1
|
Mitsutani M, Yokoyama M, Hano H, Morita A, Matsushita M, Tagami T, Moriyama K. Growth hormone is involved in GATA1 gene expression via STAT5B in human erythroleukemia and monocytic cell lines. Blood Cells Mol Dis 2025; 110:102894. [PMID: 39303396 DOI: 10.1016/j.bcmd.2024.102894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
GATAs are a family of transcription factors consisting of six members. Particularly, GATA1 and GATA2 have been reported to promote the development of erythrocytes, megakaryocytes, eosinophils, and mast cells. However, little information is available on the extracellular ligands that promote GATA1 expression. We evaluated whether growth hormone (GH) is an extracellular stimulator that participates in the signal transduction of GATAs, focusing on GATA1 expression in hematopoietic cell lineages. We used a reporter assay, RT-PCR, real-time quantitative PCR, and western blotting to evaluate GH-induced expression of GATA1 and GATA2 in the human erythroleukemic cell line K562 and the non-erythroid cell line U937. GATA1 expression in these hematopoietic cell lines increased at the transcriptional and protein levels in the presence of GH, and was inhibited by a STAT5 specific inhibitor. Cells transfected with activated STAT5B showed increased expression of GATA1. We identified functional STAT5B consensus sequences as binding site-158 bp from the transcription starting site in the GATA1 promoter region. These results suggest that GH directly induces GATA1 expression via GHR/JAK/STAT5 and is related to hematopoietic cell proliferation.
Collapse
Affiliation(s)
- Mana Mitsutani
- Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Mei Yokoyama
- Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Hiromi Hano
- Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Aoi Morita
- Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Midori Matsushita
- Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Tetsuya Tagami
- Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| | - Kenji Moriyama
- Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan; Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan.
| |
Collapse
|
2
|
Bieker JJ, Philipsen S. Erythroid Krüppel-Like Factor (KLF1): A Surprisingly Versatile Regulator of Erythroid Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:217-242. [PMID: 39017846 DOI: 10.1007/978-3-031-62731-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Erythroid Krüppel-like factor (KLF1), first discovered in 1992, is an erythroid-restricted transcription factor (TF) that is essential for terminal differentiation of erythroid progenitors. At face value, KLF1 is a rather inconspicuous member of the 26-strong SP/KLF TF family. However, 30 years of research have revealed that KLF1 is a jack of all trades in the molecular control of erythropoiesis. Initially described as a one-trick pony required for high-level transcription of the adult HBB gene, we now know that it orchestrates the entire erythroid differentiation program. It does so not only as an activator but also as a repressor. In addition, KLF1 was the first TF shown to be directly involved in enhancer/promoter loop formation. KLF1 variants underlie a wide range of erythroid phenotypes in the human population, varying from very mild conditions such as hereditary persistence of fetal hemoglobin and the In(Lu) blood type in the case of haploinsufficiency, to much more serious non-spherocytic hemolytic anemias in the case of compound heterozygosity, to dominant congenital dyserythropoietic anemia type IV invariably caused by a de novo variant in a highly conserved amino acid in the KLF1 DNA-binding domain. In this chapter, we present an overview of the past and present of KLF1 research and discuss the significance of human KLF1 variants.
Collapse
Affiliation(s)
- James J Bieker
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Chen X, Pillay S, Lohmann F, Bieker JJ. Association of DDX5/p68 protein with the upstream erythroid enhancer element (EHS1) of the gene encoding the KLF1 transcription factor. J Biol Chem 2023; 299:105489. [PMID: 38000658 PMCID: PMC10750184 DOI: 10.1016/j.jbc.2023.105489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 10/28/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
EKLF/KLF1 is an essential transcription factor that plays a global role in erythroid transcriptional activation. Regulation of KLF1 is of interest, as it displays a highly restricted expression pattern, limited to erythroid cells and its progenitors. Here we use biochemical affinity purification to identify the DDX5/p68 protein as an activator of KLF1 by virtue of its interaction with the erythroid-specific DNAse hypersensitive site upstream enhancer element (EHS1). We further show that this protein associates with DEK and CTCF. We postulate that the range of interactions of DDX5/p68 with these and other proteins known to interact with this element render it part of the enhanseosome complex critical for optimal expression of KLF1 and enables the formation of a proper chromatin configuration at the Klf1 locus. These individual interactions provide quantitative contributions that, in sum, establish the high-level activity of the Klf1 promoter and suggest they can be selectively manipulated for clinical benefit.
Collapse
Affiliation(s)
- Xiaoyong Chen
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Sanjana Pillay
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Felix Lohmann
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - James J Bieker
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA; Black Familly Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA; Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA; Mindich Child Health and Development Institute, Mount Sinai School of Medicine, New York, New York, USA.
| |
Collapse
|
4
|
Quotti Tubi L, Canovas Nunes S, Mandato E, Pizzi M, Vitulo N, D’Agnolo M, Colombatti R, Martella M, Boaro MP, Doriguzzi Breatta E, Fregnani A, Spinello Z, Nabergoj M, Filhol O, Boldyreff B, Albiero M, Fadini GP, Gurrieri C, Vianello F, Semenzato G, Manni S, Trentin L, Piazza F. CK2β Regulates Hematopoietic Stem Cell Biology and Erythropoiesis. Hemasphere 2023; 7:e978. [PMID: 38026791 PMCID: PMC10673422 DOI: 10.1097/hs9.0000000000000978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
The Ser-Thr kinase CK2 plays important roles in sustaining cell survival and resistance to stress and these functions are exploited by different types of blood tumors. Yet, the physiological involvement of CK2 in normal blood cell development is poorly known. Here, we discovered that the β regulatory subunit of CK2 is critical for normal hematopoiesis in the mouse. Fetal livers of conditional CK2β knockout embryos showed increased numbers of hematopoietic stem cells associated to a higher proliferation rate compared to control animals. Both hematopoietic stem and progenitor cells (HSPCs) displayed alterations in the expression of transcription factors involved in cell quiescence, self-renewal, and lineage commitment. HSPCs lacking CK2β were functionally impaired in supporting both in vitro and in vivo hematopoiesis as demonstrated by transplantation assays. Furthermore, KO mice developed anemia due to a reduced number of mature erythroid cells. This compartment was characterized by dysplasia, proliferative defects at early precursor stage, and apoptosis at late-stage erythroblasts. Erythroid cells exhibited a marked compromise of signaling cascades downstream of the cKit and erythropoietin receptor, with a defective activation of ERK/JNK, JAK/STAT5, and PI3K/AKT pathways and perturbations of several transcriptional programs as demonstrated by RNA-Seq analysis. Moreover, we unraveled an unforeseen molecular mechanism whereby CK2 sustains GATA1 stability and transcriptional proficiency. Thus, our work demonstrates new and crucial functions of CK2 in HSPC biology and in erythropoiesis.
Collapse
Affiliation(s)
- Laura Quotti Tubi
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Sara Canovas Nunes
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Elisa Mandato
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marco Pizzi
- Department of Medicine, Cytopathology and Surgical Pathology Unit, University of Padova, Italy
| | - Nicola Vitulo
- Department of Biotechnology, University of Verona, Italy
| | - Mirco D’Agnolo
- Department of Women’s and Child’s Health, University of Padova, Italy
| | | | | | - Maria Paola Boaro
- Department of Women’s and Child’s Health, University of Padova, Italy
| | - Elena Doriguzzi Breatta
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Anna Fregnani
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Zaira Spinello
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Mitja Nabergoj
- Hematology Service, Institut Central des Hôpitaux (ICH), Hôpital du Valais, Sion, Switzerland
| | - Odile Filhol
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1036, Institute de Reserches en Technologies et Sciences pour le Vivant/Biologie du Cancer et de l’Infection, Grenoble, France
| | | | - Mattia Albiero
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy
- Veneto Institute of Molecular Medicine, Experimental Diabetology Lab, Padova, Italy
| | - Gian Paolo Fadini
- Veneto Institute of Molecular Medicine, Experimental Diabetology Lab, Padova, Italy
- Department of Medicine, University of Padova, Italy
| | - Carmela Gurrieri
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Fabrizio Vianello
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Sabrina Manni
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Livio Trentin
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Division of Hematology, University of Padova, Italy
- Laboratory of Normal and Malignant Hematopoiesis and Pathobiology of Myeloma and Lymphoma. Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| |
Collapse
|
5
|
Gnanapragasam MN, Planutis A, Glassberg JA, Bieker JJ. Identification of a genomic DNA sequence that quantitatively modulates KLF1 transcription factor expression in differentiating human hematopoietic cells. Sci Rep 2023; 13:7589. [PMID: 37165057 PMCID: PMC10172341 DOI: 10.1038/s41598-023-34805-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 05/08/2023] [Indexed: 05/12/2023] Open
Abstract
The onset of erythropoiesis is under strict developmental control, with direct and indirect inputs influencing its derivation from the hematopoietic stem cell. A major regulator of this transition is KLF1/EKLF, a zinc finger transcription factor that plays a global role in all aspects of erythropoiesis. Here, we have identified a short, conserved enhancer element in KLF1 intron 1 that is important for establishing optimal levels of KLF1 in mouse and human cells. Chromatin accessibility of this site exhibits cell-type specificity and is under developmental control during the differentiation of human CD34+ cells towards the erythroid lineage. This site binds GATA1, SMAD1, TAL1, and ETV6. In vivo editing of this region in cell lines and primary cells reduces KLF1 expression quantitatively. However, we find that, similar to observations seen in pedigrees of families with KLF1 mutations, downstream effects are variable, suggesting that the global architecture of the site is buffered towards keeping the KLF1 genetic region in an active state. We propose that modification of intron 1 in both alleles is not equivalent to complete loss of function of one allele.
Collapse
Affiliation(s)
- M N Gnanapragasam
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1020, New York, NY, 10029, USA
- Department of Biological, Geological, and Environmental Sciences, Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA
| | - A Planutis
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1020, New York, NY, 10029, USA
| | - J A Glassberg
- Department of Emergency Medicine, Hematology and Medical Oncology, Mount Sinai School of Medicine, New York, NY, USA
| | - J J Bieker
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1020, New York, NY, 10029, USA.
- Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, USA.
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, USA.
- Mindich Child Health and Development Institute, Mount Sinai School of Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Ghersi JJ, Mahony CB, Bertrand JY. bif1, a new BMP signaling inhibitor, regulates embryonic hematopoiesis in the zebrafish. Development 2019; 146:dev.164103. [PMID: 30837221 DOI: 10.1242/dev.164103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/22/2019] [Indexed: 01/17/2023]
Abstract
Hematopoiesis maintains the entire blood system, and dysregulation of this process can lead to malignancies (leukemia), immunodeficiencies or red blood cell diseases (anemia, polycythemia vera). We took advantage of the zebrafish model that shares most of the genetic program involved in hematopoiesis with mammals to characterize a new gene of unknown function, si:ch73-299h12.2, which is expressed in the erythroid lineage during primitive, definitive and adult hematopoiesis. This gene, required during primitive and definitive erythropoiesis, encodes a C2H2 zinc-finger protein that inhibits BMP signaling. We therefore named this gene blood-inducing factor 1 and BMP inhibitory factor 1 (bif1). We identified a bif1 ortholog in Sinocyclocheilus rhinocerous, another fish, and in the mouse genome. Both genes also inhibit BMP signaling when overexpressed in zebrafish. In conclusion, we have deorphanized a new zebrafish gene of unknown function: bif1 codes for a zinc-finger protein that inhibits BMP signaling and also regulates primitive erythropoiesis and definitive hematopoiesis.
Collapse
Affiliation(s)
- Joey J Ghersi
- University of Geneva, School of Medicine, Department of Pathology and Immunology, CMU, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Christopher B Mahony
- University of Geneva, School of Medicine, Department of Pathology and Immunology, CMU, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Julien Y Bertrand
- University of Geneva, School of Medicine, Department of Pathology and Immunology, CMU, University of Geneva, CH-1211 Geneva 4, Switzerland
| |
Collapse
|
7
|
Chal J, Al Tanoury Z, Oginuma M, Moncuquet P, Gobert B, Miyanari A, Tassy O, Guevara G, Hubaud A, Bera A, Sumara O, Garnier JM, Kennedy L, Knockaert M, Gayraud-Morel B, Tajbakhsh S, Pourquié O. Recapitulating early development of mouse musculoskeletal precursors of the paraxial mesoderm in vitro. Development 2018; 145:145/6/dev157339. [DOI: 10.1242/dev.157339] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 02/06/2018] [Indexed: 12/13/2022]
Abstract
ABSTRACT
Body skeletal muscles derive from the paraxial mesoderm, which forms in the posterior region of the embryo. Using microarrays, we characterize novel mouse presomitic mesoderm (PSM) markers and show that, unlike the abrupt transcriptome reorganization of the PSM, neural tube differentiation is accompanied by progressive transcriptome changes. The early paraxial mesoderm differentiation stages can be efficiently recapitulated in vitro using mouse and human pluripotent stem cells. While Wnt activation alone can induce posterior PSM markers, acquisition of a committed PSM fate and efficient differentiation into anterior PSM Pax3+ identity further requires BMP inhibition to prevent progenitors from drifting to a lateral plate mesoderm fate. When transplanted into injured adult muscle, these precursors generated large numbers of immature muscle fibers. Furthermore, exposing these mouse PSM-like cells to a brief FGF inhibition step followed by culture in horse serum-containing medium allows efficient recapitulation of the myogenic program to generate myotubes and associated Pax7+ cells. This protocol results in improved in vitro differentiation and maturation of mouse muscle fibers over serum-free protocols and enables the study of myogenic cell fusion and satellite cell differentiation.
Collapse
Affiliation(s)
- Jérome Chal
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Masayuki Oginuma
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Philippe Moncuquet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Bénédicte Gobert
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Anagenesis Biotechnologies, Parc d'innovation, Illkirch Graffenstaden 67400, France
| | - Ayako Miyanari
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Olivier Tassy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Getzabel Guevara
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Alexis Hubaud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Agata Bera
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Olga Sumara
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Jean-Marie Garnier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Leif Kennedy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Marie Knockaert
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Barbara Gayraud-Morel
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| |
Collapse
|
8
|
Chondrou V, Kolovos P, Sgourou A, Kourakli A, Pavlidaki A, Kastrinou V, John A, Symeonidis A, Ali BR, Papachatzopoulou A, Katsila T, Patrinos GP. Whole transcriptome analysis of human erythropoietic cells during ontogenesis suggests a role of VEGFA gene as modulator of fetal hemoglobin and pharmacogenomic biomarker of treatment response to hydroxyurea in β-type hemoglobinopathy patients. Hum Genomics 2017; 11:24. [PMID: 29061162 PMCID: PMC5654038 DOI: 10.1186/s40246-017-0120-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/16/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Human erythropoiesis is characterized by distinct gene expression profiles at various developmental stages. Previous studies suggest that fetal-to-adult hemoglobin switch is regulated by a complex mechanism, in which many key players still remain unknown. Here, we report our findings from whole transcriptome analysis of erythroid cells, isolated from erythroid tissues at various developmental stages in an effort to identify distinct molecular signatures of each erythroid tissue. RESULTS From our in-depth data analysis, pathway analysis, and text mining, we opted to focus on the VEGFA gene, given its gene expression characteristics. Selected VEGFA genomic variants, identified through linkage disequilibrium analysis, were explored further for their association with elevated fetal hemoglobin levels in β-type hemoglobinopathy patients. Our downstream analysis of non-transfusion-dependent β-thalassemia patients, β-thalassemia major patients, compound heterozygous sickle cell disease/β-thalassemia patients receiving hydroxyurea as fetal hemoglobin augmentation treatment, and non-thalassemic individuals indicated that VEGFA genomic variants were associated with disease severity in β-thalassemia patients and hydroxyurea treatment efficacy in SCD/β-thalassemia compound heterozygous patients. CONCLUSIONS Our findings suggest that VEGFA may act as a modifier gene of human globin gene expression and, at the same time, serve as a genomic biomarker in β-type hemoglobinopathy disease severity and hydroxyurea treatment efficacy.
Collapse
Affiliation(s)
- Vasiliki Chondrou
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
| | - Petros Kolovos
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | - Alexandra Kourakli
- Hematology Division, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras, Greece
| | - Alexia Pavlidaki
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
- Present address: Institut de Génétique et de Biologie Moléculaire et Cellulaire IGBMC/CNRS/INSERM/UDS, 67404 ILLKIRCH, BP 10142, CU de Strasbourg, France
| | - Vlasia Kastrinou
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
| | - Anne John
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Argiris Symeonidis
- Hematology Division, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras, Greece
| | - Bassam R Ali
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | | | - Theodora Katsila
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
| | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece.
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
| |
Collapse
|
9
|
Abstract
Skeletal muscle is the largest tissue in the body and loss of its function or its regenerative properties results in debilitating musculoskeletal disorders. Understanding the mechanisms that drive skeletal muscle formation will not only help to unravel the molecular basis of skeletal muscle diseases, but also provide a roadmap for recapitulating skeletal myogenesis in vitro from pluripotent stem cells (PSCs). PSCs have become an important tool for probing developmental questions, while differentiated cell types allow the development of novel therapeutic strategies. In this Review, we provide a comprehensive overview of skeletal myogenesis from the earliest premyogenic progenitor stage to terminally differentiated myofibers, and discuss how this knowledge has been applied to differentiate PSCs into muscle fibers and their progenitors in vitro.
Collapse
Affiliation(s)
- Jérome Chal
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA .,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
10
|
Planutis A, Xue L, Trainor CD, Dangeti M, Gillinder K, Siatecka M, Nebor D, Peters LL, Perkins AC, Bieker JJ. Neomorphic effects of the neonatal anemia (Nan-Eklf) mutation contribute to deficits throughout development. Development 2017; 144:430-440. [PMID: 28143845 DOI: 10.1242/dev.145656] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/18/2016] [Indexed: 12/20/2022]
Abstract
Transcription factor control of cell-specific downstream targets can be significantly altered when the controlling factor is mutated. We show that the semi-dominant neonatal anemia (Nan) mutation in the EKLF/KLF1 transcription factor leads to ectopic expression of proteins that are not normally expressed in the red blood cell, leading to systemic effects that exacerbate the intrinsic anemia in the adult and alter correct development in the early embryo. Even when expressed as a heterozygote, the Nan-EKLF protein accomplishes this by direct binding and aberrant activation of genes encoding secreted factors that exert a negative effect on erythropoiesis and iron use. Our data form the basis for a novel mechanism of physiological deficiency that is relevant to human dyserythropoietic anemia and likely other disease states.
Collapse
Affiliation(s)
- Antanas Planutis
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Li Xue
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Cecelia D Trainor
- Laboratory of Molecular Biology, NIDDK, NIH, Bethesda, MD 20892, USA
| | - Mohan Dangeti
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Kevin Gillinder
- Mater Research Institute, University of Queensland, Woolloongabba QLD 4102, Queensland, Australia
| | - Miroslawa Siatecka
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA.,Department of Genetics, University of Adam Mickiewicz, Poznan 61-614, Poland
| | | | | | - Andrew C Perkins
- Mater Research Institute, University of Queensland, Woolloongabba QLD 4102, Queensland, Australia.,Princess Alexandra Hospital, Brisbane QLD 4102, Queensland, Australia
| | - James J Bieker
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA .,Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.,Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.,Mindich Child Health and Development Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| |
Collapse
|
11
|
Morin-Doré L, Blondin P, Vigneault C, Grand FX, Labrecque R, Sirard MA. Transcriptomic evaluation of bovine blastocysts obtained from peri-pubertal oocyte donors. Theriogenology 2017; 93:111-123. [PMID: 28257859 DOI: 10.1016/j.theriogenology.2017.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/09/2016] [Accepted: 01/03/2017] [Indexed: 12/18/2022]
Abstract
Assisted reproduction technologies (ART) and high selection pressure in the dairy industry are leading towards the use of younger females for reproduction, thereby reducing the interval between generations. This situation may have a negative impact on embryo quality, thus reducing the success rate of the procedures. This study aimed to document the effects of oocyte donor age on embryo quality, at the transcriptomic level, in order to characterize the effects of using young females for reproduction purpose. Young Holstein heifers (n = 10) were used at three different ages for ovarian stimulation protocols and oocyte collections (at 8, 11 and 14 months). All of the oocytes were fertilized in vitro with the semen of one adult bull, generating three lots of embryos per animal. Each animal was its own control for the evaluation of the effects of age. The EmbryoGENE platform was used for the assessment of gene expression patterns at the blastocyst stage. Embryos from animals at 8 vs 14 months and at 11 vs 14 months were used for microarray hybridization. Validation was done by performing RT-qPCR on seven candidate genes. Age-related contrast analysis (8 vs 14 mo and 11 vs 14 mo) identified 242 differentially expressed genes (DEGs) for the first contrast, and 296 for the second. The analysis of the molecular and biological functions of the DEGs suggests a metabolic cause to explain the differences that are observed between embryos from immature and adult subjects. The mTOR and PPAR signaling pathways, as well as the NRF2-mediated oxidative stress response pathways were among the gene expression pathways affected by donor age. In conclusion, the main differences between embryos produced at peri-pubertal ages are related to metabolic conditions resulting in a higher impact of in vitro conditions on blastocyts from younger heifers.
Collapse
Affiliation(s)
- Léonie Morin-Doré
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Département des Sciences Animales, Faculté des Sciences de l'Agriculture et de l'Alimentation, Université Laval, Québec, Canada
| | | | | | | | | | - Marc-André Sirard
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Département des Sciences Animales, Faculté des Sciences de l'Agriculture et de l'Alimentation, Université Laval, Québec, Canada.
| |
Collapse
|
12
|
Chal J, Al Tanoury Z, Hestin M, Gobert B, Aivio S, Hick A, Cherrier T, Nesmith AP, Parker KK, Pourquié O. Generation of human muscle fibers and satellite-like cells from human pluripotent stem cells in vitro. Nat Protoc 2016; 11:1833-50. [PMID: 27583644 DOI: 10.1038/nprot.2016.110] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Progress toward finding a cure for muscle diseases has been slow because of the absence of relevant cellular models and the lack of a reliable source of muscle progenitors for biomedical investigation. Here we report an optimized serum-free differentiation protocol to efficiently produce striated, millimeter-long muscle fibers together with satellite-like cells from human pluripotent stem cells (hPSCs) in vitro. By mimicking key signaling events leading to muscle formation in the embryo, in particular the dual modulation of Wnt and bone morphogenetic protein (BMP) pathway signaling, this directed differentiation protocol avoids the requirement for genetic modifications or cell sorting. Robust myogenesis can be achieved in vitro within 1 month by personnel experienced in hPSC culture. The differentiating culture can be subcultured to produce large amounts of myogenic progenitors amenable to numerous downstream applications. Beyond the study of myogenesis, this differentiation method offers an attractive platform for the development of relevant in vitro models of muscle dystrophies and drug screening strategies, as well as providing a source of cells for tissue engineering and cell therapy approaches.
Collapse
Affiliation(s)
- Jérome Chal
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Marie Hestin
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Bénédicte Gobert
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Suvi Aivio
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Aurore Hick
- Anagenesis Biotechnologies, Parc d'innovation, Illkirch-Graffenstaden, France
| | - Thomas Cherrier
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Alexander P Nesmith
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Kevin K Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Knorr DA, Wang H, Aurora M, MacMillan ML, Holtan SG, Bergerson R, Cao Q, Weisdorf DJ, Cooley S, Brunstein C, Miller JS, Wagner JE, Blazar BR, Verneris MR. Loss of T Follicular Helper Cells in the Peripheral Blood of Patients with Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2016; 22:825-33. [PMID: 26806586 PMCID: PMC5015683 DOI: 10.1016/j.bbmt.2016.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
Abstract
B cell antihost antibody production plays a central role in chronic graft-versus-host disease (cGVHD). T follicular helper (TFH) cells drive B cell responses and are implicated in this process. Given differences in cGVHD incidence between umbilical cord blood (UCB) and adult donor transplant recipients, we evaluated TFH cell reconstitution kinetics to define graft source differences and their potential pathogenic role in cGVHD. Although we observed significantly fewer TFH cells in the blood of UCB recipients (versus matched related donors [MRD]) early after transplantation, by 1 year the numbers of TFH cells were similar. Additionally, at both early (day 60) and late (1 year) time points, TFH cell phenotype was predominantly central memory cells in both cohorts. TFH cells were functional and able to produce multiple cytokines (INF-γ, TNF-α, IL-2, IL-17, and IL-21) after stimulation. In contrast to mouse models, where an enhanced frequency of splenic TFH cells contributes to cGVHD, patients with cGVHD showed significantly depleted circulating TFH cells after both UCB and MRD transplantation. Low numbers of TFH cells early after UCB transplantation could directly contribute to less cGVHD in this cohort. Additionally, systemic therapy (including steroids and calcineurin inhibitors) may contribute to decreases in TFH cells in patients with cGVHD. These data provide further evidence supporting the importance of TFH cells in cGVHD pathogenesis.
Collapse
Affiliation(s)
- David A Knorr
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Hongbo Wang
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Mukta Aurora
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Margaret L MacMillan
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Shernan G Holtan
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Rachel Bergerson
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Qing Cao
- Division of Blood and Marrow Transplantation, Department of Biostatistics, University of Minnesota, Minnesota
| | - Daniel J Weisdorf
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Sarah Cooley
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Claudio Brunstein
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Jeffery S Miller
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - John E Wagner
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Michael R Verneris
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota.
| |
Collapse
|
14
|
Lohmann F, Dangeti M, Soni S, Chen X, Planutis A, Baron MH, Choi K, Bieker JJ. The DEK Oncoprotein Is a Critical Component of the EKLF/KLF1 Enhancer in Erythroid Cells. Mol Cell Biol 2015; 35:3726-38. [PMID: 26303528 PMCID: PMC4589598 DOI: 10.1128/mcb.00382-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 05/06/2015] [Accepted: 08/17/2015] [Indexed: 02/07/2023] Open
Abstract
Understanding how transcriptional regulators are themselves controlled is important in attaining a complete picture of the intracellular effects that follow signaling cascades during early development and cell-restricted differentiation. We have addressed this issue by focusing on the regulation of EKLF/KLF1, a zinc finger transcription factor that plays a necessary role in the global regulation of erythroid gene expression. Using biochemical affinity purification, we have identified the DEK oncoprotein as a critical factor that interacts with an essential upstream enhancer element of the EKLF promoter and exerts a positive effect on EKLF levels. This element also binds a core set of erythroid transcription factors, suggesting that DEK is part of a tissue-restricted enhanceosome that contains BMP4-dependent and -independent components. Together with local enrichment of properly coded histones and an open chromatin domain, optimal transcriptional activation of the EKLF locus can be established.
Collapse
Affiliation(s)
- Felix Lohmann
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Mohan Dangeti
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Shefali Soni
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Xiaoyong Chen
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Antanas Planutis
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Margaret H Baron
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA Department of Medicine, Mount Sinai School of Medicine, New York, New York, USA
| | - Kyunghee Choi
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - James J Bieker
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| |
Collapse
|
15
|
Blobel GA, Bodine D, Brand M, Crispino J, de Bruijn MFTR, Nathan D, Papayannopoulou T, Porcher C, Strouboulis J, Zon L, Higgs DR, Stamatoyannopoulos G, Engel JD. An international effort to cure a global health problem: A report on the 19th Hemoglobin Switching Conference. Exp Hematol 2015; 43:821-37. [PMID: 26143582 DOI: 10.1016/j.exphem.2015.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 12/24/2022]
Abstract
Every 2 years since 1978, an international group of scientists, physicians, and other researchers meet to discuss the latest developments in the underlying etiology, mechanisms of action, and developmental acquisition of cellular and systemic defects exhibited and elicited by the most common inherited human disorders, the hemoglobinopathies. The 19th Hemoglobin Switching Conference, held in September 2014 at St. John's College in Oxford, once again exceeded all expectations by describing cutting edge research in cellular, molecular, developmental, and genomic advances focused on these diseases. The conference comprised about 60 short talks over 3 days by leading investigators in the field. This meeting report describes the highlights of the conference.
Collapse
Affiliation(s)
- Gerd A Blobel
- Division of Hematology, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Bodine
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marjorie Brand
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - John Crispino
- Division of Hematology/Oncology, Robert H Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Marella F T R de Bruijn
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital University of Oxford, Oxford, UK; BRC Blood Theme, NIHR Oxford Biomedical Centre, Oxford University Hospital, Oxford, UK
| | - David Nathan
- Division of Hematology and Oncology, Boston Children's Hospital, Departments of Pediatrics and Medicine, Harvard Medical School, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Catherine Porcher
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital University of Oxford, Oxford, UK; BRC Blood Theme, NIHR Oxford Biomedical Centre, Oxford University Hospital, Oxford, UK
| | - John Strouboulis
- Division of Molecular Oncology, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Len Zon
- Boston Children's Hospital/HHMI, Boston, MA, USA
| | - Douglas R Higgs
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital University of Oxford, Oxford, UK; BRC Blood Theme, NIHR Oxford Biomedical Centre, Oxford University Hospital, Oxford, UK
| | | | - James Douglas Engel
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
Perna F, Vu LP, Themeli M, Kriks S, Hoya-Arias R, Khanin R, Hricik T, Mansilla-Soto J, Papapetrou EP, Levine RL, Studer L, Sadelain M, Nimer SD. The polycomb group protein L3MBTL1 represses a SMAD5-mediated hematopoietic transcriptional program in human pluripotent stem cells. Stem Cell Reports 2015; 4:658-69. [PMID: 25754204 PMCID: PMC4400644 DOI: 10.1016/j.stemcr.2015.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 01/11/2023] Open
Abstract
Epigenetic regulation of key transcriptional programs is a critical mechanism that controls hematopoietic development, and, thus, aberrant expression patterns or mutations in epigenetic regulators occur frequently in hematologic malignancies. We demonstrate that the Polycomb protein L3MBTL1, which is monoallelically deleted in 20q- myeloid malignancies, represses the ability of stem cells to drive hematopoietic-specific transcriptional programs by regulating the expression of SMAD5 and impairing its recruitment to target regulatory regions. Indeed, knockdown of L3MBTL1 promotes the development of hematopoiesis and impairs neural cell fate in human pluripotent stem cells. We also found a role for L3MBTL1 in regulating SMAD5 target gene expression in mature hematopoietic cell populations, thereby affecting erythroid differentiation. Taken together, we have identified epigenetic priming of hematopoietic-specific transcriptional networks, which may assist in the development of therapeutic approaches for patients with anemia. L3MBTL1 is a chromatin-binding protein that represses SMAD5 expression Lack of L3MBTL1 primes the hematopoietic development of pluripotent stem cells L3MBTL1 regulates erythroid differentiation
Collapse
Affiliation(s)
- Fabiana Perna
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Ly P Vu
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria Themeli
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sonja Kriks
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ruben Hoya-Arias
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Raya Khanin
- Bioinformatics Core, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Todd Hricik
- Human Oncology and Pathogenesis Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jorge Mansilla-Soto
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Ross L Levine
- Human Oncology and Pathogenesis Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lorenz Studer
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michel Sadelain
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen D Nimer
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
17
|
Chung D, Gao F, Jegga AG, Das SK. Estrogen mediated epithelial proliferation in the uterus is directed by stromal Fgf10 and Bmp8a. Mol Cell Endocrinol 2015; 400:48-60. [PMID: 25451979 PMCID: PMC4751583 DOI: 10.1016/j.mce.2014.11.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 11/25/2022]
Abstract
To define endometrial stromal-derived paracrine mediators that participate in estradiol-17β (E2)-induced epithelial proliferation, microarray analysis of gene expression was carried out in mouse uterine epithelial-stromal co-culture systems under the condition of E2 or vehicle (control). Our results demonstrated gene alteration by E2: in epithelial cells, we found up-regulation of 119 genes and down-regulation of 28 genes, while in stroma cells we found up-regulation of 144 genes and down-regulation of 184 genes. A functional enrichment analysis of the upregulated epithelial genes implicated them for proliferation, while upregulated stromal genes were associated with extracellular functions. Quantitative RT-PCR and in situ hybridization results confirmed differential gene expression in both cell cultures and ovariectomized uteri after the above treatments. Based on our identification of stromal secretory factors, we found evidence that suppression by siRNA specifically for Bmp8a and/or Fgf10 in the stromal layer caused significant inhibition of proliferation by E2 in the co-culture system, suggesting Bmp8a and Fgf10 act as paracrine mediators during E2-dependent control of uterine proliferation. The localization of receptors and receptor activation signaling in epithelial cells in both the co-culture system and uteri was consistent with their involvement in ligand-receptor signaling. Interestingly, loss of Bmp8a or Fgf10 also caused abrogation of E2-regulated epithelial receptor signaling in co-culture systems, suggesting that stroma-derived Fgf10 and Bmp8a are responsible for epithelial communication. Overall, stromal Fgf10 and Bmp8a serve as potential paracrine factors for E2-dependent regulation of epithelial proliferation in the uterus.
Collapse
Affiliation(s)
- Daesuk Chung
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Fei Gao
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Anil G Jegga
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Sanjoy K Das
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
18
|
Transcription factor EKLF (KLF1) recruitment of the histone chaperone HIRA is essential for β-globin gene expression. Proc Natl Acad Sci U S A 2014; 111:13337-42. [PMID: 25197097 DOI: 10.1073/pnas.1405422111] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The binding of chromatin-associated proteins and incorporation of histone variants correlates with alterations in gene expression. These changes have been particularly well analyzed at the mammalian β-globin locus, where transcription factors such as erythroid Krüppel-like factor (EKLF), which is also known as Krüppel-like factor 1 (KLF1), play a coordinating role in establishing the proper chromatin structure and inducing high-level expression of adult β-globin. We had previously shown that EKLF preferentially interacts with histone H3 and that the H3.3 variant is differentially recruited to the β-globin promoter. We now find that a novel interaction between EKLF and the histone cell cycle regulation defective homolog A (HIRA) histone chaperone accounts for these effects. HIRA is not only critical for β-globin expression but is also required for activation of the erythropoietic regulators EKLF and GATA binding protein 1 (GATA1). Our results provide a mechanism by which transcription factor-directed recruitment of a generally expressed histone chaperone can lead to tissue-restricted changes in chromatin components, structure, and transcription at specific genomic sites during differentiation.
Collapse
|
19
|
Xue L, Galdass M, Gnanapragasam MN, Manwani D, Bieker JJ. Extrinsic and intrinsic control by EKLF (KLF1) within a specialized erythroid niche. Development 2014; 141:2245-54. [PMID: 24866116 DOI: 10.1242/dev.103960] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The erythroblastic island provides an important nutritional and survival support niche for efficient erythropoietic differentiation. Island integrity is reliant on adhesive interactions between erythroid and macrophage cells. We show that erythroblastic islands can be formed from single progenitor cells present in differentiating embryoid bodies, and that these correspond to erythro-myeloid progenitors (EMPs) that first appear in the yolk sac of the early developing embryo. Erythroid Krüppel-like factor (EKLF; KLF1), a crucial zinc finger transcription factor, is expressed in the EMPs, and plays an extrinsic role in erythroid maturation by being expressed in the supportive macrophage of the erythroblastic island and regulating relevant genes important for island integrity within these cells. Together with its well-established intrinsic contributions to erythropoiesis, EKLF thus plays a coordinating role between two different cell types whose interaction provides the optimal environment to generate a mature red blood cell.
Collapse
Affiliation(s)
- Li Xue
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Mariann Galdass
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Merlin Nithya Gnanapragasam
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Deepa Manwani
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - James J Bieker
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| |
Collapse
|
20
|
Hewitt KJ, Sanalkumar R, Johnson KD, Keles S, Bresnick EH. Epigenetic and genetic mechanisms in red cell biology. Curr Opin Hematol 2014; 21:155-64. [PMID: 24722192 PMCID: PMC6061918 DOI: 10.1097/moh.0000000000000034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Erythropoiesis, in which hematopoietic stem cells (HSCs) generate lineage-committed progenitors that mature into erythrocytes, is regulated by numerous chromatin modifying and remodeling proteins. We will focus on how epigenetic and genetic mechanisms mesh to establish the erythroid transcriptome and how studying erythropoiesis can yield genomic principles. RECENT FINDINGS Trans-acting factor binding to small DNA motifs (cis-elements) underlies regulatory complex assembly at specific chromatin sites, and therefore unique transcriptomes. As cis-elements are often very small, thousands or millions of copies of a given element reside in a genome. Chromatin restricts factor access in a context-dependent manner, and cis-element-binding factors recruit chromatin regulators that mediate functional outputs. Technologies to map chromatin attributes of loci in vivo, to edit genomes and to sequence whole genomes have been transformative in discovering critical cis-elements linked to human disease. SUMMARY Cis-elements mediate chromatin-targeting specificity, and chromatin regulators dictate cis-element accessibility/function, illustrating an amalgamation of genetic and epigenetic mechanisms. Cis-elements often function ectopically when studied outside of their endogenous loci, and complex strategies to identify nonredundant cis-elements require further development. Facile genome-editing technologies provide a new approach to address this problem. Extending genetic analyses beyond exons and promoters will yield a rich pipeline of cis-element alterations with importance for red cell biology and disease.
Collapse
Affiliation(s)
- Kyle J. Hewitt
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health
- UW-Madison Blood Research Program, Carbone Cancer Center
| | - Rajendran Sanalkumar
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health
- UW-Madison Blood Research Program, Carbone Cancer Center
| | - Kirby D. Johnson
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health
- UW-Madison Blood Research Program, Carbone Cancer Center
| | - Sunduz Keles
- UW-Madison Blood Research Program, Carbone Cancer Center
- Department of Biostatistics and Medical Informatics, Department of Statistics, Wisconsin Institutes for Medical Research, Madison, Wisconsin, USA
| | - Emery H. Bresnick
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health
- UW-Madison Blood Research Program, Carbone Cancer Center
| |
Collapse
|
21
|
Hou W, Xiong L, Li S, Wang Y, Jiang Z, Cheng S, Liu Y, Xiao J, Guo H, Wang Z. A continuous electromagnetic radiation exposure affected the expressions ofClockandfviigenes in mice. BIOL RHYTHM RES 2013. [DOI: 10.1080/09291016.2013.770295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
22
|
Three fingers on the switch: Krüppel-like factor 1 regulation of γ-globin to β-globin gene switching. Curr Opin Hematol 2013; 20:193-200. [PMID: 23474875 DOI: 10.1097/moh.0b013e32835f59ba] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Krüppel-like factor 1 (KLF1) regulates most aspects of erythropoiesis. Many years ago, transgenic mouse studies implicated KLF1 in the control of the human γ-globin to β-globin switch. In this review, we will integrate these initial studies with recent developments in human genetics to discuss our present understanding of how KLF1 and its target genes direct the switch. RECENT FINDINGS Recent studies have shown that human mutations in KLF1 are common and mostly asymptomatic, but lead to significant increases in levels of fetal hemoglobin (HbF) (α2γ2) and adult HbA2 (α2δ2). Genome-wide association studies (GWAS) have demonstrated that three primary loci are associated with increased HbF levels in the population: the β-globin locus itself, the BCL11A locus, and a site between MYB and HBS1L. We discuss evidence that KLF1 directly regulates BCL11A, MYB and other genes, which are involved directly or indirectly in γ-globin silencing, thus providing a link between GWAS and KLF1 in hemoglobin switching. SUMMARY KLF1 regulates the γ-globin to β-globin genetic switch by many mechanisms. Firstly, it facilitates formation of an active chromatin hub (ACH) at the β-globin gene cluster. Specifically, KLF1 conscripts the adult-stage β-globin gene to replace the γ-globin gene within the ACH in a stage-specific manner. Secondly, KLF1 acts as a direct activator of genes that encode repressors of γ-globin gene expression. Finally, KLF1 is a regulator of many components of the cell cycle machinery. We suggest that dysregulation of these genes leads to cell cycle perturbation and 'erythropoietic stress' leading to indirect upregulation of HbF.
Collapse
|
23
|
Yien YY, Bieker JJ. EKLF/KLF1, a tissue-restricted integrator of transcriptional control, chromatin remodeling, and lineage determination. Mol Cell Biol 2013; 33:4-13. [PMID: 23090966 PMCID: PMC3536305 DOI: 10.1128/mcb.01058-12] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Erythroid Krüppel-like factor (EKLF or KLF1) is a transcriptional regulator that plays a critical role in lineage-restricted control of gene expression. KLF1 expression and activity are tightly controlled in a temporal and differentiation stage-specific manner. The mechanisms by which KLF1 is regulated encompass a range of biological processes, including control of KLF1 RNA transcription, protein stability, localization, and posttranslational modifications. Intact KLF1 regulation is essential to correctly regulate erythroid function by gene transcription and to maintain hematopoietic lineage homeostasis by ensuring a proper balance of erythroid/megakaryocytic differentiation. In turn, KLF1 regulates erythroid biology by a wide variety of mechanisms, including gene activation and repression by regulation of chromatin configuration, transcriptional initiation and elongation, and localization of gene loci to transcription factories in the nucleus. An extensive series of biochemical, molecular, and genetic analyses has uncovered some of the secrets of its success, and recent studies are highlighted here. These reveal a multilayered set of control mechanisms that enable efficient and specific integration of transcriptional and epigenetic controls and that pave the way for proper lineage commitment and differentiation.
Collapse
Affiliation(s)
- Yvette Y. Yien
- Department of Developmental and Regenerative Biology
- Graduate School of Biological Sciences
| | - James J. Bieker
- Department of Developmental and Regenerative Biology
- Black Family Stem Cell Institute
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| |
Collapse
|
24
|
Kang YJ, Shin JW, Yoon JH, Oh IH, Lee SP, Kim SY, Park SH, Mamura M. Inhibition of erythropoiesis by Smad6 in human cord blood hematopoietic stem cells. Biochem Biophys Res Commun 2012; 423:750-6. [DOI: 10.1016/j.bbrc.2012.06.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 06/08/2012] [Indexed: 11/24/2022]
|
25
|
Rhee JM, Iannaccone PM. Mapping mouse hemangioblast maturation from headfold stages. Dev Biol 2012; 365:1-13. [PMID: 22426104 DOI: 10.1016/j.ydbio.2012.02.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 02/14/2012] [Accepted: 02/15/2012] [Indexed: 11/18/2022]
Abstract
The mouse posterior primitive streak at neural plate/headfold stages (NP/HF, ~7.5 dpc-8 dpc) represents an optimal window from which hemangioblasts can be isolated. We performed immunohistochemistry on this domain using established monoclonal antibodies for proteins that affect blood and endothelial fates. We demonstrate that HoxB4 and GATA1 are the first set of markers that segregate independently to endothelial or blood populations during NP/HF stages of mouse embryonic development. In a subset of cells, both proteins are co-expressed and immunoreactivities appear mutually excluded within nuclear spaces. We searched for this particular state at later sites of hematopoietic stem cell emergence, viz., the aorta-gonad-mesonephros (AGM) and the fetal liver at 10.5-11.5 dpc, and found that only a rare number of cells displayed this character. Based on this spatial-temporal argument, we propose that the earliest blood progenitors emerge either directly from the epiblast or through segregation within the allantoic core domain (ACD) through reduction of cell adhesion and pSmad1/5 nuclear signaling, followed by a stochastic decision toward a blood or endothelial fate that involves GATA1 and HoxB4, respectively. A third form in which binding distributions are balanced may represent a common condition shared by hemangioblasts and HSCs. We developed a heuristic model of hemangioblast maturation, in part, to be explicit about our assumptions.
Collapse
Affiliation(s)
- Jerry M Rhee
- Children's Memorial Research Center, Department of Pediatrics, Developmental Biology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | | |
Collapse
|
26
|
Kang K, Jung H, Nam S, Lim JS. NDRG2 Promotes GATA-1 Expression through Regulation of the JAK2/STAT Pathway in PMA-stimulated U937 Cells. Immune Netw 2011; 11:348-57. [PMID: 22346774 PMCID: PMC3275703 DOI: 10.4110/in.2011.11.6.348] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Revised: 10/18/2011] [Accepted: 10/29/2011] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND N-myc downstream-regulated gene 2 (NDRG2), a member of a newly described family of differentiation-related genes, has been characterized as a regulator of dendritic cells. However, the role of NDRG2 on the expression and activation of transcription factors in blood cells remains poorly understood. In this study, we investigated the effects of NDRG2 overexpression on GATA-1 expression in PMA-stimulated U937 cells. METHODS We generated NDRG2-overexpressing U937 cell line (U937-NDRG2) and treated the cells with PMA to investigate the role of NDRG2 on GATA-1 expression. RESULTS NDRG2 overexpression in U937 cells significantly induced GATA-1 expression in response to PMA stimulation. Interestingly, JAK2/STAT and BMP-4/Smad pathways associated with the induction of GATA-1 were activated in PMA-stimulated U937-NDRG2 cells. We found that the inhibition of JAK2 activation, but not of BMP-4/Smad signaling, can elicit a decrease of PMA-induced GATA-1 expression in U937-NDRG2 cells. CONCLUSION The results reveal that NDRG2 promotes the expression of GATA-1 through activation of the JAK2/STAT pathway, but not through the regulation of the BMP-4/Smad pathway in U937 cells. Our findings further suggest that NDRG2 may play a role as a regulator of erythrocyte and megakaryocyte differentiation during hematopoiesis.
Collapse
Affiliation(s)
- Kyeongah Kang
- Department of Biological Science and the Research Center for Women's Disease, Sookmyung Women's University, Seoul 140-742, Korea
| | | | | | | |
Collapse
|
27
|
Blancas AA, Shih AJ, Lauer NE, McCloskey KE. Endothelial cells from embryonic stem cells in a chemically defined medium. Stem Cells Dev 2011; 20:2153-61. [PMID: 21446878 PMCID: PMC3225059 DOI: 10.1089/scd.2010.0432] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 03/29/2011] [Indexed: 11/12/2022] Open
Abstract
Endothelial cells (ECs) are desired for their therapeutic potential in a variety of areas including gene therapy, cardiac regeneration, development of tissue-engineered vascular grafts, and prevascularized tissue transplants. Pluripotent embryonic stem cells (ESCs) can be induced to differentiate into ECs in vitro using embryoid bodies, monolayer cultures, or by genetic manipulation and immortalization. However, obtaining homogeneous cultures of proliferating ESC-derived ECs without genetic manipulation is a challenging undertaking and often requires optimization of protocols and rigorous purification techniques. Moreover, current differentiation methods that use medium containing fetal calf or bovine serum components introduce additional challenges because of our limited ability to control the differentiation signals and batch-to-batch variations of serum. We have explored the development of new medium formulations for deriving ECs from murine embryonic stem cells (mESCs) using only chemically defined reagents. We present 2 different medium formulations along with the detailed methodologies, including the optimization of extracellular matrix-derived substrates known to play a role in cell attachment and proliferation as well as cell differentiation. Characterization of the ESC-derived ECs indicate that (1) chemically defined medium formulations reproducibly generate superior ECs compared with previous serum-containing formulations, (2) fibronectin, and not collagen type-IV, is the optimal substrate for EC induction in our chemically defined medium formulations, (3) without additional activation of Notch-signaling, ESC-ECs develop predominantly into venous ECs, and (4) using these medium formulations, a second rigorous selection step is not required to generate proliferating ECs from ESCs, but it does enhance the final purity of the ECs.
Collapse
Affiliation(s)
- Alicia A. Blancas
- Graduate Program in Quantitative and Systems Biology, University of California, Merced, California
| | - Albert J. Shih
- School of Engineering, University of California, Merced, California
| | - Nicholas E. Lauer
- Graduate Program in Biological Engineering and Small-Scale Technologies, University of California, Merced, California
| | - Kara E. McCloskey
- Graduate Program in Quantitative and Systems Biology, University of California, Merced, California
- School of Engineering, University of California, Merced, California
- Graduate Program in Biological Engineering and Small-Scale Technologies, University of California, Merced, California
| |
Collapse
|
28
|
Abstract
Erythroid cells and megakaryocytes are derived from a common precursor, the megakaryocyte-erythroid progenitor. Although these 2 closely related hematopoietic cell types share many transcription factors, there are several key differences in their regulatory networks that lead to differential gene expression downstream of the megakaryocyte-erythroid progenitor. With the advent of next-generation sequencing and our ability to precisely define transcription factor chromatin occupancy in vivo on a global scale, we are much closer to understanding how these 2 lineages are specified and in general how transcription factor complexes govern hematopoiesis.
Collapse
|
29
|
Smad1 signaling restricts hematopoietic potential after promoting hemangioblast commitment. Blood 2011; 117:6489-97. [PMID: 21515822 DOI: 10.1182/blood-2010-10-312389] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bone morphogenetic protein (BMP) signaling regulates embryonic hematopoiesis via receptor-mediated activation of downstream SMAD proteins, including SMAD1. In previous work, we showed that Smad1 expression is sufficient to enhance commitment of mesoderm to hemangioblast fate. We also found indirect evidence to support a subsequent repressive function for Smad1 in hematopoiesis. To test this hypothesis directly, we developed a novel system allowing temporal control of Smad1 levels by conditional knockdown in embryonic stem cell derivatives. Depletion of Smad1 in embryoid body cultures before hemangioblast commitment limits hematopoietic potential because of a block in mesoderm development. Conversely, when Smad1 is depleted in FlK1(+) mesoderm, at a stage after hemangioblast commitment, the pool of hematopoietic progenitors is expanded. This involves enhanced expression levels for genes specific to hematopoiesis, including Gata1, Runx1 and Eklf, rather than factors required for earlier specification of the hemangioblast. The phenotype correlates with increased nuclear SMAD2 activity, indicating molecular cross-regulation between the BMP and TGF-β signaling pathways. Consistent with this mechanism, hematopoiesis was enhanced when Smad2 was directly expressed during this same developmental window. Therefore, this study reveals a temporally defined function for Smad1 in restricting the expansion of early hematopoietic progenitors.
Collapse
|
30
|
Abstract
The Krüppel-like factor (KLF) family of transcription factors regulates diverse biological processes that include proliferation, differentiation, growth, development, survival, and responses to external stress. Seventeen mammalian KLFs have been identified, and numerous studies have been published that describe their basic biology and contribution to human diseases. KLF proteins have received much attention because of their involvement in the development and homeostasis of numerous organ systems. KLFs are critical regulators of physiological systems that include the cardiovascular, digestive, respiratory, hematological, and immune systems and are involved in disorders such as obesity, cardiovascular disease, cancer, and inflammatory conditions. Furthermore, KLFs play an important role in reprogramming somatic cells into induced pluripotent stem (iPS) cells and maintaining the pluripotent state of embryonic stem cells. As research on KLF proteins progresses, additional KLF functions and associations with disease are likely to be discovered. Here, we review the current knowledge of KLF proteins and describe common attributes of their biochemical and physiological functions and their pathophysiological roles.
Collapse
Affiliation(s)
- Beth B McConnell
- Departments of Medicine and of Hematology and Medical Oncology, Emory University School of Medicine,Atlanta, Georgia 30322, USA
| | | |
Collapse
|
31
|
Claudin 13, a member of the claudin family regulated in mouse stress induced erythropoiesis. PLoS One 2010; 5. [PMID: 20844758 PMCID: PMC2937028 DOI: 10.1371/journal.pone.0012667] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 08/15/2010] [Indexed: 11/19/2022] Open
Abstract
Mammals are able to rapidly produce red blood cells in response to stress. The molecular pathways used in this process are important in understanding responses to anaemia in multiple biological settings. Here we characterise the novel gene Claudin 13 (Cldn13), a member of the Claudin family of tight junction proteins using RNA expression, microarray and phylogenetic analysis. We present evidence that Cldn13 appears to be co-ordinately regulated as part of a stress induced erythropoiesis pathway and is a mouse-specific gene mainly expressed in tissues associated with haematopoietic function. CLDN13 phylogenetically groups with its genomic neighbour CLDN4, a conserved tight junction protein with a putative role in epithelial to mesenchymal transition, suggesting a recent duplication event. Mechanisms of mammalian stress erythropoiesis are of importance in anaemic responses and expression microarray analyses demonstrate that Cldn13 is the most abundant Claudin in spleen from mice infected with Trypanosoma congolense. In mice prone to anaemia (C57BL/6), its expression is reduced compared to strains which display a less severe anaemic response (A/J and BALB/c) and is differentially regulated in spleen during disease progression. Genes clustering with Cldn13 on microarrays are key regulators of erythropoiesis (Tal1, Trim10, E2f2), erythrocyte membrane proteins (Rhd and Gypa), associated with red cell volume (Tmcc2) and indirectly associated with erythropoietic pathways (Cdca8, Cdkn2d, Cenpk). Relationships between genes appearing co-ordinately regulated with Cldn13 post-infection suggest new insights into the molecular regulation and pathways involved in stress induced erythropoiesis and suggest a novel, previously unreported role for claudins in correct cell polarisation and protein partitioning prior to erythroblast enucleation.
Collapse
|
32
|
Yue F, Johkura K, Tomotsune D, Shirasawa S, Yokoyama T, Nagai M, Sasaki K. Bone marrow stromal cells as an inducer for cardiomyocyte differentiation from mouse embryonic stem cells. Ann Anat 2010; 192:314-21. [DOI: 10.1016/j.aanat.2010.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 07/27/2010] [Accepted: 07/28/2010] [Indexed: 01/10/2023]
|
33
|
Abstract
PURPOSE OF REVIEW The role of bone morphogenetic proteins (BMPs) in vasculogenesis is still not well understood, despite many recent developments in this area of research. In this review, we discuss the most recent studies that identify new critical mechanisms through which BMP signaling acts with a focus on angiogenesis. RECENT FINDINGS New evidence brought to light over the last few years suggests that BMP-binding proteins, formerly thought of as antagonists, can also increase BMP activity under certain conditions. It has also recently been determined that components of the extracellular matrix are involved in the BMP signaling pathways that regulate angiogenesis. Through the BMP pathway, myosin-X and cyclooxygenase 2 serve as target genes that have been determined to play a role in blood vessel formation. BMPs also conduct Smad-independent signaling and crosstalk with other pathways. Finally, BMPs have been shown to play an antiangiogenic role in specific settings. SUMMARY Better understanding of the BMP signaling pathway and its regulators can have potentially great effects on therapeutic strategies from cardiovascular disease to cancer.
Collapse
|
34
|
Blancas AA, Lauer NE, McCloskey KE. Endothelial differentiation of embryonic stem cells. ACTA ACUST UNITED AC 2008; Chapter 1:Unit 1F.5. [PMID: 18819086 DOI: 10.1002/9780470151808.sc01f05s6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Vascular progenitor cells derived from stem cells could potentially lead to a variety of clinically relevant applications, including cell-based therapies and tissue engineering. Here, we describe methods for isolating purified proliferating populations of vascular endothelial cells from mouse embryonic stem cells (mESC) using Flk-1 positive sorted cells, VEGF supplementation, and a rigorous manual selection technique required for endothelial cell purification and expansion. Using this in vitro derivation procedure, it is possible to obtain millions of cells at various stages of differentiation, with the potential for up to 25 population doublings.
Collapse
Affiliation(s)
- Alicia A Blancas
- Graduate Program in Quantitative and Systems Biology, University of California at Merced, California, USA
| | | | | |
Collapse
|
35
|
McReynolds LJ, Tucker J, Mullins MC, Evans T. Regulation of hematopoiesis by the BMP signaling pathway in adult zebrafish. Exp Hematol 2008; 36:1604-1615. [PMID: 18973974 DOI: 10.1016/j.exphem.2008.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2007] [Revised: 08/12/2008] [Accepted: 08/21/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The zebrafish is an established model system for studying the embryonic emergence of tissues and organs, including the hematopoietic system. We hypothesized that key signaling pathways controlling embryonic hematopoiesis continue to be important in the adult, and we sought to develop approaches to test this in zebrafish, focused on the bone morphogenetic protein (BMP) signaling pathway. Functions for this pathway in adult hematopoiesis have been challenging to probe in other models. MATERIALS AND METHODS Several approaches tested the function of BMP signaling during adult zebrafish hematopoiesis. First, we evaluated steady-state hematopoiesis in adult fish that are heterozygous for mutant alleles of Smad5, or are homozygous for mutant alleles, and rescued to adulthood by injection of RNA encoding Smad5. Second, we tested the relative ability of smad5 mutant fish to recover from hemolytic anemia. Third, we generated a transgenic line that targets the expression of a dominant-negative BMP receptor to adult-stage Gata1+ progenitor cells. RESULTS Adult fish with a strong mutant smad5 allele are anemic at steady state and, in addition, respond to hemolytic anemia with kinetics that are altered compared to wild-type fish. Fish expressing a mutant BMP receptor in early Gata1+ definitive progenitors generate excessive eosinophils. CONCLUSIONS Our study provides proof of principle that regulation of adult hematopoiesis can be studied in zebrafish by altering specific pathways. We show that the BMP signaling pathway is relevant for adult hematopoiesis to maintain steady state erythropoiesis, control the erythropoietic response following stress anemia, and to generate normal numbers of eosinophils.
Collapse
Affiliation(s)
- Lisa J McReynolds
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
36
|
Abstract
The hemangioblast hypothesis was proposed a century ago. The existence of hemangioblasts is now demonstrated in mouse and human embryonic stem cell (ESC)-derived embryoid bodies (EBs), in the mouse and zebrafish gastrula, and in adults. The hemangioblast is believed to derive from mesodermal cells, and is enriched in the Bry+Flk1+ and Flk1+Scl+ cell populations in EBs and in the posterior primitive streak of the mouse gastrula and in the ventral mesoderm of the zebrafish gastrula. However, recent studies suggest that the hemangioblast does not give rise to all endothelial and hematopoietic lineages in mouse and zebrafish embryos. Although several signaling pathways are known to involve the generation of hemangioblasts, it remains largely unknown how the hemangioblast is formed and what are the master genes controlling hemangioblast development. This review will summarize our current knowledge, challenges, and future directions on molecular and developmental aspects of the hemangioblast.
Collapse
Affiliation(s)
- Jing-Wei Xiong
- The Nephrology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 01219, USA.
| |
Collapse
|
37
|
Liu S, Bhattacharya S, Han A, Suragani RNVS, Zhao W, Fry RC, Chen JJ. Haem-regulated eIF2alpha kinase is necessary for adaptive gene expression in erythroid precursors under the stress of iron deficiency. Br J Haematol 2008; 143:129-37. [PMID: 18665838 DOI: 10.1111/j.1365-2141.2008.07293.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Haem-regulated eIF2alpha kinase (HRI) is essential for the regulation of globin gene translation and the survival of erythroid precursors in iron/haem deficiency. This study found that that in iron deficiency, fetal definitive erythropoiesis is inhibited at the basophilic erythroblast stage with increased proliferation and elevated apoptosis. This hallmark of ineffective erythropoiesis is more severe in HRI deficiency. Microarray gene profiling analysis showed that HRI was required for adaptive gene expression in erythroid precursors during chronic iron deficiency. The number of genes with expression affected more than twofold increased, from 213 in iron deficiency and 73 in HRI deficiency, to 3135 in combined iron and HRI deficiencies. Many of these genes are regulated by Gata1 and Fog1. We demonstrate for the first time that Gata1 expression in developing erythroid precursors is decreased in iron deficiency, and is decreased further in combined iron and HRI deficiencies. Additionally, Fog1 expression is decreased in combined deficiencies, but not in iron or HRI deficiency alone. Our results indicate that HRI confers adaptive gene expression in developing erythroblasts during iron deficiency through maintaining Gata1/Fog1 expression.
Collapse
Affiliation(s)
- Sijin Liu
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Analysis of the temporal and concentration-dependent effects of BMP-4, VEGF, and TPO on development of embryonic stem cell-derived mesoderm and blood progenitors in a defined, serum-free media. Exp Hematol 2008; 36:1186-98. [PMID: 18550259 DOI: 10.1016/j.exphem.2008.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 04/08/2008] [Accepted: 04/09/2008] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To develop a robust serum-free (SF) system for generation of hemogenic mesoderm and blood progenitors from pluripotent cells. MATERIALS AND METHODS Embryonic stem cells (ESCs) maintained in N2B27 supplemented with leukemia inhibitory factor (LIF) and bone morphogenetic protein (BMP)-4 were induced to differentiate into Brachyury/T-expressing cells (measured using a green fluorescent protein reporter) and myeloid-erythroid colony-forming cells (ME-CFCs), by removing LIF, changing the base media formulation, and via the time- and concentration-dependent addition of other factors. RESULTS Presence of 10 ng/mL BMP-4 permitted the emergence of cells expressing T and the vascular endothelial growth factor receptor (VEGFR)-2, however, <5% of the cells were double-positive on day 4. Adjusting the SF media formulation allowed only 5 ng/mL BMP-4 to yield 24% +/- 4% Brachyury-green fluorescent protein VEGFR-2(+) cells by day 4. These cells could develop into ME-CFC, producing 4.4 +/- 0.8 CFC per 1000 cells at day 8. We also examined the timing and concentration sensitivity of BMP-4, VEGF, and thrombopoietin (TPO) during differentiation. BMP-4 with 50 ng/mL TPO generated 232 +/- 48 CFC per 5 x 10(4) cells, similar to the serum-control, and this response could be enhanced to 292 +/- 42 CFC per 5 x 10(4) cells by early (between day 0-5), but not late (after day 5) VEGF treatment. CONCLUSION Moving to SF systems facilitates directed differentiation by eliminating confounding signals. This article describes modifications to the N2B27 media that amplify mesoderm induction and extends earlier work defining blood progenitor cell induction from ESC with BMP-4, VEGF, and TPO.
Collapse
|
39
|
Non-random subcellular distribution of variant EKLF in erythroid cells. Exp Cell Res 2008; 314:1595-604. [PMID: 18329016 DOI: 10.1016/j.yexcr.2008.01.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 01/11/2008] [Accepted: 01/29/2008] [Indexed: 11/22/2022]
Abstract
EKLF protein plays a prominent role during erythroid development as a nuclear transcription factor. Not surprisingly, exogenous EKLF quickly localizes to the nucleus. However, using two different assays we have unexpectedly found that a substantial proportion of endogenous EKLF resides in the cytoplasm at steady state in all erythroid cells examined. While EKLF localization does not appear to change during either erythroid development or terminal differentiation, we find that the protein displays subtle yet distinct biochemical and functional differences depending on which subcellular compartment it is isolated from, with PEST sequences possibly playing a role in these differences. Localization is unaffected by inhibition of CRM1 activity and the two populations are not differentiated by stability. Heterokaryon assays demonstrate that EKLF is able to shuttle out of the nucleus although its nuclear re-entry is rapid. These studies suggest there is an unexplored role for EKLF in the cytoplasm that is separate from its well-characterized nuclear function.
Collapse
|
40
|
Durand C, Robin C, Bollerot K, Baron MH, Ottersbach K, Dzierzak E. Embryonic stromal clones reveal developmental regulators of definitive hematopoietic stem cells. Proc Natl Acad Sci U S A 2007; 104:20838-43. [PMID: 18087045 PMCID: PMC2409228 DOI: 10.1073/pnas.0706923105] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Indexed: 11/18/2022] Open
Abstract
Hematopoietic stem cell (HSC) self-renewal and differentiation is regulated by cellular and molecular interactions with the surrounding microenvironment. During ontogeny, the aorta-gonad-mesonephros (AGM) region autonomously generates the first HSCs and serves as the first HSC-supportive microenvironment. Because the molecular identity of the AGM microenvironment is as yet unclear, we examined two closely related AGM stromal clones that differentially support HSCs. Expression analyses identified three putative HSC regulatory factors, beta-NGF (a neurotrophic factor), MIP-1gamma (a C-C chemokine family member) and Bmp4 (a TGF-beta family member). We show here that these three factors, when added to AGM explant cultures, enhance the in vivo repopulating ability of AGM HSCs. The effects of Bmp4 on AGM HSCs were further studied because this factor acts at the mesodermal and primitive erythropoietic stages in the mouse embryo. In this report, we show that enriched E11 AGM HSCs express Bmp receptors and can be inhibited in their activity by gremlin, a Bmp antagonist. Moreover, our results reveal a focal point of Bmp4 expression in the mesenchyme underlying HSC containing aortic clusters at E11. We suggest that Bmp4 plays a relatively late role in the regulation of HSCs as they emerge in the midgestation AGM.
Collapse
Affiliation(s)
- Charles Durand
- *Department of Cell Biology and Genetics, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands; and
| | - Catherine Robin
- *Department of Cell Biology and Genetics, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands; and
| | - Karine Bollerot
- *Department of Cell Biology and Genetics, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands; and
| | - Margaret H. Baron
- Departments of Medicine, Molecular, Cell, and Developmental Biology, Gene and Cell Medicine, and Oncological Sciences and The Black Family Stem Cell Institute, Mt. Sinai School of Medicine, New York, NY 10029
| | - Katrin Ottersbach
- *Department of Cell Biology and Genetics, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands; and
| | - Elaine Dzierzak
- *Department of Cell Biology and Genetics, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands; and
| |
Collapse
|
41
|
Bruce SJ, Gardiner BB, Burke LJ, Gongora MM, Grimmond SM, Perkins AC. Dynamic transcription programs during ES cell differentiation towards mesoderm in serum versus serum-freeBMP4 culture. BMC Genomics 2007; 8:365. [PMID: 17925037 PMCID: PMC2204012 DOI: 10.1186/1471-2164-8-365] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 10/10/2007] [Indexed: 12/30/2022] Open
Abstract
Background Expression profiling of embryonic stem (ES) cell differentiation in the presence of serum has been performed previously. It remains unclear if transcriptional activation is dependent on complex growth factor mixtures in serum or whether this process is intrinsic to ES cells once the stem cell program has been inactivated. The aims of this study were to determine the transcriptional programs associated with the stem cell state and to characterize mesoderm differentiation between serum and serum-free culture. Results ES cells were differentiated as embryoid bodies in 10% FBS or serum-free media containing BMP4 (2 ng/ml), and expression profiled using 47 K Illumina(R) Sentrix arrays. Statistical methods were employed to define gene sets characteristic of stem cell, epiblast and primitive streak programs. Although the initial differentiation profile was similar between the two culture conditions, cardiac gene expression was inhibited in serum whereas blood gene expression was enhanced. Also, expression of many members of the Kruppel-like factor (KLF) family of transcription factors changed dramatically during the first few days of differentiation. KLF2 and KLF4 co-localized with OCT4 in a sub-nuclear compartment of ES cells, dynamic changes in KLF-DNA binding activities occurred upon differentiation, and strong bio-informatic evidence for direct regulation of many stem cell genes by KLFs was found. Conclusion Down regulation of stem cell genes and activation of epiblast/primitive streak genes is similar in serum and defined media, but subsequent mesoderm differentiation is strongly influenced by the composition of the media. In addition, KLF family members are likely to be important regulators of many stem cell genes.
Collapse
Affiliation(s)
- Stephen J Bruce
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
| | | | | | | | | | | |
Collapse
|
42
|
Frontelo P, Manwani D, Galdass M, Karsunky H, Lohmann F, Gallagher PG, Bieker JJ. Novel role for EKLF in megakaryocyte lineage commitment. Blood 2007; 110:3871-80. [PMID: 17715392 PMCID: PMC2190608 DOI: 10.1182/blood-2007-03-082065] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Megakaryocytes and erythroid cells are thought to derive from a common progenitor during hematopoietic differentiation. Although a number of transcriptional regulators are important for this process, they do not explain the bipotential result. We now show by gain- and loss-of-function studies that erythroid Krüppel-like factor (EKLF), a transcription factor whose role in erythroid gene regulation is well established, plays an unexpected directive role in the megakaryocyte lineage. EKLF inhibits the formation of megakaryocytes while at the same time stimulating erythroid differentiation. Quantitative examination of expression during hematopoiesis shows that, unlike genes whose presence is required for establishment of both lineages, EKLF is uniquely down-regulated in megakaryocytes after formation of the megakaryocyte-erythroid progenitor. Expression profiling and molecular analyses support these observations and suggest that megakaryocytic inhibition is achieved, at least in part, by EKLF repression of Fli-1 message levels.
Collapse
|
43
|
Loose M, Swiers G, Patient R. Transcriptional networks regulating hematopoietic cell fate decisions. Curr Opin Hematol 2007; 14:307-14. [PMID: 17534154 DOI: 10.1097/moh.0b013e3281900eee] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE OF REVIEW We provide a summary of the temporal cascade of transcriptional networks giving rise to the hematopoietic stem cell (HSC) and controlling differentiation of the erythroid lineage from it. We focus on the mechanisms by which cell fate decisions are made and comment on recent developments and additions to the networks. RECENT FINDINGS A role for an SCL/LMO2 complex in HSC emergence, as well as in subsequent erythroid differentiation, has received support. Connections between the transcriptional networks and signaling molecules are being made but more work is needed in this area. Evidence that transcriptional cross-antagonistic switches underlie the choice between lineage pathways is increasing, and we highlight how the dynamics of earlier lineage decisions can influence later ones. Mathematical models are being built and reveal a surprising degree of power in these simple motifs to explain lineage choices. SUMMARY New links in the transcriptional networks underlying cell-fate decisions are constantly emerging, and their incorporation into the evolving networks will make mathematical modeling more precise in its predictions of cell behavior, which can be tested experimentally.
Collapse
Affiliation(s)
- Matt Loose
- Institute of Genetics, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | | | | |
Collapse
|
44
|
Ren R, Charles PC, Zhang C, Wu Y, Wang H, Patterson C. Gene expression profiles identify a role for cyclooxygenase 2-dependent prostanoid generation in BMP6-induced angiogenic responses. Blood 2007; 109:2847-53. [PMID: 17119124 PMCID: PMC1852219 DOI: 10.1182/blood-2006-08-039743] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The bone morphogenetic protein (BMP) family of proteins participates in regulation of angiogenesis in physiologic and pathologic conditions. To investigate the molecular mechanisms that contribute to BMP-dependent angiogenic signaling, we performed gene expression profiling of BMP6-treated mouse endothelial cells. We detected 77 mRNAs that were differentially regulated after BMP6 stimulation. Of these, cyclooxygenase 2 (Cox2) was among the most highly up-regulated by BMP stimulation, suggesting a role for Cox2 as a downstream regulator of BMP-induced angiogenesis. Up-regulation of Cox2 by BMP6 was detected at both mRNA and protein levels in endothelial cells, and BMP6 increased production of prostaglandins in a Cox2-dependent fashion. BMP6 up-regulated Cox2 at the transcriptional level through upstream SMAD-binding sites in the Cox2 promoter. Pharmacologic inhibition of Cox2, but not Cox1, blocked BMP6-induced endothelial cell proliferation, migration, and network assembly. BMP6-dependent microvessel outgrowth was markedly attenuated in aortic rings from Cox2-/- mice or after pharmacologic inhibition of Cox2 in aortas from wild-type mice. These results support a necessary role for Cox2 in mediating proangiogenic activities of BMP6. These data indicate that Cox2 may serve as a unifying component downstream from disparate pathways to modulate angiogenic responses in diseases in which neovascularization plays an underlying pathophysiologic role.
Collapse
Affiliation(s)
- Rongqin Ren
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill 27599-7126, USA
| | | | | | | | | | | |
Collapse
|
45
|
Zafonte BT, Liu S, Lynch-Kattman M, Torregroza I, Benvenuto L, Kennedy M, Keller G, Evans T. Smad1 expands the hemangioblast population within a limited developmental window. Blood 2006; 109:516-23. [PMID: 16990609 PMCID: PMC1785093 DOI: 10.1182/blood-2006-02-004564] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bone morphogenetic protein (BMP) signaling is an important regulator of hematovascular development. However, the progenitor population that responds to BMP signaling is undefined, and the relative role of downstream mediators including Smad1 is unclear. We find that Smad1 shows a distinctive expression profile as embryonic stem (ES) cells undergo differentiation in the embryoid body (EB) system, with peak levels in cell populations enriched for the hemangioblast. To test the functional relevance of this observation, we generated an ES cell line that allows temporal control of ectopic Smad1 expression. Continuous expression of Smad1 from day 2 of EB culture does not disturb hematopoiesis, according to colony assays. In contrast, a pulse of Smad1 expression exclusively between day 2 and day 2.25 expands the population of progenitors for primitive erythroblasts and other hematopoietic lineages. This effect correlates with increased levels of transcripts encoding markers for the hemangioblast, including Runx1, Scl, and Gata2. Indeed, the pulse of Smad1 induction also expands the blast colony-forming cell (BL-CFC) population at a level that is fully sufficient to explain subsequent increases in hematopoiesis. Our data demonstrate that Smad1 expression is sufficient to expand the number of cells that commit to hemangioblast fate.
Collapse
Affiliation(s)
- Brian T. Zafonte
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Susanna Liu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| | | | - Ingrid Torregroza
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Luke Benvenuto
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Marion Kennedy
- Department of Gene and Cell Medicine, Mt Sinai School of Medicine, New York, NY
| | - Gordon Keller
- Department of Gene and Cell Medicine, Mt Sinai School of Medicine, New York, NY
| | - Todd Evans
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
- Correspondence: Todd Evans,
Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Chanin Rm 501, 1300 Morris Park Ave, Bronx, NY 10461; e-mail:
| |
Collapse
|
46
|
Cameron CM, Hu WS, Kaufman DS. Improved development of human embryonic stem cell-derived embryoid bodies by stirred vessel cultivation. Biotechnol Bioeng 2006; 94:938-48. [PMID: 16547998 DOI: 10.1002/bit.20919] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human embryonic stem cells (hESCs) represent an important resource for novel cell-based regenerative medical therapies. hESCs are known to differentiate into mature cells of defined lineages through the formation of embryoid bodies (EBs) which are amenable to suspension culture for several weeks. However, EBs derived from hESCs in standard static cultures are typically non-homogeneous, leading to inefficient cellular development. Here, we systematically compare the formation, growth, and differentiation capabilities of hESC-derived EBs in stirred and static suspension cultures. A 15-fold expansion in total number of EB-derived cells cultured for 21 days in a stirred flask was observed, compared to a fourfold expansion in static (non-stirred) cultures. Additionally, stirred vessel mediated cultures have a more homogeneous EB morphology and size. Importantly, the EBs cultivated in spinner flasks retained comparable ability to produce hematopoietic progenitor cells as those grown in static culture. These results demonstrate the decoupling between EB cultivation method and EB-derived cells' ability to form hematopoietic progenitors, and will allow for improved production of scalable quantities of hematopoietic cells or other differentiated cell lineages from hESCs in a controlled environment.
Collapse
Affiliation(s)
- C M Cameron
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | | | | |
Collapse
|
47
|
Quadrini KJ, Bieker JJ. EKLF/KLF1 is ubiquitinated in vivo and its stability is regulated by activation domain sequences through the 26S proteasome. FEBS Lett 2006; 580:2285-93. [PMID: 16579989 DOI: 10.1016/j.febslet.2006.03.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Revised: 02/15/2006] [Accepted: 03/10/2006] [Indexed: 01/30/2023]
Abstract
Erythroid Krüppel-like factor (EKLF/KLF1) is an erythroid specific, C(2)H(2) zinc finger transcription factor that is essential for the proper chromatin structure and expression of the adult beta-globin gene. Herein, we determine that 26S proteasome inhibitors lead to an accumulation of EKLF protein in murine erythroleukemia (MEL) cells. In addition, EKLF half-life in both MEL cells (<3h) and fetal liver cells (between 6 and 9h) is stabilized in the presence of these inhibitors. EKLF is ubiquitinated in vivo, however its modification does not rely on a particular internal lysine. Finally, EKLF contains two PEST sequences within its N-terminus that have no effect on the ability of EKLF to be ubiquitinated but contribute to its destabilization.
Collapse
Affiliation(s)
- Karen J Quadrini
- The Brookdale Department of Molecular, Cell and Developmental Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
48
|
Hassel S, Yakymovych M, Hellman U, Rönnstrand L, Knaus P, Souchelnytskyi S. Interaction and functional cooperation between the serine/threonine kinase bone morphogenetic protein type II receptor with the tyrosine kinase stem cell factor receptor. J Cell Physiol 2006; 206:457-67. [PMID: 16155937 DOI: 10.1002/jcp.20480] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Transmembrane receptors with intrinsic serine/threonine or tyrosine kinase domains regulate vital functions of cells in multicellular eukaryotes, e.g., differentiation, apoptosis, and proliferation. Here, we show that bone morphogenetic protein type II receptor (BMPR-II) which has a serine/threonine kinase domain, and stem cell factor receptor (c-kit) which contains a tyrosine kinase domain form a complex in vitro and in vivo; the interaction is induced upon treatment of cells with BMP2 and SCF. Stem cell factor (SCF) modulated BMP2-dependent activation of Smad1/5/8 and phosphorylation of Erk kinase. SCF also enhanced BMP2-dependent differentiation of C2C12 cells. We found that BMPR-II was phosphorylated at Ser757 upon co-expression with and activation of c-kit. BMPR-II phosphorylation required intact kinase activity of BMPR-II. Abrogation of the c-kit/SCF-dependent phosphorylation of BMPR-II at the Ser757 interfered with the cooperative effect of BMP2 and SCF. Our data suggest that the complex formation between c-kit and BMPR-II leads to phosphorylation of BMPR-II at Ser757, which modulates BMPR-II-dependent signaling.
Collapse
Affiliation(s)
- Sylke Hassel
- Ludwig Institute for Cancer Research, Biomedical Centre, Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
49
|
Nishimura Y, Hamazaki TS, Komazaki S, Kamimura S, Okochi H, Asashima M. Ciliated cells differentiated from mouse embryonic stem cells. Stem Cells 2006; 24:1381-8. [PMID: 16410384 DOI: 10.1634/stemcells.2005-0464] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the present study, we demonstrated that the mouse embryonic stem cells were differentiated into ciliated epithelial cells, with characteristics of normal ciliated cells. These cells expressed ciliary marker proteins, such as beta-tubulin IV and hepatocyte nuclear factor-3/forkhead homolog 4 (HFH-4), and processed microtubules were arranged in the 9 + 2 structure, which is the same specific alignment observed in normal ciliary microtubules. The cilia of these cells were beating at a frequency of 17-20 Hz. The differentiated embryoid bodies (EBs) containing these ciliated cells expressed respiratory marker genes such as thyroid transcription factor-1 and surfactant protein-C. For the induction of ciliated cells, culture of EBs in serum-free medium during the initial 2 days of the attachment was indispensable. When EBs were treated with bone morphogenetic proteins, the expression of HFH-4 was decreased, and the ciliated cells were scarcely differentiated. Previous methods for inducing ciliated cells in vitro from embryonic or adult tissues involved an air-liquid interface. The system used in this study more closely mimics the normal development of ciliated cells; thus, an added advantage of the system is as a tool for studying the differentiation mechanism of normal ciliated epithelial cells.
Collapse
Affiliation(s)
- Yusuke Nishimura
- Department of Biological Science, The University of Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
50
|
He Z, Li JJ, Zhen CH, Feng LY, Ding XY. Effect of leukemia inhibitory factor on embryonic stem cell differentiation: implications for supporting neuronal differentiation. Acta Pharmacol Sin 2006; 27:80-90. [PMID: 16364214 DOI: 10.1111/j.1745-7254.2006.00254.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
AIM Leukemia inhibitory factor (LIF), a pleiotropic cytokine, has been used extensively in the maintenance of mouse embryonic stem cell pluripotency. In this current work, we examined the effect of the LIF signaling pathway in embryonic stem (ES) cell differentiation to a neural fate. METHODS In the presence of LIF (1000 U/mL), the production of neuronal cells derived from embryoid bodies (EB) was tested under various culture conditions. Inhibition of the LIF pathway was examined with specific inhibitors. The effects of cell apoptosis and proliferation on neural differentiation were examined. ES cell differentiation into three-germ layers was compared. RESULTS Under various culture conditions, neuronal differentiation was increased in the presence of LIF. Blocking the LIF-activated STAT3 signaling pathway with specific inhibitors abolished the neuronal differentiation of ES cells, whereas inhibition of the LIF-activated MEK signaling pathway impaired the differentiation of ES cells toward a glial fate. LIF suppressed cell apoptosis and promoted cell proliferation during ES cell differentiation. LIF inhibited the differentiation of ES cells to both mesoderm and extraembryonic endoderm fates, but enhanced the determination of neural progenitors. CONCLUSION These results suggest that LIF plays a positive role during the differentiation of ES cells into neuronal cells.
Collapse
Affiliation(s)
- Zhao He
- Laboratory of Molecular and Cell Biology and Laboratory of Stem Cell Biology, Institute of Biochemistry and Cell Biology; Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China
| | | | | | | | | |
Collapse
|