251
|
Hematopoietic stem cell and mesenchymal stem cell population size in bone marrow samples depends on patient's age and harvesting technique. Cytotechnology 2018; 70:1575-1583. [PMID: 30229373 DOI: 10.1007/s10616-018-0250-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are heterogeneous population of cells with great potential for regenerative medicine. MSCs are relatively easy to expand in a cell culture, however determination of their concentration in harvested tissue is more complex and is not implemented as routine procedure. To identify MSCs collected from bone marrow we have used two combinations of cell markers (CD45-/CD73+/CD90+/CD105+ and CD45-/CD271+) and fibroblast colony-forming unit (CFU-F) assay. Further, in donors of various ages, mesenchymal stem cell concentration was compared with the result of CFU-F assay and with hematopoietic stem cell concentration, determined by a standardized flow cytometric assay. A positive correlation of MSC populations to the CFU-F numbers is observed, the population of the CD45-/CD271+ cells correlates better with CFU-F numbers than the population of the CD45-/CD73+/CD90+/CD105+ cells. The relationship between the hematopoietic CD45dim/CD34+ cell concentration and mesenchymal CFU-Fs or CD45-/CD271+ cells shows a positive linear regression. An age-related quantitative reduction of hematopoietic CD45dim/CD34+, mesenchymal CD45-/CD73+/CD90+/CD105+ and CD45-/CD271+ stem cells, and CFU-F numbers were noted. Additionally, statistically significant higher CFU-F numbers were observed when bone marrow samples were harvested from three different sites from the anterior iliac crest instead of harvesting the same sample amount only from one site.
Collapse
|
252
|
Rijal G, Li W. Native-mimicking in vitro microenvironment: an elusive and seductive future for tumor modeling and tissue engineering. J Biol Eng 2018; 12:20. [PMID: 30220913 PMCID: PMC6136168 DOI: 10.1186/s13036-018-0114-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Human connective tissues are complex physiological microenvironments favorable for optimal survival, function, growth, proliferation, differentiation, migration, and death of tissue cells. Mimicking native tissue microenvironment using various three-dimensional (3D) tissue culture systems in vitro has been explored for decades, with great advances being achieved recently at material, design and application levels. These achievements are based on improved understandings about the functionalities of various tissue cells, the biocompatibility and biodegradability of scaffolding materials, the biologically functional factors within native tissues, and the pathophysiological conditions of native tissue microenvironments. Here we discuss these continuously evolving physical aspects of tissue microenvironment important for human disease modeling, with a focus on tumors, as well as for tissue repair and regeneration. The combined information about human tissue spaces reflects the necessities of considerations when configuring spatial microenvironments in vitro with native fidelity to culture cells and regenerate tissues that are beyond the formats of 2D and 3D cultures. It is important to associate tissue-specific cells with specific tissues and microenvironments therein for a better understanding of human biology and disease conditions and for the development of novel approaches to treat human diseases.
Collapse
Affiliation(s)
- Girdhari Rijal
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99210 USA
| | - Weimin Li
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99210 USA
| |
Collapse
|
253
|
Shepherd MS, Li J, Wilson NK, Oedekoven CA, Li J, Belmonte M, Fink J, Prick JCM, Pask DC, Hamilton TL, Loeffler D, Rao A, Schröder T, Göttgens B, Green AR, Kent DG. Single-cell approaches identify the molecular network driving malignant hematopoietic stem cell self-renewal. Blood 2018; 132:791-803. [PMID: 29991556 PMCID: PMC6107881 DOI: 10.1182/blood-2017-12-821066] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 07/03/2018] [Indexed: 12/24/2022] Open
Abstract
Recent advances in single-cell technologies have permitted the investigation of heterogeneous cell populations at previously unattainable resolution. Here we apply such approaches to resolve the molecular mechanisms driving disease in mouse hematopoietic stem cells (HSCs), using JAK2V617F mutant myeloproliferative neoplasms (MPNs) as a model. Single-cell gene expression and functional assays identified a subset of JAK2V617F mutant HSCs that display defective self-renewal. This defect is rescued at the single HSC level by crossing JAK2V617F mice with mice lacking TET2, the most commonly comutated gene in patients with MPN. Single-cell gene expression profiling of JAK2V617F-mutant HSCs revealed a loss of specific regulator genes, some of which were restored to normal levels in single TET2/JAK2 mutant HSCs. Of these, Bmi1 and, to a lesser extent, Pbx1 and Meis1 overexpression in JAK2-mutant HSCs could drive a disease phenotype and retain durable stem cell self-renewal in functional assays. Together, these single-cell approaches refine the molecules involved in clonal expansion of MPNs and have broad implications for deconstructing the molecular network of normal and malignant stem cells.
Collapse
Affiliation(s)
- Mairi S Shepherd
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Juan Li
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Nicola K Wilson
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Caroline A Oedekoven
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Jiangbing Li
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Miriam Belmonte
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Juergen Fink
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Janine C M Prick
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Dean C Pask
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Tina L Hamilton
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Dirk Loeffler
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland; and
| | - Anjana Rao
- La Jolla Institute and Department of Pharmacology, University of California, San Diego, La Jolla, CA
| | - Timm Schröder
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland; and
| | - Berthold Göttgens
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| | - Anthony R Green
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
- Department of Haematology, Addenbrooke's Hospital, Hills Road, Cambridge, United Kingdom
| | - David G Kent
- Wellcome MRC Cambridge Stem Cell Institute and
- Department of Haematology, University of Cambridge, United Kingdom
| |
Collapse
|
254
|
Mesuraca M, Amodio N, Chiarella E, Scicchitano S, Aloisio A, Codispoti B, Lucchino V, Montalcini Y, Bond HM, Morrone G. Turning Stem Cells Bad: Generation of Clinically Relevant Models of Human Acute Myeloid Leukemia through Gene Delivery- or Genome Editing-Based Approaches. Molecules 2018; 23:E2060. [PMID: 30126100 PMCID: PMC6222541 DOI: 10.3390/molecules23082060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/09/2018] [Accepted: 08/14/2018] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML), the most common acute leukemia in the adult, is believed to arise as a consequence of multiple molecular events that confer on primitive hematopoietic progenitors unlimited self-renewal potential and cause defective differentiation. A number of genetic aberrations, among which a variety of gene fusions, have been implicated in the development of a transformed phenotype through the generation of dysfunctional molecules that disrupt key regulatory mechanisms controlling survival, proliferation, and differentiation in normal stem and progenitor cells. Such genetic aberrations can be recreated experimentally to a large extent, to render normal hematopoietic stem cells "bad", analogous to the leukemic stem cells. Here, we wish to provide a brief outline of the complementary experimental approaches, largely based on gene delivery and more recently on gene editing, employed over the last two decades to gain insights into the molecular mechanisms underlying AML development and progression and on the prospects that their applications offer for the discovery and validation of innovative therapies.
Collapse
Affiliation(s)
- Maria Mesuraca
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Nicola Amodio
- Laboratory of Medical Oncology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Emanuela Chiarella
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Stefania Scicchitano
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Annamaria Aloisio
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Bruna Codispoti
- Tecnologica Research Institute-Marrelli Hospital, 88900 Crotone, Italy.
| | - Valeria Lucchino
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.
| | - Ylenia Montalcini
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Heather M Bond
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Giovanni Morrone
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| |
Collapse
|
255
|
Koenis DS, Medzikovic L, Vos M, Beldman TJ, van Loenen PB, van Tiel CM, Hamers AAJ, Otermin Rubio I, de Waard V, de Vries CJM. Nur77 variants solely comprising the amino-terminal domain activate hypoxia-inducible factor-1α and affect bone marrow homeostasis in mice and humans. J Biol Chem 2018; 293:15070-15083. [PMID: 30111591 DOI: 10.1074/jbc.ra118.002720] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 07/30/2018] [Indexed: 01/03/2023] Open
Abstract
Gene targeting via homologous recombination can occasionally result in incomplete disruption of the targeted gene. Here, we show that a widely used Nur77-deficient transgenic mouse model expresses a truncated protein encoding for part of the N-terminal domain of nuclear receptor Nur77. This truncated Nur77 protein is absent in a newly developed Nur77-deficient mouse strain generated using Cre-Lox recombination. Comparison of these two mouse strains using immunohistochemistry, flow cytometry, and colony-forming assays shows that homologous recombination-derived Nur77-deficient mice, but not WT or Cre-Lox-derived Nur77-deficient mice, suffer from liver immune cell infiltrates, loss of splenic architecture, and increased numbers of bone marrow hematopoietic stem cells and splenic colony-forming cells with age. Mechanistically, we demonstrate that the truncated Nur77 N-terminal domain protein maintains the stability and activity of hypoxia-inducible factor (HIF)-1, a transcription factor known to regulate bone marrow homeostasis. Additionally, a previously discovered, but uncharacterized, human Nur77 transcript variant that encodes solely for its N-terminal domain, designated TR3β, can also stabilize and activate HIF-1α. Meta-analysis of publicly available microarray data sets shows that TR3β is highly expressed in human bone marrow cells and acute myeloid leukemia samples. In conclusion, our study provides evidence that a transgenic mouse model commonly used to study the biological function of Nur77 has several major drawbacks, while simultaneously identifying the importance of nongenomic Nur77 activity in the regulation of bone marrow homeostasis.
Collapse
Affiliation(s)
- Duco S Koenis
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Lejla Medzikovic
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Mariska Vos
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Thijs J Beldman
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Pieter B van Loenen
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Claudia M van Tiel
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Anouk A J Hamers
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Iker Otermin Rubio
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Vivian de Waard
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Carlie J M de Vries
- From the Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
256
|
Ramond C, Beydag-Tasöz BS, Azad A, van de Bunt M, Petersen MBK, Beer NL, Glaser N, Berthault C, Gloyn AL, Hansson M, McCarthy MI, Honoré C, Grapin-Botton A, Scharfmann R. Understanding human fetal pancreas development using subpopulation sorting, RNA sequencing and single-cell profiling. Development 2018; 145:dev.165480. [PMID: 30042179 PMCID: PMC6124547 DOI: 10.1242/dev.165480] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/06/2018] [Indexed: 12/30/2022]
Abstract
To decipher the populations of cells present in the human fetal pancreas and their lineage relationships, we developed strategies to isolate pancreatic progenitors, endocrine progenitors and endocrine cells. Transcriptome analysis of the individual populations revealed a large degree of conservation among vertebrates in the drivers of gene expression changes that occur at different steps of differentiation, although notably, sometimes, different members of the same gene family are expressed. The transcriptome analysis establishes a resource to identify novel genes and pathways involved in human pancreas development. Single-cell profiling further captured intermediate stages of differentiation and enabled us to decipher the sequence of transcriptional events occurring during human endocrine differentiation. Furthermore, we evaluate how well individual pancreatic cells derived in vitro from human pluripotent stem cells mirror the natural process occurring in human fetuses. This comparison uncovers a few differences at the progenitor steps, a convergence at the steps of endocrine induction, and the current inability to fully resolve endocrine cell subtypes in vitro.
Collapse
Affiliation(s)
- Cyrille Ramond
- Department of Endocrinology, Metabolism and Diabetes, Inserm U1016, Cochin Institute, Paris 75014, France,CNRS UMR 8104, Paris 75014, France,University of Paris Descartes, Sorbonne Paris Cité, Paris 75006, France
| | - Belin Selcen Beydag-Tasöz
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Ajuna Azad
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Martijn van de Bunt
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK,Global Research Informatics, Novo Nordisk A/S, Novo Nordisk Park, Måløv 2760, Denmark
| | - Maja Borup Kjær Petersen
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen 2200, Denmark,Department of Stem Cell Biology, Novo Nordisk A/S, Novo Nordisk Park, Måløv 2760, Denmark
| | - Nicola L. Beer
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| | - Nicolas Glaser
- Department of Endocrinology, Metabolism and Diabetes, Inserm U1016, Cochin Institute, Paris 75014, France,CNRS UMR 8104, Paris 75014, France,University of Paris Descartes, Sorbonne Paris Cité, Paris 75006, France
| | - Claire Berthault
- Department of Endocrinology, Metabolism and Diabetes, Inserm U1016, Cochin Institute, Paris 75014, France,CNRS UMR 8104, Paris 75014, France,University of Paris Descartes, Sorbonne Paris Cité, Paris 75006, France
| | - Anna L. Gloyn
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| | - Mattias Hansson
- Stem Cell Research, Novo Nordisk A/S, Novo Nordisk Park, Måløv 2760, Denmark
| | - Mark I. McCarthy
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| | - Christian Honoré
- Department of Stem Cell Biology, Novo Nordisk A/S, Novo Nordisk Park, Måløv 2760, Denmark
| | - Anne Grapin-Botton
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen 2200, Denmark,Authors for correspondence (; )
| | - Raphael Scharfmann
- Department of Endocrinology, Metabolism and Diabetes, Inserm U1016, Cochin Institute, Paris 75014, France,CNRS UMR 8104, Paris 75014, France,University of Paris Descartes, Sorbonne Paris Cité, Paris 75006, France,Authors for correspondence (; )
| |
Collapse
|
257
|
Hui T, Cao Q, Wegrzyn-Woltosz J, O'Neill K, Hammond CA, Knapp DJHF, Laks E, Moksa M, Aparicio S, Eaves CJ, Karsan A, Hirst M. High-Resolution Single-Cell DNA Methylation Measurements Reveal Epigenetically Distinct Hematopoietic Stem Cell Subpopulations. Stem Cell Reports 2018; 11:578-592. [PMID: 30078558 PMCID: PMC6093082 DOI: 10.1016/j.stemcr.2018.07.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/08/2018] [Accepted: 07/09/2018] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence of functional and transcriptional heterogeneity in phenotypically similar cells examined individually has prompted interest in obtaining parallel methylome data. We describe the development and application of such a protocol to index-sorted murine and human hematopoietic cells that are highly enriched in their content of functionally defined stem cells. Utilizing an optimized single-cell bisulfite sequencing protocol, we obtained quantitative DNA methylation measurements of up to 5.7 million CpGs in single hematopoietic cells. In parallel, we developed an analytical strategy (PDclust) to define single-cell DNA methylation states through pairwise comparisons of single-CpG methylation measurements. PDclust revealed that a single-cell epigenetic state can be described by a small (<1%) stochastically sampled fraction of CpGs and that these states are reflective of cell identity and state. Using relationships revealed by PDclust, we derive near complete methylomes for epigenetically distinct subpopulations of hematopoietic cells enriched for functional stem cell content.
Collapse
Affiliation(s)
- Tony Hui
- Department of Microbiology and Immunology and Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - Qi Cao
- Department of Microbiology and Immunology and Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Joanna Wegrzyn-Woltosz
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Kieran O'Neill
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Terry Fox Laboratory, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - Colin A Hammond
- Terry Fox Laboratory, BC Cancer, Vancouver, BC V5Z 1L3, Canada; Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - David J H F Knapp
- Terry Fox Laboratory, BC Cancer, Vancouver, BC V5Z 1L3, Canada; Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Emma Laks
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - Michelle Moksa
- Department of Microbiology and Immunology and Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Samuel Aparicio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - Connie J Eaves
- Terry Fox Laboratory, BC Cancer, Vancouver, BC V5Z 1L3, Canada; Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Aly Karsan
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Terry Fox Laboratory, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - Martin Hirst
- Department of Microbiology and Immunology and Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada.
| |
Collapse
|
258
|
A metabolic interplay coordinated by HLX regulates myeloid differentiation and AML through partly overlapping pathways. Nat Commun 2018; 9:3090. [PMID: 30082823 PMCID: PMC6078963 DOI: 10.1038/s41467-018-05311-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 06/29/2018] [Indexed: 02/07/2023] Open
Abstract
The H2.0-like homeobox transcription factor (HLX) regulates hematopoietic differentiation and is overexpressed in Acute Myeloid Leukemia (AML), but the mechanisms underlying these functions remain unclear. We demonstrate here that HLX overexpression leads to a myeloid differentiation block both in zebrafish and human hematopoietic stem and progenitor cells (HSPCs). We show that HLX overexpression leads to downregulation of genes encoding electron transport chain (ETC) components and upregulation of PPARδ gene expression in zebrafish and human HSPCs. HLX overexpression also results in AMPK activation. Pharmacological modulation of PPARδ signaling relieves the HLX-induced myeloid differentiation block and rescues HSPC loss upon HLX knockdown but it has no effect on AML cell lines. In contrast, AMPK inhibition results in reduced viability of AML cell lines, but minimally affects myeloid progenitors. This newly described role of HLX in regulating the metabolic state of hematopoietic cells may have important therapeutic implications. HLX transcription factor regulates haematopoietic stem and progenitor cell (HSPC) differentiation and is overexpressed in acute myeloid leukemia. Here the authors show that HLX overexpression leads to myeloid differentiation block in zebrafish and human HSPCs by direct regulation of metabolic pathways.
Collapse
|
259
|
Salas LA, Wiencke JK, Koestler DC, Zhang Z, Christensen BC, Kelsey KT. Tracing human stem cell lineage during development using DNA methylation. Genome Res 2018; 28:1285-1295. [PMID: 30072366 PMCID: PMC6120629 DOI: 10.1101/gr.233213.117] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 07/27/2018] [Indexed: 12/22/2022]
Abstract
Stem cell maturation is a fundamental, yet poorly understood aspect of human development. We devised a DNA methylation signature deeply reminiscent of embryonic stem cells (a fetal cell origin signature, FCO) to interrogate the evolving character of multiple human tissues. The cell fraction displaying this FCO signature was highly dependent upon developmental stage (fetal versus adult), and in leukocytes, it described a dynamic transition during the first 5 yr of life. Significant individual variation in the FCO signature of leukocytes was evident at birth, in childhood, and throughout adult life. The genes characterizing the signature included transcription factors and proteins intimately involved in embryonic development. We defined and applied a DNA methylation signature common among human fetal hematopoietic progenitor cells and have shown that this signature traces the lineage of cells and informs the study of stem cell heterogeneity in humans under homeostatic conditions.
Collapse
Affiliation(s)
- Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03756, USA
| | - John K Wiencke
- Department of Neurological Surgery, Institute for Human Genetics, University of California San Francisco, San Francisco, California 94158, USA
| | - Devin C Koestler
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Ze Zhang
- Department of Epidemiology, Brown University, Providence, Rhode Island 02912, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03756, USA.,Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03756, USA.,Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03756, USA
| | - Karl T Kelsey
- Department of Epidemiology, Brown University, Providence, Rhode Island 02912, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912, USA
| |
Collapse
|
260
|
Abstract
Purpose of Review Clonal hematopoiesis of indeterminate potential (CHIP) is a common, age-associated condition characterized by the acquisition of somatic mutations. This concise review explores our current understanding of the mechanisms that influence the development of clonality with aging and its potential malignant and non-malignant clinical implications. Recent Findings Aging of the hematopoietic system results in phenotypic changes that favor clonal dominance. Cell-extrinsic factors provide additional selective pressures that further shape clonal architecture. Even so, small clones with candidate driver mutations appear to be ubiquitous with age and largely benign in the absence of strong selective pressures. Benign clonal expansion may compensate for the loss of regenerative HSC capacity as we age. Summary CHIP is a marker of aging that reflects the biologic interplay between HSC aging and cell-extrinsic factors. The clinical significance of CHIP is highly variable and dependent on clinical context. Distinguishing the causal relationships and confounding factors that regulate clonal behavior will be essential to define the mechanistic role of CHIP in aging and potentially mitigate its clinical consequences.
Collapse
Affiliation(s)
- Soo J Park
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Rafael Bejar
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|
261
|
Wilkinson AC, Yamazaki S. The hematopoietic stem cell diet. Int J Hematol 2018; 107:634-641. [PMID: 29605874 DOI: 10.1007/s12185-018-2451-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 03/27/2018] [Indexed: 02/07/2023]
Abstract
Hematopoietic stem cells (HSCs) are responsible for sustaining life-long blood formation or hematopoiesis and are also used clinically in a form of bone marrow transplantation, a curative cellular therapy for a range of hematological diseases. HSCs are maintained throughout adult life by a complex biological niche or microenvironment, which is thought to be composed of a range of cellular, molecular, and metabolic components. The metabolic components of the HSC niche have become of increasing interest over the past few years. It is now well-recognized that metabolic activity is intimately linked to HSC function, and dysregulation of these metabolic pathways result in hematological pathologies such as leukemia. Here, we review the recent progress in this field including our current understanding of the "dietary" requirements of HSCs and how nutrition influences HSC activity. These recent findings have suggested promising new metabolic approaches to improve clinical HSC transplantation and leukemia therapies.
Collapse
Affiliation(s)
- Adam C Wilkinson
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Satoshi Yamazaki
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan.
| |
Collapse
|
262
|
Extracellular Vesicles: A New Prospective in Crosstalk between Microenvironment and Stem Cells in Hematological Malignancies. Stem Cells Int 2018; 2018:9863194. [PMID: 29977309 PMCID: PMC5994264 DOI: 10.1155/2018/9863194] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/31/2018] [Indexed: 02/06/2023] Open
Abstract
The bone marrow (BM) microenvironment in hematological malignancies (HMs) comprises heterogeneous populations of neoplastic and nonneoplastic cells. Cancer stem cells (CSCs), neoplastic cells, hematopoietic stem cells (HSCs), and mesenchymal stromal/stem cells (MSCs) are all components of this microenvironment. CSCs are the HM initiators and are associated with neoplastic growth and drug resistance, while HSCs are able to reconstitute the entire hematopoietic system; finally, MSCs actively support hematopoiesis. In some HMs, CSCs and neoplastic cells compromise the normal development of HSCs and perturb BM-MSCs. In response, "reprogrammed" MSCs generate a favorable environment to support neoplastic cells. Extracellular vesicles (EVs) are an important cell-to-cell communication type in physiological and pathological conditions. In particular, in HMs, EV secretion participates to unidirectional and bidirectional interactions between neoplastic cells and BM cells. The transfer of EV molecular cargo triggers different responses in target cells; in particular, malignant EVs modify the BM environment in favor of neoplastic cells at the expense of normal HSCs, by interfering with antineoplastic immunity and participating in resistance to treatment. Here, we review the role of EVs in BM cell communication in physiological conditions and in HMs, focusing on the effects of BM niche EVs on HSCs and MSCs.
Collapse
|
263
|
Single-cell analysis identifies a CD33 + subset of human cord blood cells with high regenerative potential. Nat Cell Biol 2018; 20:710-720. [PMID: 29802403 DOI: 10.1038/s41556-018-0104-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 04/13/2018] [Indexed: 12/15/2022]
Abstract
Elucidation of the identity and diversity of mechanisms that sustain long-term human blood cell production remains an important challenge. Previous studies indicate that, in adult mice, this property is vested in cells identified uniquely by their ability to clonally regenerate detectable, albeit highly variable levels and types, of mature blood cells in serially transplanted recipients. From a multi-parameter analysis of the molecular features of very primitive human cord blood cells that display long-term cell outputs in vitro and in immunodeficient mice, we identified a prospectively separable CD33+CD34+CD38-CD45RA-CD90+CD49f+ phenotype with serially transplantable, but diverse, cell output profiles. Single-cell measurements of the mitogenic response, and the transcriptional, DNA methylation and 40-protein content of this and closely related phenotypes revealed subtle but consistent differences both within and between each subset. These results suggest that multiple regulatory mechanisms combine to maintain different cell output activities of human blood cell precursors with high regenerative potential.
Collapse
|
264
|
Dessels C, Alessandrini M, Pepper MS. Factors Influencing the Umbilical Cord Blood Stem Cell Industry: An Evolving Treatment Landscape. Stem Cells Transl Med 2018; 7:643-650. [PMID: 29777574 PMCID: PMC6127225 DOI: 10.1002/sctm.17-0244] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/03/2018] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is common practice today for life threatening malignant and non-malignant diseases of the blood and immune systems. Umbilical cord blood (UCB) is rich in hematopoietic stem cells (HSCs) and is an attractive alternative to harvesting HSCs from bone marrow or when mobilized into peripheral blood. One of the most appealing attributes of UCB is that it can be banked for future use and hence provides an off-the-shelf solution for patients in urgent need of a transplantation. This has led to the establishment of publicly funded and private UCB banks, as seen by the rapid growth of the UCB industry in the early part of this century. However, from about 2010, the release of UCB units for treatment purposes plateaued and started to decrease year-on-year from 2013 to 2016. Our interest has been to investigate the factors contributing to these changes. Key drivers influencing the UCB industry include the emergence of haploidentical HSCT and the increasing use of UCB units for regenerative medicine purposes. Further influencing this dynamic is the high cost associated with UCB transplantation, the economic impact of sustaining public bank operations and an active private UCB banking sector. We foresee that these factors will continue in a tug-of-war fashion to shape and finally determine the fate of the UCB industry. Stem Cells Translational Medicine 2018 Stem Cells Translational Medicine 2018;7:643-650.
Collapse
Affiliation(s)
- Carla Dessels
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Marco Alessandrini
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Michael Sean Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
265
|
Grinenko T, Eugster A, Thielecke L, Ramasz B, Krüger A, Dietz S, Glauche I, Gerbaulet A, von Bonin M, Basak O, Clevers H, Chavakis T, Wielockx B. Hematopoietic stem cells can differentiate into restricted myeloid progenitors before cell division in mice. Nat Commun 2018; 9:1898. [PMID: 29765026 PMCID: PMC5954009 DOI: 10.1038/s41467-018-04188-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 04/10/2018] [Indexed: 02/06/2023] Open
Abstract
Hematopoietic stem cells (HSCs) continuously replenish all blood cell types through a series of differentiation steps and repeated cell divisions that involve the generation of lineage-committed progenitors. However, whether cell division in HSCs precedes differentiation is unclear. To this end, we used an HSC cell-tracing approach and Ki67RFP knock-in mice, in a non-conditioned transplantation model, to assess divisional history, cell cycle progression, and differentiation of adult HSCs. Our results reveal that HSCs are able to differentiate into restricted progenitors, especially common myeloid, megakaryocyte-erythroid and pre-megakaryocyte progenitors, without undergoing cell division and even before entering the S phase of the cell cycle. Additionally, the phenotype of the undivided but differentiated progenitors correlated with the expression of lineage-specific genes and loss of multipotency. Thus HSC fate decisions can be uncoupled from physical cell division. These results facilitate a better understanding of the mechanisms that control fate decisions in hematopoietic cells. Dependence of hematopoietic stem cell (HSC) fate on the phase of the cell cycle has not been demonstrated in vivo. Here, the authors find that HSCs can differentiate into a downstream progenitor without physical division, even before progressing into the S phase of the cell cycle.
Collapse
Affiliation(s)
- Tatyana Grinenko
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
| | - Anne Eugster
- DFG Research Centre and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Lars Thielecke
- Institute for Medical Informatics and Biometry (IMB), Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Beáta Ramasz
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Anja Krüger
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Sevina Dietz
- DFG Research Centre and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Ingmar Glauche
- Institute for Medical Informatics and Biometry (IMB), Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Alexander Gerbaulet
- Institute for Immunology, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Malte von Bonin
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden, Fetscherstraße 74, 01307, Dresden, Germany.,German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Onur Basak
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, Netherlands.,Cancer Genomics Netherlands, UMC Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, Netherlands.,Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht and Utrecht University, 3584 CG, Utrecht, Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, Netherlands.,Cancer Genomics Netherlands, UMC Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, Netherlands.,Princess Máxima Centre, Lundlaan 6, 3584, EA Utrecht, Netherlands
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.,DFG Research Centre and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Ben Wielockx
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany. .,DFG Research Centre and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany.
| |
Collapse
|
266
|
Berrun A, Harris E, Stachura DL. Isthmin 1 (ism1) is required for normal hematopoiesis in developing zebrafish. PLoS One 2018; 13:e0196872. [PMID: 29758043 PMCID: PMC5951578 DOI: 10.1371/journal.pone.0196872] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023] Open
Abstract
Hematopoiesis is an essential and highly regulated biological process that begins with hematopoietic stem cells (HSCs). In healthy organisms, HSCs are responsible for generating a multitude of mature blood cells every day, yet the molecular pathways that instruct HSCs to self-renew and differentiate into post-mitotic blood cells are not fully known. To understand these molecular pathways, we investigated novel genes expressed in hematopoietic-supportive cell lines from the zebrafish (Danio rerio), a model system increasingly utilized to uncover molecular pathways important in the development of other vertebrate species. We performed RNA sequencing of the transcriptome of three stromal cell lines derived from different stages of embryonic and adult zebrafish and identified hundreds of highly expressed transcripts. For our studies, we focused on isthmin 1 (ism1) due to its shared synteny with its human gene ortholog and because it is a secreted protein. To characterize ism1, we performed loss-of-function experiments to identify if mature blood cell production was disrupted. Myeloid and erythroid lineages were visualized and scored with transgenic zebrafish expressing lineage-specific markers. ism1 knockdown led to reduced numbers of neutrophils, macrophages, and erythrocytes. Analysis of clonal methylcellulose assays from ism1 morphants also showed a reduction in total hematopoietic stem and progenitor cells (HSPCs). Overall, we demonstrate that ism1 is required for normal generation of HSPCs and their downstream progeny during zebrafish hematopoiesis. Further investigation into ism1 and its importance in hematopoiesis may elucidate evolutionarily conserved processes in blood formation that can be further investigated for potential clinical utility.
Collapse
Affiliation(s)
- Arturo Berrun
- Department of Biological Sciences, California State University Chico, Chico, CA, United States of America
| | - Elena Harris
- Department of Computer Sciences, California State University Chico, Chico, CA, United States of America
| | - David L Stachura
- Department of Biological Sciences, California State University Chico, Chico, CA, United States of America
| |
Collapse
|
267
|
Nowek K, Wiemer EA, Jongen-Lavrencic M. The versatile nature of miR-9/9 * in human cancer. Oncotarget 2018; 9:20838-20854. [PMID: 29755694 PMCID: PMC5945517 DOI: 10.18632/oncotarget.24889] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 02/26/2018] [Indexed: 12/22/2022] Open
Abstract
miR-9 and miR-9* (miR-9/9*) were first shown to be expressed in the nervous system and to function as versatile regulators of neurogenesis. The variable expression levels of miR-9/9* in human cancer prompted researchers to investigate whether these small RNAs may also have an important role in the deregulation of physiological and biochemical networks in human disease. In this review, we present a comprehensive overview of the involvement of miR-9/9* in various human malignancies focusing on their opposing roles in supporting or suppressing tumor development and metastasis. Importantly, it is shown that the capacity of miR-9/9* to impact tumor formation is independent from their influence on the metastatic potential of tumor cells. Moreover, data suggest that miR-9/9* may increase malignancy of one cancer cell population at the expense of another. The functional versatility of miR-9/9* emphasizes the complexity of studying miRNA function and the importance to perform functional studies of both miRNA strands in a relevant cellular context. The possible application of miR-9/9* as targets for miRNA-based therapies is discussed, emphasizing the need to obtain a better understanding of the functional properties of these miRNAs and to develop safe delivery methods to target specific cell populations.
Collapse
Affiliation(s)
- Katarzyna Nowek
- Department of Hematology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Erik A.C. Wiemer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mojca Jongen-Lavrencic
- Department of Hematology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
268
|
Systems for localized release to mimic paracrine cell communication in vitro. J Control Release 2018; 278:24-36. [PMID: 29601931 DOI: 10.1016/j.jconrel.2018.03.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/27/2022]
Abstract
Paracrine cell communication plays a pivotal role for signal exchange between proximal cells in vivo. However, this localized, gradient type release of mediators at very low concentrations (pg/ml), relevant during physiological and pathological processes, is rarely reflected within in vitro approaches. This review gives an overview on state-of-the-art approaches, which transfer the paracrine cell-to-cell communication into in vitro cell culture model setups. The traditional methods like trans-well assays and more advanced microfluidic approaches are included. The review focusses on systems for localized release, mostly based on microparticles, which tightly mimic the paracrine interaction between single cells in 3D microenvironments. Approaches based on single microparticles, with the main focus on affinity-controlled storage and release of cytokines, are reviewed and their importance for understanding paracrine communication is highlighted. Various methods to study the cytokine release and their advantages and disadvantages are discussed. Basic principles of the release characteristics, like diffusion mechanisms, are quantitatively described, including the formation of resulting gradients around the local sources. In vitro cell experiments using such localized microparticle release systems in approaches to increase understanding of stem cell behavior within their niches and regulation of wound healing are highlighted as examples of successful localized release systems for mimicking paracrine cell communication.
Collapse
|
269
|
Galkowski D, Ratajczak MZ, Kocki J, Darzynkiewicz Z. Of Cytometry, Stem Cells and Fountain of Youth. Stem Cell Rev Rep 2018; 13:465-481. [PMID: 28364326 DOI: 10.1007/s12015-017-9733-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Outlined are advances of cytometry applications to identify and sort stem cells, of laser scanning cytometry and ImageStream imaging instrumentation to further analyze morphometry of these cells, and of mass cytometry to classify a multitude of cellular markers in large cell populations. Reviewed are different types of stem cells, including potential candidates for cancer stem cells, with respect to their "stemness", and other characteristics. Appraised is further progress in identification and isolation of the "very small embryonic-like stem cells" (VSELs) and their autogenous transplantation for tissue repair and geroprotection. Also assessed is a function of hyaluronic acid, the major stem cells niche component, as a guardian and controller of stem cells. Briefly appraised are recent advances and challenges in the application of stem cells in regenerative medicine and oncology and their future role in different disciplines of medicine, including geriatrics.
Collapse
Affiliation(s)
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University in Lublin, 20-080, Lublin, Poland
| | - Zbigniew Darzynkiewicz
- Brander Cancer Research Institute and Department of Pathology, New York Medical College, Valhalla, NY, 10095, USA.
| |
Collapse
|
270
|
Houshmand M, Nakhlestani Hagh M, Soleimani M, Hamidieh AA, Abroun S, Nikougoftar Zarif M. MicroRNA Microarray Profiling during Megakaryocyte Differentiation of Cord Blood CD133+ Hematopoietic Stem Cells. CELL JOURNAL 2018; 20:195-203. [PMID: 29633597 PMCID: PMC5893291 DOI: 10.22074/cellj.2018.5021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 04/30/2017] [Indexed: 12/18/2022]
Abstract
Objective In order to clarify the role of microRNAs (miRNA) in megakaryocyte differentiation, we ran a microRNA microarray
experiment to measure the expression level of 961 human miRNA in megakaryocytes differentiated from human umbilical
cord blood CD133+ cells.
Materials and Methods In this experimental study, human CD133+ hematopoietic stem cells were collected from three
human umbilical cord blood (UCB) samples, and then differentiated to the megakaryocytic lineage and characterized
by flow cytometry, CFU-assay and ploidy analysis. Subsequently, microarray analysis was undertaken followed by
quantitative polymerase chain reaction (qPCR) to validate differentially expressed miRNA identified in the microarray
analysis.
Results A total of 10 and 14 miRNAs were upregulated (e.g. miR-1246 and miR-148-a) and down-regulated (e.g. miR-
551b and miR-10a) respectively during megakaryocyte differentiation, all of which were confirmed by qPCR. Analysis
of targets of these miRNA showed that the majority of targets are transcription factors involved in megakaryopoiesis.
Conclusion We conclude that miRNA play an important role in megakaryocyte differentiation and may be used as
targets to change the rate of differentiation and further our understanding of the biology of megakaryocyte commitment.
Collapse
Affiliation(s)
- Mohammad Houshmand
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.,Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Mozhde Nakhlestani Hagh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Ali Hamidieh
- Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Saeed Abroun
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahin Nikougoftar Zarif
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.,HSCT Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
271
|
Zylbersztejn F, Flores-Violante M, Voeltzel T, Nicolini FE, Lefort S, Maguer-Satta V. The BMP pathway: A unique tool to decode the origin and progression of leukemia. Exp Hematol 2018; 61:36-44. [PMID: 29477370 DOI: 10.1016/j.exphem.2018.02.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/08/2018] [Accepted: 02/13/2018] [Indexed: 12/25/2022]
Abstract
The microenvironment (niche) governs the fate of stem cells (SCs) by balancing self-renewal and differentiation. Increasing evidence indicates that the tumor niche plays an active role in cancer, but its important properties for tumor initiation progression and resistance remain to be identified. Clinical data show that leukemic stem cell (LSC) survival is responsible for disease persistence and drug resistance, probably due to their sustained interactions with the tumor niche. Bone morphogenetic protein (BMP) signaling is a key pathway controlling stem cells and their niche. BMP2 and BMP4 are important in both the normal and the cancer context. Several studies have revealed profound alterations of the BMP signaling in cancer SCs, with major deregulations of the BMP receptors and their downstream signaling elements. This was illustrated in the hematopoietic system by pioneer studies in chronic myelogenous leukemia that may now be expanded to acute myeloid leukemia and lymphoid leukemia, as reviewed here. At diagnosis, cells from the leukemic microenvironment are the major providers of soluble BMPs. Conversely, LSCs display altered receptors and downstream BMP signaling elements accompanied by altered functional responses to BMPs. These studies reveal the role of BMPs in tumor initiation, in addition to their known effects in later stages of transformation and progression. They also reveal the importance of BMPs in fueling cell transformation and expansion by overamplifying a natural SC response. This mechanism may explain the survival of LSCs independently of the initial oncogenic event and therefore may be involved in resistance processes.
Collapse
Affiliation(s)
- Florence Zylbersztejn
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France
| | - Mario Flores-Violante
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France
| | - Thibault Voeltzel
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France
| | - Franck-Emmanuel Nicolini
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France; Centre Léon Bérard, 69000 Lyon, France
| | - Sylvain Lefort
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France
| | - Véronique Maguer-Satta
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Université de Lyon, 69000, Lyon, France; Department of Signaling of Tumor Escape, Lyon, France.
| |
Collapse
|
272
|
Oikonomidou PR, Rivella S. What can we learn from ineffective erythropoiesis in thalassemia? Blood Rev 2018; 32:130-143. [PMID: 29054350 PMCID: PMC5882559 DOI: 10.1016/j.blre.2017.10.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 09/30/2017] [Accepted: 10/02/2017] [Indexed: 02/07/2023]
Abstract
Erythropoiesis is a dynamic process regulated at multiple levels to balance proliferation, differentiation and survival of erythroid progenitors. Ineffective erythropoiesis is a key feature of various diseases, including β-thalassemia. The pathogenic mechanisms leading to ineffective erythropoiesis are complex and still not fully understood. Altered survival and decreased differentiation of erythroid progenitors are both critical processes contributing to reduced production of mature red blood cells. Recent studies have identified novel important players and provided major advances in the development of targeted therapeutic approaches. In this review, β-thalassemia is used as a paradigmatic example to describe our current knowledge on the mechanisms leading to ineffective erythropoiesis and novel treatments that may have the potential to improve the clinical phenotype of associated diseases in the future.
Collapse
Affiliation(s)
- Paraskevi Rea Oikonomidou
- Department of Pediatrics, Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA.
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; Cell and Molecular Biology Graduate Group (CAMB), University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
273
|
Comoglio F, Park HJ, Schoenfelder S, Barozzi I, Bode D, Fraser P, Green AR. Thrombopoietin signaling to chromatin elicits rapid and pervasive epigenome remodeling within poised chromatin architectures. Genome Res 2018; 28:295-309. [PMID: 29429976 PMCID: PMC5848609 DOI: 10.1101/gr.227272.117] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 01/26/2018] [Indexed: 12/13/2022]
Abstract
Thrombopoietin (TPO) is a critical cytokine regulating hematopoietic stem cell maintenance and differentiation into the megakaryocytic lineage. However, the transcriptional and chromatin dynamics elicited by TPO signaling are poorly understood. Here, we study the immediate early transcriptional and cis-regulatory responses to TPO in hematopoietic stem/progenitor cells (HSPCs) and use this paradigm of cytokine signaling to chromatin to dissect the relationship between cis-regulatory activity and chromatin architecture. We show that TPO profoundly alters the transcriptome of HSPCs, with key hematopoietic regulators being transcriptionally repressed within 30 min of TPO. By examining cis-regulatory dynamics and chromatin architectures, we demonstrate that these changes are accompanied by rapid and extensive epigenome remodeling of cis-regulatory landscapes that is spatially coordinated within topologically associating domains (TADs). Moreover, TPO-responsive enhancers are spatially clustered and engage in preferential homotypic intra- and inter-TAD interactions that are largely refractory to TPO signaling. By further examining the link between cis-regulatory dynamics and chromatin looping, we show that rapid modulation of cis-regulatory activity is largely independent of chromatin looping dynamics. Finally, we show that, although activated and repressed cis-regulatory elements share remarkably similar DNA sequence compositions, transcription factor binding patterns accurately predict rapid cis-regulatory responses to TPO.
Collapse
Affiliation(s)
- Federico Comoglio
- Cambridge Institute for Medical Research, Medical Research Council/Wellcome Trust Stem Cell Institute, and Department of Haematology, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Hyun Jung Park
- Cambridge Institute for Medical Research, Medical Research Council/Wellcome Trust Stem Cell Institute, and Department of Haematology, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Stefan Schoenfelder
- Nuclear Dynamics Programme, The Babraham Institute, Cambridge CB22 3AT, United Kingdom
| | - Iros Barozzi
- Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | - Daniel Bode
- Cambridge Institute for Medical Research, Medical Research Council/Wellcome Trust Stem Cell Institute, and Department of Haematology, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Peter Fraser
- Nuclear Dynamics Programme, The Babraham Institute, Cambridge CB22 3AT, United Kingdom
- Department of Biological Science, Florida State University, Tallahassee, Florida 32301, USA
| | - Anthony R Green
- Cambridge Institute for Medical Research, Medical Research Council/Wellcome Trust Stem Cell Institute, and Department of Haematology, University of Cambridge, Cambridge CB2 0XY, United Kingdom
- Department of Haematology, Addenbrooke's Hospital, Cambridge CB2 0XY, United Kingdom
| |
Collapse
|
274
|
Abstract
Central to the classical hematopoietic stem cell (HSC) paradigm is the concept that the maintenance of blood cell numbers is exclusively executed by a discrete physical entity: the transplantable HSC. The HSC paradigm has served as a stereotypic template in stem cell biology, yet the search for rare, hardwired professional stem cells has remained futile in most other tissues. In a more open approach, the focus on the search for stem cells as a physical entity may need to be replaced by the search for stem cell function, operationally defined as the ability of an organ to replace lost cells. The nature of such a cell may be different under steady state conditions and during tissue repair. We discuss emerging examples including the renewal strategies of the skin, gut epithelium, liver, lung, and mammary gland in comparison with those of the hematopoietic system. While certain key housekeeping and developmental signaling pathways are shared between different stem cell systems, there may be no general, deeper principles underlying the renewal mechanisms of the various individual tissues.
Collapse
Affiliation(s)
- Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, 3584CT Utrecht, The Netherlands;
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, United Kingdom;
| |
Collapse
|
275
|
Shi X, Wei S, Simms KJ, Cumpston DN, Ewing TJ, Zhang P. Sonic Hedgehog Signaling Regulates Hematopoietic Stem/Progenitor Cell Activation during the Granulopoietic Response to Systemic Bacterial Infection. Front Immunol 2018. [PMID: 29535725 PMCID: PMC5834434 DOI: 10.3389/fimmu.2018.00349] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Activation and reprogramming of hematopoietic stem/progenitor cells play a critical role in the granulopoietic response to bacterial infection. Our current study determined the significance of Sonic hedgehog (SHH) signaling in the regulation of hematopoietic precursor cell activity during the host defense response to systemic bacterial infection. Bacteremia was induced in male Balb/c mice via intravenous injection (i.v.) of Escherichia coli (5 × 107 CFUs/mouse). Control mice received i.v. saline. SHH protein level in bone marrow cell (BMC) lysates was markedly increased at both 24 and 48 h of bacteremia. By contrast, the amount of soluble SHH ligand in marrow elutes was significantly reduced. These contrasting alterations suggested that SHH ligand release from BMCs was reduced and/or binding of soluble SHH ligand to BMCs was enhanced. At both 12 and 24 h of bacteremia, SHH mRNA expression by BMCs was significantly upregulated. This upregulation of SHH mRNA expression was followed by a marked increase in SHH protein expression in BMCs. Activation of the ERK1/2–SP1 pathway was involved in mediating the upregulation of SHH gene expression. The major cell type showing the enhancement of SHH expression in the bone marrow was lineage positive cells. Gli1 positioned downstream of the SHH receptor activation serves as a key component of the hedgehog (HH) pathway. Primitive hematopoietic precursor cells exhibited the highest level of baseline Gli1 expression, suggesting that they were active cells responding to SHH ligand stimulation. Along with the increased expression of SHH in the bone marrow, expression of Gli1 by marrow cells was significantly upregulated at both mRNA and protein levels following bacteremia. This enhancement of Gli1 expression was correlated with activation of hematopoietic stem/progenitor cell proliferation. Mice with Gli1 gene deletion showed attenuation in activation of marrow hematopoietic stem/progenitor cell proliferation and inhibition of increase in blood granulocytes following bacteremia. Our results indicate that SHH signaling is critically important in the regulation of hematopoietic stem/progenitor cell activation and reprogramming during the granulopoietic response to serious bacterial infection.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Shengcai Wei
- Department of Dermatology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Kevin J Simms
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Devan N Cumpston
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Thomas J Ewing
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Ping Zhang
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, United States
| |
Collapse
|
276
|
Anitua E, Troya M, Zalduendo M. Progress in the use of dental pulp stem cells in regenerative medicine. Cytotherapy 2018; 20:479-498. [PMID: 29449086 DOI: 10.1016/j.jcyt.2017.12.011] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/11/2017] [Accepted: 12/27/2017] [Indexed: 12/13/2022]
Abstract
The field of tissue engineering is emerging as a multidisciplinary area with promising potential for regenerating new tissues and organs. This approach requires the involvement of three essential components: stem cells, scaffolds and growth factors. To date, dental pulp stem cells have received special attention because they represent a readily accessible source of stem cells. Their high plasticity and multipotential capacity to differentiate into a large array of tissues can be explained by its neural crest origin, which supports applications beyond the scope of oral tissues. Many isolation, culture and cryopreservation protocols have been proposed that are known to affect cell phenotype, proliferation rate and differentiation capacity. The clinical applications of therapies based on dental pulp stem cells demand the development of new biomaterials suitable for regenerative purposes that can act as scaffolds to handle, carry and implant stem cells into patients. Currently, the development of xeno-free culture media is emerging as a means of standardization to improve safe and reproducibility. The present review aims to describe the current knowledge of dental pulp stem cells, considering in depth the key aspects related to the characterization, establishment, maintenance and cryopreservation of primary cultures and their involvement in the multilineage differentiation potential. The main clinical applications for these stem cells and their combination with several biomaterials is also covered.
Collapse
Affiliation(s)
- Eduardo Anitua
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine and Oral Implantology UIRMI, UPV/EHU-Fundación Eduardo Anitua, Vitoria, Spain.
| | - María Troya
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine and Oral Implantology UIRMI, UPV/EHU-Fundación Eduardo Anitua, Vitoria, Spain
| | - Mar Zalduendo
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine and Oral Implantology UIRMI, UPV/EHU-Fundación Eduardo Anitua, Vitoria, Spain
| |
Collapse
|
277
|
Culen M, Borsky M, Nemethova V, Razga F, Smejkal J, Jurcek T, Dvorakova D, Zackova D, Weinbergerova B, Semerad L, Sadovnik I, Eisenwort G, Herrmann H, Valent P, Mayer J, Racil Z. Quantitative assessment of the CD26+ leukemic stem cell compartment in chronic myeloid leukemia: patient-subgroups, prognostic impact, and technical aspects. Oncotarget 2018; 7:33016-24. [PMID: 27145281 PMCID: PMC5078071 DOI: 10.18632/oncotarget.9108] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/10/2016] [Indexed: 12/01/2022] Open
Abstract
Little is known about the function and phenotype of leukemic stem cells (LSCs) in chronic myeloid leukemia (CML) or about specific markers that discriminate LSCs from normal hematopoietic stem cells (HSCs). CD26 has recently been described as a specific marker of CML LSCs. In the current study, we investigated this marker in a cohort of 31 unselected CML patients. BCR/ABL1 positivity was analyzed in highly enriched stem cell fractions using fluorescence in situ hybridization (FISH) and reverse transcription PCR (RT-PCR). The proportion of CD26+ LSCs and CD26− HSCs varied considerably among the patients analyzed, and the percentage of CD26+ cells correlated with leukocyte count. The CD26 expression robustly discriminated LSCs from HSCs. This required a strict gating of the stem cell compartment. Thus, in patients with very low LSC or HSC numbers, only the highly sensitive RT-PCR method discriminated between clonal and non-clonal cells, while a robust FISH analysis required larger numbers of cells in both compartments. Finally, our data show that the numbers of CD26+ CML LSCs correlate with responses to treatment with BCR-ABL1 inhibitors.
Collapse
Affiliation(s)
- Martin Culen
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marek Borsky
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Veronika Nemethova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic.,Polymer Institute of the Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Filip Razga
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic.,Polymer Institute of the Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Jiri Smejkal
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Tomas Jurcek
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Dana Dvorakova
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Daniela Zackova
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Barbora Weinbergerova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Lukas Semerad
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Irina Sadovnik
- Department of Internal Medicine I, Division of Haematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Gregor Eisenwort
- Department of Internal Medicine I, Division of Haematology and Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Harald Herrmann
- Department of Internal Medicine I, Division of Haematology and Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Haematology and Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic.,Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Zdenek Racil
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic.,Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| |
Collapse
|
278
|
Carrelha J, Meng Y, Kettyle LM, Luis TC, Norfo R, Alcolea V, Boukarabila H, Grasso F, Gambardella A, Grover A, Högstrand K, Lord AM, Sanjuan-Pla A, Woll PS, Nerlov C, Jacobsen SEW. Hierarchically related lineage-restricted fates of multipotent haematopoietic stem cells. Nature 2018; 554:106-111. [PMID: 29298288 DOI: 10.1038/nature25455] [Citation(s) in RCA: 247] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 12/18/2017] [Indexed: 01/03/2023]
Abstract
Rare multipotent haematopoietic stem cells (HSCs) in adult bone marrow with extensive self-renewal potential can efficiently replenish all myeloid and lymphoid blood cells, securing long-term multilineage reconstitution after physiological and clinical challenges such as chemotherapy and haematopoietic transplantations. HSC transplantation remains the only curative treatment for many haematological malignancies, but inefficient blood-lineage replenishment remains a major cause of morbidity and mortality. Single-cell transplantation has uncovered considerable heterogeneity among reconstituting HSCs, a finding that is supported by studies of unperturbed haematopoiesis and may reflect different propensities for lineage-fate decisions by distinct myeloid-, lymphoid- and platelet-biased HSCs. Other studies suggested that such lineage bias might reflect generation of unipotent or oligopotent self-renewing progenitors within the phenotypic HSC compartment, and implicated uncoupling of the defining HSC properties of self-renewal and multipotency. Here we use highly sensitive tracking of progenitors and mature cells of the megakaryocyte/platelet, erythroid, myeloid and B and T cell lineages, produced from singly transplanted HSCs, to reveal a highly organized, predictable and stable framework for lineage-restricted fates of long-term self-renewing HSCs. Most notably, a distinct class of HSCs adopts a fate towards effective and stable replenishment of a megakaryocyte/platelet-lineage tree but not of other blood cell lineages, despite sustained multipotency. No HSCs contribute exclusively to any other single blood-cell lineage. Single multipotent HSCs can also fully restrict towards simultaneous replenishment of megakaryocyte, erythroid and myeloid lineages without executing their sustained lymphoid lineage potential. Genetic lineage-tracing analysis also provides evidence for an important role of platelet-biased HSCs in unperturbed adult haematopoiesis. These findings uncover a limited repertoire of distinct HSC subsets, defined by a predictable and hierarchical propensity to adopt a fate towards replenishment of a restricted set of blood lineages, before loss of self-renewal and multipotency.
Collapse
Affiliation(s)
- Joana Carrelha
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Yiran Meng
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Laura M Kettyle
- Department of Cell and Molecular Biology, Wallenberg Institute for Regenerative Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Karolinska Institutet, Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm SE-141 86, Sweden
| | - Tiago C Luis
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Ruggiero Norfo
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Verónica Alcolea
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Hanane Boukarabila
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Francesca Grasso
- Karolinska Institutet, Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm SE-141 86, Sweden
- Karolinska University Hospital, Stockholm SE-141 86, Sweden
| | - Adriana Gambardella
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Amit Grover
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Kari Högstrand
- Department of Cell and Molecular Biology, Wallenberg Institute for Regenerative Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Karolinska Institutet, Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm SE-141 86, Sweden
| | - Allegra M Lord
- Department of Cell and Molecular Biology, Wallenberg Institute for Regenerative Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Karolinska Institutet, Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm SE-141 86, Sweden
| | - Alejandra Sanjuan-Pla
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Petter S Woll
- Karolinska Institutet, Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm SE-141 86, Sweden
- Karolinska University Hospital, Stockholm SE-141 86, Sweden
| | - Claus Nerlov
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Sten Eirik W Jacobsen
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
- Department of Cell and Molecular Biology, Wallenberg Institute for Regenerative Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Karolinska Institutet, Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm SE-141 86, Sweden
- Karolinska University Hospital, Stockholm SE-141 86, Sweden
| |
Collapse
|
279
|
Antagonism of PPAR-γ signaling expands human hematopoietic stem and progenitor cells by enhancing glycolysis. Nat Med 2018; 24:360-367. [PMID: 29377004 DOI: 10.1038/nm.4477] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Hematopoietic stem cells (HSCs) quiescently reside in bone marrow niches and have the capacity to self-renew or differentiate to form all of the blood cells throughout the lifespan of an animal. Allogeneic HSC transplantation is a life-saving treatment for malignant and nonmalignant disorders. HSCs isolated from umbilical cord blood (CB) are used for hematopoietic cell transplantation (HCT), but due to the limited numbers of HSCs in single units of umbilical CB, a number of methods have been proposed for ex vivo expansion of human HSCs. We show here that antagonism of peroxisome proliferator-activated receptor (PPAR)-γ promotes ex vivo expansion of phenotypically and functionally defined subsets of human CB HSCs and hematopoietic progenitor cells (HSPCs). PPAR-γ antagonism in CB HSPCs strongly downregulated expression of several differentiation-associated genes, as well as fructose-bisphosphatase 1 (FBP1; which encodes a negative regulator of glycolysis), and enhanced glycolysis without compromising mitochondrial metabolism. The expansion of CB HSPCs by PPAR-γ antagonism was completely suppressed by removal of glucose or inhibition of glycolysis. Moreover, knockdown of FBP1 expression promoted glycolysis and ex vivo expansion of long-term repopulating CB HSPCs, whereas overexpression of FBP1 suppressed the expansion of CB HSPCs that was induced by PPAR-γ antagonism. Our study suggests the possibility for a new and simple means for metabolic reprogramming of CB HSPCs to improve the efficacy of HCT.
Collapse
|
280
|
Wang Y, Tian H, Cai W, Lian Z, Bhavanasi D, Wu C, Sato T, Kurokawa M, Wu D, Fu L, Wang H, Shen H, Liang D, Huang J. Tracking hematopoietic precursor division ex vivo in real time. Stem Cell Res Ther 2018; 9:16. [PMID: 29361987 PMCID: PMC5781326 DOI: 10.1186/s13287-017-0767-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Deciphering molecular mechanisms underlying the division of hematopoietic stem cells (HSCs) and malignant precursors would improve our understanding of the basis of stem cell-fate decisions and oncogenic transformation. METHODS Using a novel reporter of hematopoietic precursor, Evi1-GFP, we tracked the division of hematopoietic precursors in culture in real time. RESULTS First, we confirmed that Evi1-GFP is a faithful reporter of HSC activity and identified three dividing patterns of HSCs: symmetric renewal, symmetric differentiation, and asymmetric division. Moreover, we found that the cytokine and growth factor combination (STIF) promotes symmetric renewal, whereas OP9 stromal cells balance symmetric renewal and differentiation of HSCs ex vivo. Interestingly, we found that Tet2 knockout HSCs underwent more symmetric differentiation in culture compared with the wild-type control. Intriguingly, OP9 stromal cells reverse the phenotype of Tet2 knockout HSCs ex vivo. Furthermore, we demonstrated that Tet2 -/- ;Flt3ITD acute myeloid leukemia (AML) precursors primarily underwent symmetric renewal divisions in culture. Mechanistically, we demonstrated that inhibiting DNA methylation can reverse the aberrant division phenotypes of Tet2 -/- and Tet2 -/- ;FLT3ITD precursors, suggesting that abnormal DNA methylation plays an important role in controlling (pre-)leukemic precursor fate decision ex vivo. CONCLUSIONS Our study exploited a new system to explore the molecular mechanisms of the regulation of benign and malignant hematopoietic precursor division ex vivo. The knowledge learned from these studies will provide new insights into the molecular mechanisms of HSC fate decision and leukemogenesis.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Science, Peking University, Beijing, People's Republic of China.,Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Hong Tian
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Wenzhi Cai
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Zhaorui Lian
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Dheeraj Bhavanasi
- Department of Medicine (Hematology-Oncology), University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Chao Wu
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.,Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Tomohiko Sato
- Department of Hematology and Oncology, University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Mineo Kurokawa
- Department of Hematology and Oncology, University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Depei Wu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Li Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Hong Wang
- Center for Metabolic Disease Research, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dong Liang
- Department of Prenatal Diagnosis, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, Jiangsu, People's Republic of China
| | - Jian Huang
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
281
|
Yoder MC. Endothelial stem and progenitor cells (stem cells): (2017 Grover Conference Series). Pulm Circ 2018; 8:2045893217743950. [PMID: 29099663 PMCID: PMC5731724 DOI: 10.1177/2045893217743950] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/31/2017] [Indexed: 12/11/2022] Open
Abstract
The capacity of existing blood vessels to give rise to new blood vessels via endothelial cell sprouting is called angiogenesis and is a well-studied biologic process. In contrast, little is known about the mechanisms for endothelial cell replacement or regeneration within established blood vessels. Since clear definitions exist for identifying cells with stem and progenitor cell properties in many tissues and organs of the body, several groups have begun to accumulate evidence that endothelial stem and progenitor cells exist within the endothelial intima of existing blood vessels. This paper will review stem and progenitor cell definitions and highlight several recent papers purporting to have identified resident vascular endothelial stem and progenitor cells.
Collapse
Affiliation(s)
- Mervin C. Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
282
|
Arany EJ, Waseem M, Strutt BJ, Chamson-Reig A, Bernardo A, Eng E, Hill DJ. Direct comparison of the abilities of bone marrow mesenchymal versus hematopoietic stem cells to reverse hyperglycemia in diabetic NOD.SCID mice. Islets 2018; 10:137-150. [PMID: 30110202 PMCID: PMC6281365 DOI: 10.1080/19382014.2018.1480285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Both bone marrow-derived hematopoietic stem cells (HSC) and mesenchymal stem cells (MSC) improve glycemic control in diabetic mice, but their kinetics and associated changes in pancreatic morphology have not been directly compared. Our goal was to examine the time course of improvements in glucose tolerance and associated changes in β-cell mass and proliferation following transplantation of equivalent numbers of HSC or MSC from the same bone marrow into diabetic non-obese diabetic severe combined immune deficiency (NOD.SCID) mice. We used transgenic mice with a targeted expression of yellow fluorescent protein (YFP) driven by the Vav1 gene promoter to genetically tag HSC and progeny. HSC were separated from bone marrow by fluorescence-activated cell sorting and MSC following cell culture. Equivalent numbers of isolated HSC or MSC were transplanted directly into the pancreas of NOD.SCID mice previously made diabetic with streptozotocin. Glucose tolerance, serum insulin, β-cell mass and β-cell proliferation were examined up to 28 days following transplant. Transplantation with MSC improved glucose tolerance within 7 days and serum insulin levels increased, but with no increase in β-cell mass. Mice transplanted with HSC showed improved glucose tolerance only after 3 weeks associated with increased β-cell proliferation and mass. We conclude that single injections of either MSC or HSC transiently improved glycemic control in diabetic NOD.SCID mice, but with different time courses. However, only HSC infiltrated the islets and were associated with an expanded β-cell mass. This suggests that MSC and HSC have differing mechanisms of action.
Collapse
Affiliation(s)
- Edith J. Arany
- Lawson Health Research Institute, London, ON, Canada
- Department of Medicine, Western University, London, ON, Canada
- Department of Pathology, Western University, London, ON, Canada
- CONTACT Dr. David J. Hill Lawson Health Research Institute, St. Joseph’s Health Care, 268 Grosvenor St, London ON Canada N6A 4V2
| | - Muhammad Waseem
- Lawson Health Research Institute, London, ON, Canada
- International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | | | - Adam Bernardo
- Lawson Health Research Institute, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Elizabeth Eng
- Lawson Health Research Institute, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - David J. Hill
- Lawson Health Research Institute, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Department of Medicine, Western University, London, ON, Canada
| |
Collapse
|
283
|
Rossmann MP, Orkin SH, Chute JP. Hematopoietic Stem Cell Biology. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
284
|
Draper JE, Sroczynska P, Fadlullah MZH, Patel R, Newton G, Breitwieser W, Kouskoff V, Lacaud G. A novel prospective isolation of murine fetal liver progenitors to study in utero hematopoietic defects. PLoS Genet 2018; 14:e1007127. [PMID: 29300724 PMCID: PMC5754050 DOI: 10.1371/journal.pgen.1007127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/26/2017] [Indexed: 12/29/2022] Open
Abstract
In recent years, highly detailed characterization of adult bone marrow (BM) myeloid progenitors has been achieved and, as a result, the impact of somatic defects on different hematopoietic lineage fate decisions can be precisely determined. Fetal liver (FL) hematopoietic progenitor cells (HPCs) are poorly characterized in comparison, potentially hindering the study of the impact of genetic alterations on midgestation hematopoiesis. Numerous disorders, for example infant acute leukemias, have in utero origins and their study would therefore benefit from the ability to isolate highly purified progenitor subsets. We previously demonstrated that a Runx1 distal promoter (P1)-GFP::proximal promoter (P2)-hCD4 dual-reporter mouse (Mus musculus) model can be used to identify adult BM progenitor subsets with distinct lineage preferences. In this study, we undertook the characterization of the expression of Runx1-P1-GFP and P2-hCD4 in FL. Expression of P2-hCD4 in the FL immunophenotypic Megakaryocyte-Erythroid Progenitor (MEP) and Common Myeloid Progenitor (CMP) compartments corresponded to increased granulocytic/monocytic/megakaryocytic and decreased erythroid specification. Moreover, Runx1-P2-hCD4 expression correlated with several endogenous cell surface markers' expression, including CD31 and CD45, providing a new strategy for prospective identification of highly purified fetal myeloid progenitors in transgenic mouse models. We utilized this methodology to compare the impact of the deletion of either total RUNX1 or RUNX1C alone and to determine the fetal HPCs lineages most substantially affected. This new prospective identification of FL progenitors therefore raises the prospect of identifying the underlying gene networks responsible with greater precision than previously possible.
Collapse
Affiliation(s)
- Julia E. Draper
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Patrycja Sroczynska
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Muhammad Z. H. Fadlullah
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Rahima Patel
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Gillian Newton
- Molecular Biology Core Facility, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Wolfgang Breitwieser
- Molecular Biology Core Facility, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Valerie Kouskoff
- Division of Developmental Biology & Medicine, Michael Smith Building, The University of Manchester, Manchester, United Kingdom
| | - Georges Lacaud
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
285
|
Dale DC. Editorial: The mysteries of the spleen. J Leukoc Biol 2017; 100:249-51. [PMID: 27481938 DOI: 10.1189/jlb.4ce0216-088r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/29/2016] [Indexed: 11/24/2022] Open
Affiliation(s)
- David C Dale
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
286
|
Wang C, Christin JR, Oktay MH, Guo W. Lineage-Biased Stem Cells Maintain Estrogen-Receptor-Positive and -Negative Mouse Mammary Luminal Lineages. Cell Rep 2017; 18:2825-2835. [PMID: 28329676 DOI: 10.1016/j.celrep.2017.02.071] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 01/08/2017] [Accepted: 02/23/2017] [Indexed: 02/02/2023] Open
Abstract
Delineating the mammary differentiation hierarchy is important for the study of mammary gland development and tumorigenesis. Mammary luminal cells are considered a major origin of human breast cancers. However, how estrogen-receptor-positive (ER+) and ER- luminal cells are developed and maintained remains poorly understood. The prevailing model suggests that a common stem/progenitor cell generates both cell types. Through genetic lineage tracing in mice, we find that SOX9-expressing cells specifically contribute to the development and maintenance of ER- luminal cells and, to a lesser degree, basal cells. In parallel, PROM1-expressing cells give rise only to ER+ luminal cells. Both SOX9+ and PROM1+ cells specifically sustain their respective lineages even after pregnancy-caused tissue remodeling or serial transplantation, demonstrating characteristic properties of long-term repopulating stem cells. Thus, our data reveal that mouse mammary ER+ and ER- luminal cells are two independent lineages that are maintained by distinct stem cells, providing a revised mammary epithelial cell hierarchy.
Collapse
Affiliation(s)
- Chunhui Wang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John R Christin
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maja H Oktay
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10467, USA; Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Wenjun Guo
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
287
|
Peng H, Kasada A, Ueno M, Hoshii T, Tadokoro Y, Nomura N, Ito C, Takase Y, Vu HT, Kobayashi M, Xiao B, Worley PF, Hirao A. Distinct roles of Rheb and Raptor in activating mTOR complex 1 for the self-renewal of hematopoietic stem cells. Biochem Biophys Res Commun 2017; 495:1129-1135. [PMID: 29175333 DOI: 10.1016/j.bbrc.2017.11.140] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 11/20/2017] [Indexed: 02/05/2023]
Abstract
The mammalian target of rapamycin (mTOR) complex 1 (mTORC1) senses a cell's energy status and environmental levels of nutrients and growth factors. In response, mTORC1 mediates signaling that controls protein translation and cellular metabolism. Although mTORC1 plays a critical role in hematopoiesis, it remains unclear which upstream stimuli regulate mTORC1 activity in the context of hematopoietic stem cells (HSC) maintenance in vivo. In this study, we investigated the function of Rheb, a critical regulator of mTORC1 activity controlled by the PI3K-AKT-TSC axis, both in HSC maintenance in mice at steady-state and in HSC-derived hematopoiesis post-transplantation. In contrast to the severe hematopoietic dysfunction caused by Raptor deletion, which completely inactivates mTORC1, Rheb deficiency in adult mice did not show remarkable hematopoietic failure. Lack of Rheb caused abnormalities in myeloid cells but did not have impact on hematopoietic regeneration in mice subjected to injury by irradiation. As previously reported, Rheb deficiency resulted in defective HSC-derived hematopoiesis post-transplantation. However, while Raptor is essential for HSC competitiveness in vivo, Rheb is dispensable for HSC maintenance under physiological conditions, indicating that the PI3K-AKT-TSC pathway does not contribute to mTORC1 activity for sustaining HSC self-renewal activity at steady-state. Thus, the various regulatory elements that impinge upstream of mTORC1 activation pathways are differentially required for HSC homeostasis in vivo.
Collapse
Affiliation(s)
- Hui Peng
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Atsuo Kasada
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Masaya Ueno
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Takayuki Hoshii
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Yuko Tadokoro
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Naho Nomura
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Chiaki Ito
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Yusuke Takase
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Ha Thi Vu
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Masahiko Kobayashi
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Bo Xiao
- Laboratory of Developmental Neurobiology & Metabolism, State Key Laboratory of Bio-Therapy, West-China Hospital, Sichuan University, Chengdu 610041, China
| | - Paul F Worley
- Departments of Neuroscience and Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan; WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan.
| |
Collapse
|
288
|
McGonagle D, Baboolal TG, Jones E. Native joint-resident mesenchymal stem cells for cartilage repair in osteoarthritis. Nat Rev Rheumatol 2017; 13:719-730. [DOI: 10.1038/nrrheum.2017.182] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
289
|
High Level P2X7-Mediated Signaling Impairs Function of Hematopoietic Stem/Progenitor Cells. Stem Cell Rev Rep 2017; 12:305-14. [PMID: 27059869 DOI: 10.1007/s12015-016-9651-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nucleotides, which bind to P2 receptors, have emerged as a family of mediators in intercellular communication. P2X7 is a member of the P2X family ligand-gated ion channels respond to extracellular ATP. High level expression of P2X7 was detected in leukemia samples, especially in relapsed cases. However, the role of P2X7 mediated signaling in hematopoietic stem/progenitor cells (HSPCs) as well as its potential role in leukemogenesis have not been established. In this study, the expression of P2X7 in hematopoietic cells in different lineages and stages was analyzed. Over-expression of P2X7 in HSPCs was carried out by retrovirus infection to study the impact on HSPCs. The results showed that low level expression of P2X7 was detected in HSPCs. Over-expression of P2X7 in HSPCs resulted in decreased colony forming ability in vitro and engraftment potential in vivo. These results suggested that high level purinergic signaling by P2X7 impaired function of HSPCs.
Collapse
|
290
|
Abstract
Purpose of review Hematopoietic stem cell (HSC) transplantation has yielded tremendous information on experimental properties of HSCs. Yet, it remains unclear whether transplantation reflects the physiology of hematopoiesis. A limitation is the difficulty in accessing HSC functions without isolation, in-vitro manipulation and readout for potential. New genetic fate mapping and clonal marking techniques now shed light on hematopoiesis under physiological conditions. Recent findings Transposon-based genetic marks were introduced across the entire hematopoietic system to follow the clonal dynamics of these tags over time. A polyclonal source downstream from stem cells was found responsible for the production of at least granulocytes. In independent experiments, HSCs were genetically marked in adult mice, and the kinetics of label emergence throughout the system was followed over time. These experiments uncovered that during physiological steady-state hematopoiesis large numbers of HSCs yield differentiated progeny. Individual HSCs were active only rarely, indicating their very slow periodicity of differentiation rather than quiescence. Summary Noninvasive genetic experiments in mice have identified a major role of stem and progenitor cells downstream from HSCs as drivers of adult hematopoiesis, and revealed that post-transplantation hematopoiesis differs quantitatively from normal steady-state hematopoiesis.
Collapse
|
291
|
Impairment of Hematopoietic Precursor Cell Activation during the Granulopoietic Response to Bacteremia in Mice with Chronic-Plus-Binge Alcohol Administration. Infect Immun 2017; 85:IAI.00369-17. [PMID: 28784931 DOI: 10.1128/iai.00369-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/03/2017] [Indexed: 01/10/2023] Open
Abstract
Alcohol abuse impairs immune defense. To study the effect of chronic-plus-binge alcohol exposure on the granulopoietic response, acute alcohol intoxication (intraperitoneal injection of 5 g alcohol/kg body weight) was introduced to mice chronically fed on the Lieber-DeCarli low-fat liquid alcohol diet for 5 weeks. Bacteremia was induced by intravenous injection of Escherichia coli Bacteremia caused a remarkable increase in marrow lin- c-kit+ Sca-1+ cells. Activation of cell proliferation supported the increase in marrow lin- c-kit+ Sca-1+ cells. Alcohol administration inhibited this activation of lin- c-kit+ Sca-1+ cells. The bone marrow of pair-fed control mice receiving intraperitoneal saline stored a large number of mature granulocytes expressing a high level of Gr1 (Gr1hi cells). The proportion of Gr1hi cells and the total number of Gr1+ cells were markedly reduced in the bone marrow, along with an increase in the ratio of Gr1+ granulocytes in peripheral white blood cells following bacteremia. E. coli infection stimulated proliferation of granulopoietic precursor cells, resulting in a marked increase in the ratio of immature Gr1lo cells in the bone marrow. Alcohol administration itself triggered marrow release of Gr1+ cells, resulting in reduction of the marrow granulocyte reserve with an elevation of granulocytes in the circulation. Alcohol also impaired activation of granulopoietic precursor proliferation following bacteremia. Alcohol disrupted lipopolysaccharide (LPS)-TLR4-ERK1/2-cyclin D1 signaling and inhibited upregulation of Sca-1 and C/EBPβ expression by lineage-negative marrow cells in response to bacteremia. These results indicate that chronic-plus-binge alcohol exposure inhibits the granulopoietic response by disrupting key cell signaling for hematopoietic precursor cell activation and commitment to granulocyte lineage development.
Collapse
|
292
|
Steinhoff G, Nesteruk J, Wolfien M, Große J, Ruch U, Vasudevan P, Müller P. Stem cells and heart disease - Brake or accelerator? Adv Drug Deliv Rev 2017; 120:2-24. [PMID: 29054357 DOI: 10.1016/j.addr.2017.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022]
Abstract
After two decades of intensive research and attempts of clinical translation, stem cell based therapies for cardiac diseases are not getting closer to clinical success. This review tries to unravel the obstacles and focuses on underlying mechanisms as the target for regenerative therapies. At present, the principal outcome in clinical therapy does not reflect experimental evidence. It seems that the scientific obstacle is a lack of integration of knowledge from tissue repair and disease mechanisms. Recent insights from clinical trials delineate mechanisms of stem cell dysfunction and gene defects in repair mechanisms as cause of atherosclerosis and heart disease. These findings require a redirection of current practice of stem cell therapy and a reset using more detailed analysis of stem cell function interfering with disease mechanisms. To accelerate scientific development the authors suggest intensifying unified computational data analysis and shared data knowledge by using open-access data platforms.
Collapse
Affiliation(s)
- Gustav Steinhoff
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Julia Nesteruk
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Markus Wolfien
- University Rostock, Institute of Computer Science, Department of Systems Biology and Bioinformatics, Ulmenstraße 69, 18057 Rostock, Germany.
| | - Jana Große
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Ulrike Ruch
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Praveen Vasudevan
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Paula Müller
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| |
Collapse
|
293
|
Gundry MC, Dever DP, Yudovich D, Bauer DE, Haas S, Wilkinson AC, Singbrant S. Technical considerations for the use of CRISPR/Cas9 in hematology research. Exp Hematol 2017; 54:4-11. [PMID: 28757433 PMCID: PMC5603407 DOI: 10.1016/j.exphem.2017.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/19/2017] [Indexed: 11/23/2022]
Abstract
The hematopoietic system is responsible for transporting oxygen and nutrients, fighting infections, and repairing tissue damage. Hematopoietic system dysfunction therefore causes a range of serious health consequences. Lifelong hematopoiesis is maintained by repopulating multipotent hematopoietic stem cells (HSCs) that replenish shorter-lived, mature blood cell types. A prokaryotic mechanism of immunity, the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 nuclease system, has been recently "repurposed" to mutate mammalian genomes efficiently and in a sequence-specific manner. The application of this genome-editing technology to hematology has afforded new approaches for functional genomics and even the prospect of "correcting" dysfunctional HSCs in the treatment of serious genetic hematological diseases. In this Perspective, we provide an overview of three recent CRISPR/Cas9 methods in hematology: gene disruption, gene targeting, and saturating mutagenesis. We also summarize the technical considerations and advice provided during the May 2017 International Society of Experimental Hematology New Investigator Committee webinar on the same topic.
Collapse
Affiliation(s)
- Michael C Gundry
- Department of Molecular and Human Genetics, Stem Cells and Regenerative Medicine Center, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Daniel P Dever
- Department of Pediatrics, Stanford University, Stanford, CA
| | - David Yudovich
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine and Division of Stem Cells and Cancer, DKFZ German Cancer Research Centre, Heidelberg, Germany
| | - Adam C Wilkinson
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA.
| | - Sofie Singbrant
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
294
|
Tsukada M, Ota Y, Wilkinson AC, Becker HJ, Osato M, Nakauchi H, Yamazaki S. In Vivo Generation of Engraftable Murine Hematopoietic Stem Cells by Gfi1b, c-Fos, and Gata2 Overexpression within Teratoma. Stem Cell Reports 2017; 9:1024-1033. [PMID: 28943250 PMCID: PMC5639260 DOI: 10.1016/j.stemcr.2017.08.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 12/21/2022] Open
Abstract
Generation of hematopoietic stem cells (HSCs) from pluripotent stem cells (PSCs) could potentially provide unlimited HSCs for clinical transplantation, a curative treatment for numerous blood diseases. However, to date, bona fide HSC generation has been largely unsuccessful in vitro. We have previously described proof of concept for in vivo HSC generation from PSCs via teratoma formation. However, our first-generation system was complex and the output low. Here, we further optimize this technology and demonstrate the following: (1) simplified HSC generation using transcription factor overexpression; (2) improved HSC output using c-Kit-deficient host mice, and (3) that teratomas can be transplanted and cryopreserved. We demonstrate that overexpression of Gfi1b, c-Fos, and Gata2, previously reported to transdifferentiate fibroblasts into hematopoietic progenitors in vitro, can induce long-term HSC formation in vivo. Our in vivo system provides a useful platform to investigate new strategies and re-evaluate existing strategies to generate HSCs and study HSC development. iPSC overexpressing Gfi1b, c-Fos, and Gata2 (GFG) form HSC-producing teratomas Hemogenic endothelial-like and peripheral blood cells emerge in GFG teratomas Depletion of host HSCs promotes expansion of long-term, transplantable HSCs Teratomas can be cryopreserved and transplanted
Collapse
Affiliation(s)
- Masao Tsukada
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yasunori Ota
- Department of Pathology, Research Hospital, The Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Adam C Wilkinson
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Hans J Becker
- Project Division of Advanced Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Motomi Osato
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiromitsu Nakauchi
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.
| | - Satoshi Yamazaki
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Project Division of Advanced Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.
| |
Collapse
|
295
|
Tosato G. Ephrin ligands and Eph receptors contribution to hematopoiesis. Cell Mol Life Sci 2017; 74:3377-3394. [PMID: 28589441 PMCID: PMC11107787 DOI: 10.1007/s00018-017-2566-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/12/2017] [Accepted: 06/01/2017] [Indexed: 12/12/2022]
Abstract
Hematopoietic stem and progenitor cells reside predominantly in the bone marrow. They supply billions of mature blood cells every day during life through maturation into multilineage progenitors and self-renewal. Newly produced mature cells serve to replenish the pool of circulating blood cells at the end of their life-span. These mature blood cells and a few hematopoietic progenitors normally exit the bone marrow through the sinusoidal vessels, a specialized venous vascular system that spreads throughout the bone marrow. Many signals regulate the coordinated mobilization of hematopoietic cells from the bone marrow to the circulation. In this review, we present recent advances on hematopoiesis and hematopoietic cell mobilization with a focus on the role of Ephrin ligands and their Eph receptors. These constitute a large family of transmembrane ligands and receptors that play critical roles in development and postnatally. New insights point to distinct roles of ephrin and Eph in different aspects of hematopoiesis.
Collapse
Affiliation(s)
- Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 4124, Bethesda, MD, 20892, USA.
| |
Collapse
|
296
|
A New Chapter for Mesenchymal Stem Cells: Decellularized Extracellular Matrices. Stem Cell Rev Rep 2017; 13:587-597. [DOI: 10.1007/s12015-017-9757-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
297
|
Wang J, Holmes MC. Engineering hematopoietic stem cells toward a functional cure of human immunodeficiency virus infection. Cytotherapy 2017; 18:1370-1381. [PMID: 27745602 DOI: 10.1016/j.jcyt.2016.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/05/2016] [Accepted: 07/21/2016] [Indexed: 12/23/2022]
Abstract
The battle with human immunodeficiency virus (HIV) has been ongoing for more than 30 years, and although progress has been made, there are still significant challenges remaining. A few unique features render HIV to be one of the toughest viruses to conquer in the modern medicine era, such as the ability to target the host immune system, persist by integrating into the host genome and adapt to a hostile environment such as a single anti-HIV medication by continuously evolving. The finding of combination anti-retroviral therapy (cART) about 2 decades ago has transformed the treatment options for HIV-infected patients and significantly improved patient outcomes. However, finding an HIV cure has proven to be extremely challenging with the only known exception being the so-called "Berlin patient," whose immune system was replaced by stem cell transplants from a donor missing one of HIV's key co-receptors (CCR5). The broad application of this approach is limited by the requirement of an HLA-matched donor who is also homozygous for the rare CCR5 delta32 deletion. On the other hand, the Berlin patient provided the proof of concept of a potential cure for HIV using HIV-resistant hematopoietic stem cells (HSCs), revitalizing the hope to find an HIV cure that is broadly applicable. Here we will review strategies and recent attempts to engineer HIV-resistant HSCs as a path to an HIV cure.
Collapse
Affiliation(s)
- Jianbin Wang
- Sangamo BioSciences Inc., Richmond, California, USA.
| | | |
Collapse
|
298
|
miR-27b-3p, miR-181a-1-3p, and miR-326-5p are involved in the inhibition of macrophage activation in chronic liver injury. J Mol Med (Berl) 2017; 95:1091-1105. [DOI: 10.1007/s00109-017-1570-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/29/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023]
|
299
|
Ramond C, Glaser N, Berthault C, Ameri J, Kirkegaard JS, Hansson M, Honoré C, Semb H, Scharfmann R. Reconstructing human pancreatic differentiation by mapping specific cell populations during development. eLife 2017; 6. [PMID: 28731406 PMCID: PMC5540466 DOI: 10.7554/elife.27564] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/17/2017] [Indexed: 12/17/2022] Open
Abstract
Information remains scarce on human development compared to animal models. Here, we reconstructed human fetal pancreatic differentiation using cell surface markers. We demonstrate that at 7weeks of development, the glycoprotein 2 (GP2) marks a multipotent cell population that will differentiate into the acinar, ductal or endocrine lineages. Development towards the acinar lineage is paralleled by an increase in GP2 expression. Conversely, a subset of the GP2+ population undergoes endocrine differentiation by down-regulating GP2 and CD142 and turning on NEUROG3, a marker of endocrine differentiation. Endocrine maturation progresses by up-regulating SUSD2 and lowering ECAD levels. Finally, in vitro differentiation of pancreatic endocrine cells derived from human pluripotent stem cells mimics key in vivo events. Our work paves the way to extend our understanding of the origin of mature human pancreatic cell types and how such lineage decisions are regulated. DOI:http://dx.doi.org/10.7554/eLife.27564.001
Collapse
Affiliation(s)
- Cyrille Ramond
- INSERM U1016, Cochin Institute, Paris, France.,CNRS UMR 8104, Paris, France.,University of Paris Descartes, Paris, France
| | - Nicolas Glaser
- INSERM U1016, Cochin Institute, Paris, France.,CNRS UMR 8104, Paris, France.,University of Paris Descartes, Paris, France
| | | | - Jacqueline Ameri
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, Denmark, Europe
| | | | - Mattias Hansson
- Global Research External Affairs, Novo Nordisk A/S, Denmark, Europe
| | - Christian Honoré
- Department of Islet and Stem Cell Biology, Novo Nordisk A/S, Denmark, Europe
| | - Henrik Semb
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, Denmark, Europe
| | - Raphaël Scharfmann
- INSERM U1016, Cochin Institute, Paris, France.,CNRS UMR 8104, Paris, France.,University of Paris Descartes, Paris, France
| |
Collapse
|
300
|
Rowe RG, Mandelbaum J, Zon LI, Daley GQ. Engineering Hematopoietic Stem Cells: Lessons from Development. Cell Stem Cell 2017; 18:707-720. [PMID: 27257760 DOI: 10.1016/j.stem.2016.05.016] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Cell engineering has brought us tantalizingly close to the goal of deriving patient-specific hematopoietic stem cells (HSCs). While directed differentiation and transcription factor-mediated conversion strategies have generated progenitor cells with multilineage potential, to date, therapy-grade engineered HSCs remain elusive due to insufficient long-term self-renewal and inadequate differentiated progeny functionality. A cross-species approach involving zebrafish and mammalian systems offers complementary methodologies to improve understanding of native HSCs. Here, we discuss the role of conserved developmental timing processes in vertebrate hematopoiesis, highlighting how identification and manipulation of stage-specific factors that specify HSC developmental state must be harnessed to engineer HSCs for therapy.
Collapse
Affiliation(s)
- R Grant Rowe
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Joseph Mandelbaum
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - George Q Daley
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA; Manton Center for Orphan Disease Research, Boston, MA 02115, USA.
| |
Collapse
|