results of NOPHO ALL2008 treatment for patients aged 1-45 years with acute lymphoblastic leukemia. Leukemia. 2018; 32(3):606-615. https://doi.org/10.1038/leu.2017.265PubMedGoogle Scholar
- Ho YH, Del Toro R, Rivera-Torres J. Remodeling of bone marrow hematopoietic stem cell niches promotes myeloid cell expansion during premature or physiological aging. Cell Stem Cell. 2019; 25(3):407-418. https://doi.org/10.1016/j.stem.2019.06.007PubMedPubMed CentralGoogle Scholar
- Jaiswal S, Fontanillas P, Flannick J. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014; 371(26):2488-2498. https://doi.org/10.1056/NEJMoa1408617PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.