The Molecular Signature of Megakaryocyte-Erythroid Progenitors Reveals a Role for the Cell Cycle in Fate Specification

SUMMARY Megakaryocytic-erythroid progenitors (MEPs) give rise to the cells that produce red blood cells and platelets. Although the mechanisms underlying megakaryocytic (MK) and erythroid (E) maturation have been described, those controlling their specification from MEPs are unknown. Single-cell RNA sequencing of primary human MEPs, common myeloid progenitors (CMPs), megakaryocyte progenitors, and E progenitors revealed a distinct transitional MEP signature. Inferred regulatory transcription factors (TFs) were associated with differential expression of cell cycle regulators. Genetic manipulation of selected TFs validated their role in lineage specification and demonstrated coincident modulation of the cell cycle. Genetic and pharmacologic modulation demonstrated that cell cycle activation is sufficient to promote E versus MK specification. These findings, obtained from healthy human cells, lay a foundation to study the mechanisms underlying benign and malignant disease states of the megakaryocytic and E lineages.

[1]  J. León,et al.  Myc roles in hematopoiesis and leukemia. , 2010, Genes & cancer.

[2]  S. Yamanaka,et al.  Transient activation of c-MYC expression is critical for efficient platelet generation from human induced pluripotent stem cells , 2010, The Journal of experimental medicine.

[3]  R. Hardison,et al.  SCL and associated proteins distinguish active from repressive GATA transcription factor complexes. , 2008, Blood.

[4]  L. Doré,et al.  Transcription factor networks in erythroid cell and megakaryocyte development. , 2011, Blood.

[5]  A. Migliaccio,et al.  Identification and characterization of a bipotent (erythroid and megakaryocytic) cell precursor from the spleen of phenylhydrazine-treated mice. , 2000, Blood.

[6]  Gazelle Zerafati,et al.  Adult human megakaryocyte-erythroid progenitors are in the CD34+CD38mid fraction. , 2016, Blood.

[7]  R. Eisenman,et al.  Max: a helix-loop-helix zipper protein that forms a sequence-specific DNA-binding complex with Myc. , 1991, Science.

[8]  Randy J. Read,et al.  Transcriptional diversity during lineage commitment of human blood progenitors , 2014, Science.

[9]  T. Graf,et al.  CD41 expression marks myeloid-biased adult hematopoietic stem cells and increases with age. , 2013, Blood.

[10]  Xiaowei Wang,et al.  PrimerBank: a resource of human and mouse PCR primer pairs for gene expression detection and quantification , 2009, Nucleic Acids Res..

[11]  Hao Yuan Kueh,et al.  Positive Feedback Between PU.1 and the Cell Cycle Controls Myeloid Differentiation , 2013, Science.

[12]  S. Saad,et al.  Hematopoietic defects in response to reduced Arhgap21 , 2017, Stem cell research.

[13]  M. Weiss,et al.  Early block to erythromegakaryocytic development conferred by loss of transcription factor GATA-1. , 2005, Blood.

[14]  Ye Ding,et al.  An extensive network of TET2-targeting MicroRNAs regulates malignant hematopoiesis. , 2013, Cell reports.

[15]  S. Soneji,et al.  Human and Murine Hematopoietic Stem Cell Aging Is Associated with Functional Impairments and Intrinsic Megakaryocytic/Erythroid Bias , 2016, PloS one.

[16]  T. Höfer,et al.  CCND1–CDK4–mediated cell cycle progression provides a competitive advantage for human hematopoietic stem cells in vivo , 2015, The Journal of experimental medicine.

[17]  Cyrille F. Dunant,et al.  Distinct routes of lineage development reshape the human blood hierarchy across ontogeny , 2016, Science.

[18]  F. Hamey,et al.  Demystifying blood stem cell fates , 2017, Nature Cell Biology.

[19]  David R. Kelley,et al.  Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks , 2012, Nature Protocols.

[20]  I. Macaulay,et al.  Platelet-biased stem cells reside at the apex of the haematopoietic stem-cell hierarchy , 2013, Nature.

[21]  Sydney M. Shaffer,et al.  Rare Cell Detection by Single-Cell RNA Sequencing as Guided by Single-Molecule RNA FISH. , 2018, Cell systems.

[22]  M. Warr,et al.  Autophagy maintains the metabolism and function of young and old (hematopoietic) stem cells , 2017, Nature.

[23]  Rong Fan,et al.  Nonstochastic Reprogramming from a Privileged Somatic Cell State , 2014, Cell.

[24]  N. Rhind,et al.  Global increase in replication fork speed during a p57KIP2-regulated erythroid cell fate switch , 2016, Science Advances.

[25]  N. Salomonis,et al.  Cross-platform single cell analysis of kidney development shows stromal cells express Gdnf. , 2017, Developmental biology.

[26]  D. Krause,et al.  Concise Review: Bipotent Megakaryocytic‐Erythroid Progenitors: Concepts and Controversies , 2018, Stem cells.

[27]  I. Amit,et al.  Transcriptional Heterogeneity and Lineage Commitment in Myeloid Progenitors , 2016, Cell.

[28]  Chi V Dang,et al.  MYC on the Path to Cancer , 2012, Cell.

[29]  J. Crispino,et al.  Combinatorial regulation of tissue specification by GATA and FOG factors , 2012, Development.

[30]  S. Dalton,et al.  Concise Review: Control of Cell Fate Through Cell Cycle and Pluripotency Networks , 2016, Stem cells.

[31]  L. Steinmetz,et al.  Human haematopoietic stem cell lineage commitment is a continuous process , 2017, Nature Cell Biology.

[32]  C. Niu,et al.  c-Myc-mediated control of cell fate in megakaryocyte-erythrocyte progenitors. , 2009, Blood.

[33]  Andrew J. Hill,et al.  Single-cell mRNA quantification and differential analysis with Census , 2017, Nature Methods.

[34]  Shangqin Guo,et al.  Choosing Cell Fate Through a Dynamic Cell Cycle , 2015, Current Stem Cell Reports.

[35]  Dorothea Emig,et al.  AltAnalyze and DomainGraph: analyzing and visualizing exon expression data , 2010, Nucleic Acids Res..

[36]  G. Blobe,et al.  Gfi-1B controls human erythroid and megakaryocytic differentiation by regulating TGF-beta signaling at the bipotent erythro-megakaryocytic progenitor stage. , 2010, Blood.

[37]  I. Weissman,et al.  A clonogenic common myeloid progenitor that gives rise to all myeloid lineages , 2000, Nature.

[38]  Xiaoming Wang,et al.  mTORC signaling in hematopoiesis , 2016, International Journal of Hematology.

[39]  W. Vainchenker,et al.  Characterization of a bipotent erythro-megakaryocytic progenitor in human bone marrow. , 1996, Blood.

[40]  Cole Trapnell,et al.  The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells , 2014, Nature Biotechnology.

[41]  Sean C. Bendall,et al.  Single-Cell Mass Cytometry of Differential Immune and Drug Responses Across a Human Hematopoietic Continuum , 2011, Science.

[42]  Katsu Takahashi,et al.  Mitochondrial function provides instructive signals for activation-induced B-cell fates , 2015, Nature Communications.

[43]  S. Dalton,et al.  Cycling through developmental decisions: how cell cycle dynamics control pluripotency, differentiation and reprogramming , 2016, Development.

[44]  Levon M. Khachigian,et al.  Protein-Protein Interaction between Fli-1 and GATA-1 Mediates Synergistic Expression of Megakaryocyte-Specific Genes through Cooperative DNA Binding , 2003, Molecular and Cellular Biology.

[45]  J. Koch,et al.  RUNX1 represses the erythroid gene expression program during megakaryocytic differentiation. , 2015, Blood.

[46]  Bruce J. Aronow,et al.  Single-cell analysis of mixed-lineage states leading to a binary cell fate choice , 2016, Nature.

[47]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[48]  Donald Metcalf,et al.  The critical regulator of embryonic hematopoiesis, SCL, is vital in the adult for megakaryopoiesis, erythropoiesis, and lineage choice in CFU-S12 , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[49]  Yukio Kondo,et al.  TGF-β–FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia , 2010, Nature.