15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Single-Cell Analyses Reveal Megakaryocyte-Biased Hematopoiesis in Myelofibrosis and Identify Mutant Clone-Specific Targets

      research-article
      1 , 2 , 3 , 4 , 14 , , 1 , 2 , 3 , 5 , 14 , 1 , 2 , 3 , 5 , 1 , 2 , 3 , 4 , 1 , 2 , 3 , 6 , 6 , 1 , 2 , 3 , 1 , 2 , 3 , 7 , 8 , 9 , 9 , NIH Intramural Sequencing Center 10 , 11 , 12 , 11 , 2 , 3 , 13 , 4 , 3 , 5 , ∗∗ , 1 , 2 , 3 , 15 , ∗∗∗
      Molecular Cell
      Cell Press
      megakaryopoiesis, myeloproliferative neoplasm, platelets, TARGET-seq, immunotherapy, single-cell multi-omics, G6B, fibrosis, bone marrow

      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.

          Summary

          Myelofibrosis is a severe myeloproliferative neoplasm characterized by increased numbers of abnormal bone marrow megakaryocytes that induce fibrosis, destroying the hematopoietic microenvironment. To determine the cellular and molecular basis for aberrant megakaryopoiesis in myelofibrosis, we performed single-cell transcriptome profiling of 135,929 CD34 + lineage hematopoietic stem and progenitor cells (HSPCs), single-cell proteomics, genomics, and functional assays. We identified a bias toward megakaryocyte differentiation apparent from early multipotent stem cells in myelofibrosis and associated aberrant molecular signatures. A sub-fraction of myelofibrosis megakaryocyte progenitors (MkPs) are transcriptionally similar to healthy-donor MkPs, but the majority are disease specific, with distinct populations expressing fibrosis- and proliferation-associated genes. Mutant-clone HSPCs have increased expression of megakaryocyte-associated genes compared to wild-type HSPCs, and we provide early validation of G6B as a potential immunotherapy target. Our study paves the way for selective targeting of the myelofibrosis clone and illustrates the power of single-cell multi-omics to discover tumor-specific therapeutic targets and mediators of tissue fibrosis.

          Graphical Abstract

          Highlights

          • Single-cell-omics demonstrate megakaryocyte-biased hematopoiesis in myelofibrosis (MF)

          • Megakaryocyte progenitors (MkPs) show high expression of a fibrosis signature in MF

          • MkPs are heterogeneous in MF with aberrant metabolic and inflammatory signatures

          • Targeting aberrant surface G6B expression may selectively ablate the MF clone

          Abstract

          Myelofibrosis (MF) is characterized by increased numbers of morphologically abnormal megakaryocytes (Mks). Single-cell RNA sequencing of >120,000 hematopoietic stem and progenitor cells demonstrated Mk-biased hematopoiesis across clinical and molecular MF subgroups. Mk progenitors were heterogeneous, with distinct expression of inflammatory mediators. Aberrant surface G6B expression on MF stem and progenitors could allow selective immunotherapeutic targeting of the MF clone.

          Related collections

          Most cited references55

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

          Accounting for technical noise in single-cell RNA-seq experiments.

          Single-cell RNA-seq can yield valuable insights about the variability within a population of seemingly homogeneous cells. We developed a quantitative statistical method to distinguish true biological variability from the high levels of technical noise in single-cell experiments. Our approach quantifies the statistical significance of observed cell-to-cell variability in expression strength on a gene-by-gene basis. We validate our approach using two independent data sets from Arabidopsis thaliana and Mus musculus.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Identification of clonogenic common lymphoid progenitors in mouse bone marrow.

            The existence of a common lymphoid progenitor that can only give rise to T cells, B cells, and natural killer (NK) cells remains controversial and constitutes an important gap in the hematopoietic lineage maps. Here, we report that the Lin(-)IL-7R(+)Thy-1(-)Sca-1loc-Kit(lo) population from adult mouse bone marrow possessed a rapid lymphoid-restricted (T, B, and NK) reconstitution capacity in vivo but completely lacked myeloid differentiation potential either in vivo or in vitro. A single Lin(-)IL-7R(+)Thy-1(-)Sca-1loc-Kit(lo) cell could generate at least both T and B cells. These data provide direct evidence for the existence of common lymphoid progenitors in sites of early hematopoiesis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              From haematopoietic stem cells to complex differentiation landscapes.

              The development of mature blood cells from haematopoietic stem cells has long served as a model for stem-cell research, with the haematopoietic differentiation tree being widely used as a model for the maintenance of hierarchically organized tissues. Recent results and new technologies have challenged the demarcations between stem and progenitor cell populations, the timing of cell-fate choices and the contribution of stem and multipotent progenitor cells to the maintenance of steady-state blood production. These evolving views of haematopoiesis have broad implications for our understanding of the functions of adult stem cells, as well as the development of new therapies for malignant and non-malignant haematopoietic diseases.
                Bookmark

                Author and article information

                Contributors
                Journal
                Mol Cell
                Mol. Cell
                Molecular Cell
                Cell Press
                1097-2765
                1097-4164
                07 May 2020
                07 May 2020
                : 78
                : 3
                : 477-492.e8
                Affiliations
                [1 ]Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX3 9DS, UK
                [2 ]MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK
                [3 ]NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK
                [4 ]Hematopoiesis Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-4442, USA
                [5 ]MRC WIMM Centre for Computational Biology, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK
                [6 ]York Biomedical Research Institute and Department of Biology, University of York, Heslington, York YO10 5DD, UK
                [7 ]NHGRI Flow Cytometry Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-4442, USA
                [8 ]Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche-S 1255, Etablissement Français du Sang Grand Est, Strasbourg 67065, France
                [9 ]Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
                [10 ]National Institutes of Health, Bethesda MD 20892-442, USA
                [11 ]Centre for Haematology, Hammersmith Hospital, Imperial College of Medicine, London W12 OHS, UK
                [12 ]Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
                [13 ]Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK
                Author notes
                []Corresponding author bethan.psaila@ 123456ndcls.ox.ac.uk
                [∗∗ ]Corresponding author supat.thongjuea@ 123456imm.ox.ac.uk
                [∗∗∗ ]Corresponding author adam.mead@ 123456imm.ox.ac.uk
                [14]

                These authors contributed equally

                [15]

                Lead Contact

                Article
                S1097-2765(20)30234-3
                10.1016/j.molcel.2020.04.008
                7217381
                32386542
                14f34700-bc46-4e4e-a66f-60de58b460c9
                © 2020 The Authors

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 20 May 2019
                : 4 February 2020
                : 6 April 2020
                Categories
                Article

                Molecular biology
                megakaryopoiesis,myeloproliferative neoplasm,platelets,target-seq,immunotherapy,single-cell multi-omics,g6b,fibrosis,bone marrow

                Comments

                Comment on this article