11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Reduced CXCL4/PF4 expression as a driver of increased human hematopoietic stem and progenitor cell proliferation in polycythemia vera

      letter

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Dear Editor, Polycythemia vera (PV) is a myeloproliferative neoplasm (MPN) marked by hyperproliferation of all myeloid cell lineages and characterized by activated JAK-STAT signaling due to an activating mutation in JAK2 1 . Disease-driving pathogenic changes in MPNs are thought to arise in hematopoietic stem cells (HSCs) that give rise to the diseased clonal progeny 2,3 . We recently developed a new data-independent acquisition (DIA) mass spectrometry (MS) technology for rare human hematopoietic stem and progenitor cell (HSPC) subpopulations 4 . This DIA-MS proteomic analysis was applied to human hematopoietic stem/multipotent progenitor cells (HSC/MPPs) and common myeloid/megakaryocyte-erythrocyte progenitors (CMP/MEPs) isolated from 123 blood samples of 18 PV patients and 21 controls (Supplementary Table 1). The proteomic dataset was complemented with RNA-sequencing data of the same patient and control samples. Comparing the proteomic and transcriptomic datasets in the corresponding patient and control samples demonstrated mainly positive correlations: 70% of genes had positive Spearman’s correlation coefficients for protein and RNA expression. In line with our previous observations in HSC/MPPs of healthy stem cell donors 4 , 30% of genes with altered expression in PV patients and their controls demonstrated negative correlation coefficients for protein and RNA expression, indicating additional information gained by proteomic compared to transcriptomic data. Downregulation of megakaryocyte differentiation and upregulation of cell proliferation in PV HSPCs and its reversal upon treatment with hydroxyurea (HU) To further examine the added information provided by protein compared to RNA expression data, we performed enrichment analyses for gene ontologies in HSPC subpopulations of PV patients on protein and RNA levels. Megakaryocyte differentiation and regulation was significantly downregulated in untreated PV patients at the protein level, but not at the RNA level in all HSPCs analyzed (Fig. 1A with individual gene ontology protein members shown in Fig. 1B). Similarly, RNAs and proteins implicated in DNA replication and G1/S transition of the mitotic cell cycle were discordantly regulated in HSC/MPPs of untreated PV patients (Fig. 1A). In contrast to the discordant protein and RNA expression in these gene sets, we observed concordant expression on the protein and RNA level for erythrocyte differentiation, regulation, development and maturation, receptor signaling via JAK-STAT, interferon-gamma signaling, cholesterol biosynthetic process, and TGFβ and MAPK signaling and regulation in HSPCs of untreated PV patients compared to controls (Fig. 1A). Fig. 1 Comparative unbiased proteomic analysis identifies downregulation of CXCL4/PF4 in HSC/MPPs of PV patients. A Gene set enrichment analysis (GSEA) comparing untreated PV patients against controls (PV.UT.HSC/MPP versus Control.HSC/MPP; PV.UT.CMP/MEP versus Control.CMP/MEP) and assessing for the effect of treatment with hydroxyurea (PV.HU.HSC/MPP versus PV.UT.HSC/MPP; PV.HU.CMP/MEP versus PV.UT.CMP/MEP). Shown are normalized enrichment scores (NES) for individual gene sets. Significantly upregulated gene sets are marked in red color, significantly downregulated gene sets are marked in blue color. Only RNAs and proteins expressed in at least half of the replicates in both comparison groups were considered. UT untreated, HU patient under treatment with hydroxyurea. B Heatmap of proteins enriched in different patient and control groups for megakaryocyte differentiation and regulation. C Volcano plot of protein intensity fold changes and p-values comparing HSC/MPPs of untreated PV patients (PV.UT.HSC/MPP) against controls (Control.HSC/MPP). D Normalized protein intensities for CXCL4/PF4 in the subgroups of controls, chronic PV patients without cytoreductive therapy (PVchron.UT), chronic PV patients with hydroxyurea therapy (PVchron.HU), and progressed PV patients (PVprog). Error bars represent standard deviations. *adj. P < 0.05; **adj. P < 0.01; ***adj. P < 0.001 Peptide profiles for CXCL4/PF4 are provided in Supplementary Fig. 1. E Graphical summary of intracellular FACS staining experiments for CXCL4/PF4 in six untreated chronic PV patients and age- and gender-matched controls. Error bars represent standard deviations. F Normalized CXCL4/PF4 expression values for protein and RNA in individual patient and control samples. Discrepant high RNA and low protein levels were seen in progressed PV patients with fibrosis (post-PV MF) but not in post-PV AML. Cytoreductive therapy with hydroxyurea reverted downregulation of megakaryocyte differentiation and regulation on protein level (Fig. 1A, B). It also reversed upregulation of cell proliferation (DNA replication, G1/S transition of the mitotic cell cycle, DNA repair) on RNA level and downregulation of TGFβ signaling on protein and RNA level (Fig. 1A). Erythrocyte differentiation and regulation were reversed by hydroxyurea on the protein but not RNA level, whereas interferon-gamma signaling was not affected by the treatment of patients with hydroxyurea (Fig. 1A). Reduced CXCL4/PF4 expression in HSC/MPPs of untreated PV patients To better characterize the molecular phenotype underlying PV stem and progenitor cell biology, we next focused on individual differentially regulated proteins in HSC/MPPs of untreated PV patients compared to controls. Consistent with the downregulation of megakaryocyte differentiation and regulation observed in the gene ontology enrichment analysis (Fig. 1A, B), we observed significant downregulation of the megakaryocytic lineage protein CXCL4/PF4 (platelet factor 4) (Fig. 1C, D and Supplementary Fig. 1). Intracellular flow cytometry confirmed the downregulation of CXCL4/PF4 in HSC/MPPs of untreated chronic phase PV patients in comparison to age- and gender-matched controls (Fig. 1E). Treatment with hydroxyurea abrogated the downregulation of CXCL4/PF4 protein expression in PV HSC/MPPs (Fig. 1D). Since all PV patients analyzed in this study carried the JAK2V617F mutation, we tested for a potential direct relationship between the JAK2V617F allele burden and CXCL4/PF4 protein expression. No significant correlation was observed (Supplementary Fig. 2), suggesting that reduced CXCL4/PF4 expression in PV is a JAK2V617F allele burden-independent disease manifestation. CXCL4/PF4 did not show equally significant expression changes on the RNA level as on the protein level. Exploring protein-RNA-correlations in more detail, we observed that protein and RNA levels became discordant upon disease progression (Fig. 1F). Upregulated RNA and downregulated protein levels for CXCL4/PF4 were observed in HSC/MPPs of post-PV myelofibrosis (MF) patients but not in post-PV acute myeloid leukemia (AML) stem cells. In controls and non-progressed PV patients, protein and RNA levels of CXCL4/PF4 were better aligned even if not systematically coordinated (Fig. 1F). Decreased ELF1 and USF2 activity explains reduced CXCL4/PF4 expression, which is linked to downregulated TGFβ signaling and loss of stem cell quiescence We next assessed the activities of transcription factors (TFs), which bind the CXCL4/PF4 promoter and may regulate CXCL4/PF4 expression in PV HSC/MPPs (Fig. 2A) 5–8 . We found a significantly decreased activity for ELF1 and to a lesser extent also for USF2 in HSC/MPPs of untreated PV compared to controls and HU-treated PV (Fig. 2A), suggesting a potential link between diminished ELF1 and USF2 activity and a reduction of CXCL4/PF4 expression in HSC/MPPs of untreated PV patients 5,8 . Upon progression to post-PV MF, the activities of the TFs USF2 and also NKX2-2 increased, potentially explaining upregulated RNA levels of CXCL4/PF4 in these patients (Supplementary Fig. 3) 5 . Fig. 2 CXCL4/PF4 protein is associated with transcriptional stem cell quiescence in untreated PV and inhibits the colony-formation capacity of PV HSC/MPPs. A Activity analysis of transcription factors (TFs) regulating CXCL4/PF4 expression showed significantly downregulated activity for the TF ELF1 and to a lesser degree for the TF USF2 in untreated PV patients compared to controls and HU-treated PV patients. Error bars represent standard errors. *P < 0.05. B Gene set enrichment analysis showed strong and significant enrichment for genes downregulated in stem cell quiescence in HSC/MPPs of untreated PV patients (PV.UT) compared to HSC/MPPs of controls, suggesting cessation of stem cell quiescence in PV stem cells. C Treatment of patients with hydroxyurea (PV.HU.HSC/MPP versus PV.UT.HSC/MPP) reversed the loss of stem cell quiescence in the PV HSC/MPP subpopulation. NES normalized enrichment score. D Heatmap of RNAs enriched in different patient/control groups for genes downregulated in stem cell quiescence. The corresponding enrichment plots and heatmap for the protein level are shown in Supplementary Fig. 4A-C. E Functional single-cell CXCL4/PF4 assay: colony growth of FACS-sorted HSC/MPPs from untreated chronic PV patients (PVchron.UT) singly incubated in cytokine-enriched serum-free medium was assessed in presence or absence of CXCL4/PF4. Colony growth as depicted was evaluated in 864 wells after 7 days of incubation. In individual patients, significance was determined using Fisher’s exact test; significance overall was calculated using two-tailed and paired Student’s t test (see Supplementary Methods). A strong growth inhibitory role of CXCL4/PF4 was observed. F Methylcellulose colony assay: the growth of different myeloid colony subtypes from FACS-sorted HSC/MPPs of untreated chronic PV patients (PVchron.UT) was evaluated in presence or absence of CXCL4/PF4. Reductions in colony growth due to CXCL4/PF4 protein were observed for all colony subtypes with significant inhibitions seen in the CFU-GM and BFU-E colony subtypes as well as in composite granulocyte/macrophage (CFU-GM and CFU-G and CFU-M) and erythroid (BFU-E and CFU-E) colonies. Relative numbers of colonies related to the total cell numbers plated are plotted. Error bars represent standard deviations. *P < 0.05. CFU-GEMM (mixed colonies), CFU-GM colony-forming unit–granulocyte/macrophage, CFU-G colony-forming unit–granulocyte, CFU-M colony-forming unit–macrophage, BFU-E burst-forming unit–erythroid, CFU-E colony-forming unit–erythroid. Reduced CXCL4/PF4 expression in HSC/MPPs of PV patients was associated with significant upregulation of genes downregulated in stem cell quiescence both on RNA (Fig. 2B, D) and protein (Supplementary Fig. 4A and 4C) level 9 . This pattern was reversed by treatment of patients with cytoreductive hydroxyurea (Fig. 2C, D and Supplementary Fig. 4B, C). Mechanistically, CXCL4/PF4 upregulates TGFβ 10 , which promotes stem cell quiescence 11 . We observed downregulation of TGFβ signaling in untreated PV patients that also exhibited downregulated CXCL4/PF4 levels (Fig. 1A). These data suggest that reduced CXCL4/PF4 expression in PV HSC/MPPs leads to the cessation of stem cell quiescence via downregulation of TGFβ signaling. CXCL4/PF4 inhibits colony formation of HSC/MPPs isolated from untreated chronic phase PV patients To assess the functional relevance of CXCL4/PF4 downregulation and to examine its causal link to HSC/MPP proliferation and differentiation in PV, we cultured HSC/MPPs of untreated chronic phase PV patients in the presence or absence of CXCL4/PF4 using two independent colony formation assays. Supplementation with CXCL4/PF4 inhibited colony formation of singly sorted PV HSC/MPPs after 7 days in the cytokine-enriched serum-free medium by 63–71% (Fig. 2E). In methylcellulose assays, a 54–61% inhibition of total colony formation was observed upon reconstitution/treatment with CXCL4/PF4 for FACS-sorted HSC/MPPs from PV patients (Fig. 2F). Detailed analyses for colony subtypes as defined by Manz et al. 12 showed significant reductions in colony growth from PV HSC/MPPs upon exposure to supplemental CXCL4/PF4 for CFU-GM (colony-forming unit–granulocyte/macrophage) and BFU-E (burst-forming unit–erythroid) (Fig. 2F). These results demonstrate that CXCL4/PF4 is linked to HSC/MPP proliferation and differentiation in PV, and reduced CXCL4/PF4 expression in HSC/MPPs may contribute to the proliferative state of PV. Discussion This study identifies reduced CXCL4/PF4 protein expression in HSC/MPPs of untreated PV patients compared to controls. Treatment of patients with cytoreductive hydroxyurea abrogated this effect. We demonstrated transcriptional cessation of stem cell quiescence to be associated with reduced CXCL4/PF4 expression in PV HSC/MPPs, and supplementation with CXCL4/PF4 strongly inhibited the in vitro colony-formation capacity of HSC/MPPs from PV patients. These findings extend previous reports in healthy hematopoietic stem cells and non-PV JAK2-mutated erythroblast-like cells 13 . CXCL4/PF4-/- mice were shown to exhibit increased numbers and proliferation of HSCs and MPPs 14 , and high CXCL4/PF4 levels were found to strongly inhibit hematopoiesis in non-PV mice 14 . The inhibitory role of CXCL4/PF4 on HSPC proliferation in our PV patients supports a key role of downregulated CXCL4/PF4 for the proliferative phenotype of PV HSC/MPPs. Mechanistically, we observed downregulation of TGFβ signaling in untreated PV patients showing reduced CXCL4/PF4 expression. CXCL4/PF4 was previously shown to activate stem cell quiescence-inducing TGFβ 10,11 . Upstream of CXCL4/PF4, we identified reduced activity of the CXCL4/PF4-regulating transcription factors ELF1 8 and USF2 5 in HSC/MPPs of untreated PV patients. Our data thus support a model in which decreased ELF1 and USF2 activity in PV HSC/MPPs leads to reduced CXCL4/PF4 expression, which leads to decreased TGFβ signaling and concomitant cessation of stem cell quiescence. Upon progression to post-PV MF, USF2 activity increased, explaining elevated CXCL4/PF4 RNA levels in this patient group. The PV disease stages and controls were better separated by CXCL4/PF4 protein expression than by CXCL4/PF4 RNA expression. Whereas transcriptomics reinforced the proteomic results for CXCL4/PF4 in chronic phase PV, discrepant RNA and protein levels were observed in HSC/MPPs of post-PV MF patients. Our findings in fibrotic patients are in line with recent observations of upregulated CXCL4/PF4 RNA levels in co-cultured HSPCs of a murine PMF model 15 . The congruent CXCL4/PF4 protein and RNA expression in control, chronic PV, and post-PV AML, but not in post-PV MF suggests post-transcriptional regulation at the base of discrepant RNA and protein levels in post-PV MF. In summary, this study identified downregulation of CXCL4/PF4 expression in HSC/MPPs with the cessation of stem cell quiescence through downregulation of TGFβ signaling as a potential new driver of the proliferative state of PV. Supplementary information Supplemental material

          Related collections

          Most cited references15

          • Record: found
          • Abstract: found
          • Article: not found

          Molecular regulation of stem cell quiescence.

          Subsets of mammalian adult stem cells reside in the quiescent state for prolonged periods of time. This state, which is reversible, has long been viewed as dormant and with minimal basal activity. Recent advances in adult stem cell isolation have provided insights into the epigenetic, transcriptional and post-transcriptional control of quiescence and suggest that quiescence is an actively maintained state in which signalling pathways are involved in maintaining a poised state that allows rapid activation. Deciphering the molecular mechanisms regulating adult stem cell quiescence will increase our understanding of tissue regeneration mechanisms and how they are dysregulated in pathological conditions and in ageing.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Benchmark and integration of resources for the estimation of human transcription factor activities

            The prediction of transcription factor (TF) activities from the gene expression of their targets (i.e., TF regulon) is becoming a widely used approach to characterize the functional status of transcriptional regulatory circuits. Several strategies and data sets have been proposed to link the target genes likely regulated by a TF, each one providing a different level of evidence. The most established ones are (1) manually curated repositories, (2) interactions derived from ChIP-seq binding data, (3) in silico prediction of TF binding on gene promoters, and (4) reverse-engineered regulons from large gene expression data sets. However, it is not known how these different sources of regulons affect the TF activity estimations and, thereby, downstream analysis and interpretation. Here we compared the accuracy and biases of these strategies to define human TF regulons by means of their ability to predict changes in TF activities in three reference benchmark data sets. We assembled a collection of TF–target interactions for 1541 human TFs and evaluated how different molecular and regulatory properties of the TFs, such as the DNA-binding domain, specificities, or mode of interaction with the chromatin, affect the predictions of TF activity. We assessed their coverage and found little overlap on the regulons derived from each strategy and better performance by literature-curated information followed by ChIP-seq data. We provide an integrated resource of all TF–target interactions derived through these strategies, with confidence scores, as a resource for enhanced prediction of TF activities.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Megakaryocytes regulate hematopoietic stem cell quiescence via Cxcl4 secretion

              In the bone marrow (BM), hematopoietic stem cells (HSCs) lodge in specialized microenvironments that tightly control their proliferative state to adapt to the varying needs for replenishment of blood cells while also preventing exhaustion 1 . All putative niche cells suggested thus far have a non-hematopoietic origin 2-8 . Thus, it remains unclear how feedback from mature cells is conveyed to HSCs to adjust proliferation. Here we show that megakaryocytes (Mk) can directly regulate HSC pool size. Three-dimensional whole-mount imaging revealed that endogenous HSCs are frequently located adjacent to Mk in a non-random fashion. Selective in vivo depletion of Mk resulted in specific loss of HSC quiescence and led to a marked expansion of functional HSCs. Gene expression analyses revealed that Mk were the source of chemokine C-X-C motif ligand 4 (Cxcl4, also named platelet factor 4, Pf4) in the BM and Cxcl4 injection reduced HSC numbers via increased quiescence. By contrast, Cxcl4 −/− mice exhibited increased HSC numbers and proliferation. Combined use of whole-mount imaging and computational modelling was highly suggestive of a megakaryocytic niche capable of influencing independently HSC maintenance by regulating quiescence. Thus, these results indicate that a terminally differentiated HSC progeny contributes to niche activity by directly regulating HSC behavior.
                Bookmark

                Author and article information

                Contributors
                fabienne.meier-abt@usz.ch
                Journal
                Blood Cancer J
                Blood Cancer J
                Blood Cancer Journal
                Nature Publishing Group UK (London )
                2044-5385
                11 February 2021
                11 February 2021
                February 2021
                : 11
                : 2
                : 31
                Affiliations
                [1 ]GRID grid.412004.3, ISNI 0000 0004 0478 9977, Department of Medical Oncology and Hematology, , University Hospital Zurich and University of Zurich, ; Zurich, Switzerland
                [2 ]GRID grid.5801.c, ISNI 0000 0001 2156 2780, Department of Biology, Institute of Molecular Systems Biology, , ETH Zurich, ; Zurich, Switzerland
                [3 ]GRID grid.5801.c, ISNI 0000 0001 2156 2780, Functional Genomics Center Zurich, , University and ETH Zurich, ; Zurich, Switzerland
                [4 ]GRID grid.419765.8, ISNI 0000 0001 2223 3006, Swiss Institute of Bioinformatics, ; Lausanne, Switzerland
                [5 ]GRID grid.7400.3, ISNI 0000 0004 1937 0650, Faculty of Science, University of Zurich, ; Zurich, Switzerland
                Author information
                http://orcid.org/0000-0001-5337-6210
                http://orcid.org/0000-0003-0026-8739
                Article
                423
                10.1038/s41408-021-00423-5
                7878875
                33574218
                67225b87-bf86-4f5a-a1d7-97dbcb355dd9
                © The Author(s) 2021

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 13 November 2020
                : 5 January 2021
                : 20 January 2021
                Funding
                Funded by: Personalized Health and Related Technologies (PHRT), ETH Zurich Filling-the-Gap, University of Zurich Krebsliga Zurich
                Funded by: Functional Genomics Center Zurich
                Funded by: FundRef https://doi.org/10.13039/501100001711, Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation);
                Award ID: 3100A0-688 107679
                Award Recipient :
                Categories
                Correspondence
                Custom metadata
                © The Author(s) 2021

                Oncology & Radiotherapy
                myeloproliferative disease,preclinical research
                Oncology & Radiotherapy
                myeloproliferative disease, preclinical research

                Comments

                Comment on this article