1
|
Wang J, Fendler NL, Shukla A, Wu SY, Challa A, Lee J, Joachimiak LA, Minna JD, Chiang CM, Vos SM, D'Orso I. ARF alters PAF1 complex integrity to selectively repress oncogenic transcription programs upon p53 loss. Mol Cell 2024; 84:4538-4557.e12. [PMID: 39532099 PMCID: PMC12001331 DOI: 10.1016/j.molcel.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 09/03/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
The polymerase associated factor 1 (PAF1) complex (PAF1c) promotes RNA polymerase II (RNA Pol II) transcription at the elongation step; however, how PAF1c transcription activity is selectively regulated during cell fate transitions remains poorly understood. Here, we reveal that the alternative reading frame (ARF) tumor suppressor operates at two levels to restrain PAF1c-dependent oncogenic transcriptional programs upon p53 loss in mouse cells. First, ARF assembles into homo-oligomers to bind the PAF1 subunit to promote PAF1c disassembly, consequently dampening PAF1c interaction with RNA Pol II and PAF1c-dependent transcription. Second, ARF targets the RUNX family transcription factor 1 (RUNX1) to selectively tune gene transcription. Consistently, ARF loss triggers RUNX1- and PAF1c-dependent transcriptional activation of pro-growth ligands (growth differentiation factor/bone morphogenetic protein [GDF/BMP]), promoting a cell-intrinsic GDF/BMP-Smad1/5 axis that aberrantly induce cell growth. Notably, pharmacologic inactivation of GDF/BMP signaling and genetic perturbation of RUNX1 significantly attenuate cell proliferation mediated by dual p53 and ARF loss, offering therapeutic utility. Our data underscore the significance of selective ARF-mediated tumor-suppressive functions through a universal transcriptional regulator.
Collapse
Affiliation(s)
- Jinli Wang
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nikole L Fendler
- Massachusetts Institute of Technology, Department of Biology, Cambridge, MA, USA
| | - Ashutosh Shukla
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shwu-Yuan Wu
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ashwini Challa
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeon Lee
- Lydia Hill Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lukasz A Joachimiak
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John D Minna
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Hamon Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cheng-Ming Chiang
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Seychelle M Vos
- Massachusetts Institute of Technology, Department of Biology, Cambridge, MA, USA; Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Iván D'Orso
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
2
|
Sánchez-Lanzas R, Jiménez-Pompa A, Ganuza M. The evolving hematopoietic niche during development. Front Mol Biosci 2024; 11:1488199. [PMID: 39417006 PMCID: PMC11480086 DOI: 10.3389/fmolb.2024.1488199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Mammalian hematopoietic stem cells (HSCs) emerge from the hemogenic endothelium in the major embryonic arteries. HSCs undergo a complex journey first migrating to the fetal liver (FL) and from there to the fetal bone marrow (FBM), where they mostly remain during adult life. In this process, a pool of adult HSCs is produced, which sustains lifelong hematopoiesis. Multiple cellular components support HSC maturation and expansion and modulate their response to environmental and developmental cues. While the adult HSC niche has been extensively studied over the last two decades, the niches present in the major embryonic arteries, FL, FBM and perinatal bone marrow (BM) are poorly described. Recent investigations highlight important differences among FL, FBM and adult BM niches and emphasize the important role that inflammation, microbiota and hormonal factors play regulating HSCs and their niches. We provide a review on our current understanding of these important cellular microenvironments across ontogeny. We mainly focused on mice, as the most widely used research model, and, when possible, include relevant insights from other vertebrates including birds, zebrafish, and human. Developing a comprehensive picture on these processes is critical to understand the earliest origins of childhood leukemia and to achieve multiple goals in regenerative medicine, such as mimicking HSC development in vitro to produce HSCs for broad transplantation purposes in leukemia, following chemotherapy, bone marrow failure, and in HSC-based gene therapy.
Collapse
Affiliation(s)
| | | | - Miguel Ganuza
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
3
|
Mozibullah M, Eslampanah Seyedi H, Khatun M, Solayman M. Identification and analysis of oncogenic non-synonymous single nucleotide polymorphisms in the human NRAS gene: An exclusive in silico study. J Genet Eng Biotechnol 2024; 22:100378. [PMID: 38797553 PMCID: PMC11087716 DOI: 10.1016/j.jgeb.2024.100378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/19/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND N-ras protein is encoded by the NRAS gene and operates as GDP-GTP-controlled on/off switching. N-ras interacts with cellular signaling networks that regulate various cellular activities including cell proliferation and survival. The nonsynonymous single nucleotide polymorphism (nsSNPs)-mediated alteration can substantially disrupt the structure and activity of the corresponding protein. N-ras has been reported to be associated with numerous diseases including cancers due to the nsSNPs. A comprehensive study on the NRAS gene to unveil the potentially damaging and oncogenic nsSNPs is yet to be accomplished. Hence, this extensive in silico study is intended to identify the disease-associated, specifically oncogenic nsSNPs of the NRAS gene. RESULTS Out of 140 missense variants, 7 nsSNPs (I55R, G60E, G60R, Y64D, L79F, D119G, and V152F) were identified to be damaging utilizing 10 computational tools that works based on different algorithms with high accuracy. Among those, G60E, G60R, and D119G variants were further filtered considering their location in the highly conserved region and later identified as oncogenic variants. Interestingly, G60E and G60R variants were revealed to be particularly associated with lung adenocarcinoma, rhabdomyosarcoma, and prostate adenocarcinoma. Therefore, D119G could be subjected to detailed investigation for identifying its association with specific cancer. CONCLUSION This in silico study identified the deleterious and oncogenic missense variants of the human NRAS gene that could be utilized for designing further experimental investigation. The outcomes of this study would be worthwhile in future research for developing personalized medicine.
Collapse
Affiliation(s)
- Md Mozibullah
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh
| | | | - Marina Khatun
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh
| | - Md Solayman
- Department of Biochemistry and Molecular Biology, Primeasia University, Bangladesh.
| |
Collapse
|
4
|
Hong L, Zhang Z, Wang Z, Yu X, Zhang J. Phase separation provides a mechanism to drive phenotype switching. Phys Rev E 2024; 109:064414. [PMID: 39021038 DOI: 10.1103/physreve.109.064414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024]
Abstract
Phenotypic switching plays a crucial role in cell fate determination across various organisms. Recent experimental findings highlight the significance of protein compartmentalization via liquid-liquid phase separation in influencing such decisions. However, the precise mechanism through which phase separation regulates phenotypic switching remains elusive. To investigate this, we established a mathematical model that couples a phase separation process and a gene expression process with feedback. We used the chemical master equation theory and mean-field approximation to study the effects of phase separation on the gene expression products. We found that phase separation can cause bistability and bimodality. Furthermore, phase separation can control the bistable properties of the system, such as bifurcation points and bistable ranges. On the other hand, in stochastic dynamics, the droplet phase exhibits double peaks within a more extensive phase separation threshold range than the dilute phase, indicating the pivotal role of the droplet phase in cell fate decisions. These findings propose an alternative mechanism that influences cell fate decisions through the phase separation process. As phase separation is increasingly discovered in gene regulatory networks, related modeling research can help build biomolecular systems with desired properties and offer insights into explaining cell fate decisions.
Collapse
|
5
|
Day RB, Hickman JA, Xu Z, Katerndahl CD, Ferraro F, Ramakrishnan SM, Erdmann-Gilmore P, Sprung RW, Mi Y, Townsend RR, Miller CA, Ley TJ. Proteogenomic analysis reveals cytoplasmic sequestration of RUNX1 by the acute myeloid leukemia-initiating CBFB::MYH11 oncofusion protein. J Clin Invest 2023; 134:e176311. [PMID: 38061017 PMCID: PMC10866659 DOI: 10.1172/jci176311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/06/2023] [Indexed: 02/16/2024] Open
Abstract
Several canonical translocations produce oncofusion genes that can initiate acute myeloid leukemia (AML). Although each translocation is associated with unique features, the mechanisms responsible remain unclear. While proteins interacting with each oncofusion are known to be relevant for how they act, these interactions have not yet been systematically defined. To address this issue in an unbiased fashion, we fused a promiscuous biotin ligase (TurboID) in-frame with 3 favorable-risk AML oncofusion cDNAs (PML::RARA, RUNX1::RUNX1T1, and CBFB::MYH11) and identified their interacting proteins in primary murine hematopoietic cells. The PML::RARA- and RUNX1::RUNX1T1-TurboID fusion proteins labeled common and unique nuclear repressor complexes, implying their nuclear localization. However, CBFB::MYH11-TurboID-interacting proteins were largely cytoplasmic, probably because of an interaction of the MYH11 domain with several cytoplasmic myosin-related proteins. Using a variety of methods, we showed that the CBFB domain of CBFB::MYH11 sequesters RUNX1 in cytoplasmic aggregates; these findings were confirmed in primary human AML cells. Paradoxically, CBFB::MYH11 expression was associated with increased RUNX1/2 expression, suggesting the presence of a sensor for reduced functional RUNX1 protein, and a feedback loop that may attempt to compensate by increasing RUNX1/2 transcription. These findings may have broad implications for AML pathogenesis.
Collapse
Affiliation(s)
- Ryan B. Day
- Section of Stem Cell Biology, Division of Oncology, Department of Internal Medicine, and
| | - Julia A. Hickman
- Section of Stem Cell Biology, Division of Oncology, Department of Internal Medicine, and
| | - Ziheng Xu
- Section of Stem Cell Biology, Division of Oncology, Department of Internal Medicine, and
| | - Casey D.S. Katerndahl
- Section of Stem Cell Biology, Division of Oncology, Department of Internal Medicine, and
| | - Francesca Ferraro
- Section of Stem Cell Biology, Division of Oncology, Department of Internal Medicine, and
| | | | - Petra Erdmann-Gilmore
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert W. Sprung
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yiling Mi
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - R. Reid Townsend
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Christopher A. Miller
- Section of Stem Cell Biology, Division of Oncology, Department of Internal Medicine, and
| | - Timothy J. Ley
- Section of Stem Cell Biology, Division of Oncology, Department of Internal Medicine, and
| |
Collapse
|
6
|
In vivo clonal tracking reveals evidence of haemangioblast and haematomesoblast contribution to yolk sac haematopoiesis. Nat Commun 2023; 14:41. [PMID: 36596806 PMCID: PMC9810727 DOI: 10.1038/s41467-022-35744-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/22/2022] [Indexed: 01/05/2023] Open
Abstract
During embryogenesis, haematopoietic and endothelial lineages emerge closely in time and space. It is thought that the first blood and endothelium derive from a common clonal ancestor, the haemangioblast. However, investigation of candidate haemangioblasts in vitro revealed the capacity for mesenchymal differentiation, a feature more compatible with an earlier mesodermal precursor. To date, no evidence for an in vivo haemangioblast has been discovered. Using single cell RNA-Sequencing and in vivo cellular barcoding, we have unravelled the ancestral relationships that give rise to the haematopoietic lineages of the yolk sac, the endothelium, and the mesenchyme. We show that the mesodermal derivatives of the yolk sac are produced by three distinct precursors with dual-lineage outcomes: the haemangioblast, the mesenchymoangioblast, and a previously undescribed cell type: the haematomesoblast. Between E5.5 and E7.5, this trio of precursors seeds haematopoietic, endothelial, and mesenchymal trajectories.
Collapse
|
7
|
Chandrakanthan V, Rorimpandey P, Zanini F, Chacon D, Olivier J, Joshi S, Kang YC, Knezevic K, Huang Y, Qiao Q, Oliver RA, Unnikrishnan A, Carter DR, Lee B, Brownlee C, Power C, Brink R, Mendez-Ferrer S, Enikolopov G, Walsh W, Göttgens B, Taoudi S, Beck D, Pimanda JE. Mesoderm-derived PDGFRA + cells regulate the emergence of hematopoietic stem cells in the dorsal aorta. Nat Cell Biol 2022; 24:1211-1225. [PMID: 35902769 PMCID: PMC9359911 DOI: 10.1038/s41556-022-00955-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 06/06/2022] [Indexed: 12/13/2022]
Abstract
Mouse haematopoietic stem cells (HSCs) first emerge at embryonic day 10.5 (E10.5), on the ventral surface of the dorsal aorta, by endothelial-to-haematopoietic transition. We investigated whether mesenchymal stem cells, which provide an essential niche for long-term HSCs (LT-HSCs) in the bone marrow, reside in the aorta-gonad-mesonephros and contribute to the development of the dorsal aorta and endothelial-to-haematopoietic transition. Here we show that mesoderm-derived PDGFRA+ stromal cells (Mesp1der PSCs) contribute to the haemogenic endothelium of the dorsal aorta and populate the E10.5-E11.5 aorta-gonad-mesonephros but by E13.5 were replaced by neural-crest-derived PSCs (Wnt1der PSCs). Co-aggregating non-haemogenic endothelial cells with Mesp1der PSCs but not Wnt1der PSCs resulted in activation of a haematopoietic transcriptional programme in endothelial cells and generation of LT-HSCs. Dose-dependent inhibition of PDGFRA or BMP, WNT and NOTCH signalling interrupted this reprogramming event. Together, aorta-gonad-mesonephros Mesp1der PSCs could potentially be harnessed to manufacture LT-HSCs from endothelium.
Collapse
Affiliation(s)
- Vashe Chandrakanthan
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia. .,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia.
| | - Prunella Rorimpandey
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Fabio Zanini
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia.,Garvan-Weizmann Centre for Cellular Genomics, Sydney, Australia.,UNSW Futures Institute for Cellular Genomics, Sydney, Australia
| | - Diego Chacon
- Centre for Health Technologies and the School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Jake Olivier
- School of Mathematics and Statistics, UNSW Sydney, Sydney, NSW, Australia
| | - Swapna Joshi
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Young Chan Kang
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Kathy Knezevic
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Yizhou Huang
- School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia.,School of Mathematics and Statistics, UNSW Sydney, Sydney, NSW, Australia.,Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Qiao Qiao
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Rema A Oliver
- Surgical & Orthopaedic Research Laboratories, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Ashwin Unnikrishnan
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Daniel R Carter
- School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia.,School of Mathematics and Statistics, UNSW Sydney, Sydney, NSW, Australia.,Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Brendan Lee
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Chris Brownlee
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Carl Power
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Robert Brink
- Garvan Institute of Medical Research, Sydney, NSW, Australia.,UNSW Sydney, Sydney, NSW, Australia
| | - Simon Mendez-Ferrer
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge, UK
| | - Grigori Enikolopov
- Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - William Walsh
- Surgical & Orthopaedic Research Laboratories, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Berthold Göttgens
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge, UK
| | - Samir Taoudi
- Epigenetics and development division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Dominik Beck
- Centre for Health Technologies and the School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - John E Pimanda
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia. .,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia. .,Department of Haematology, The Prince of Wales Hospital, Sydney, NSW, Australia.
| |
Collapse
|
8
|
Zug R. Developmental disorders caused by haploinsufficiency of transcriptional regulators: a perspective based on cell fate determination. Biol Open 2022; 11:bio058896. [PMID: 35089335 PMCID: PMC8801891 DOI: 10.1242/bio.058896] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Many human birth defects and neurodevelopmental disorders are caused by loss-of-function mutations in a single copy of transcription factor (TF) and chromatin regulator genes. Although this dosage sensitivity has long been known, how and why haploinsufficiency (HI) of transcriptional regulators leads to developmental disorders (DDs) is unclear. Here I propose the hypothesis that such DDs result from defects in cell fate determination that are based on disrupted bistability in the underlying gene regulatory network (GRN). Bistability, a crucial systems biology concept to model binary choices such as cell fate decisions, requires both positive feedback and ultrasensitivity, the latter often achieved through TF cooperativity. The hypothesis explains why dosage sensitivity of transcriptional regulators is an inherent property of fate decisions, and why disruption of either positive feedback or cooperativity in the underlying GRN is sufficient to cause disease. I present empirical and theoretical evidence in support of this hypothesis and discuss several issues for which it increases our understanding of disease, such as incomplete penetrance. The proposed framework provides a mechanistic, systems-level explanation of HI of transcriptional regulators, thus unifying existing theories, and offers new insights into outstanding issues of human disease. This article has an associated Future Leader to Watch interview with the author of the paper.
Collapse
Affiliation(s)
- Roman Zug
- Department of Biology, Lund University, 22362 Lund, Sweden
| |
Collapse
|
9
|
Tsuruda M, Morino-Koga S, Ogawa M. Bone morphogenetic protein 4 differently promotes distinct VE-cadherin + precursor stages during the definitive hematopoietic development from embryonic stem cell-derived mesodermal cells. Exp Hematol 2021; 103:40-51.e7. [PMID: 34464660 DOI: 10.1016/j.exphem.2021.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 08/04/2021] [Accepted: 08/23/2021] [Indexed: 11/30/2022]
Abstract
Definitive hematopoietic cells develop from fetal liver kinase 1 (Flk1)+ mesodermal cells during the in vitro differentiation of mouse embryonic stem cells (ESCs). VE-cadherin+CD41-CD45-(V+41-45-) hemogenic endothelial cells (HECs) and VE-cadherin+CD41+CD45- (V+41+45-) cells mediate the definitive hematopoietic development from Flk1+ cells. Bone morphogenetic protein 4 (BMP4) is known to be essential for the formation of mesoderm. However, the role of BMP4 in differentiation of the VE-cadherin+ definitive hematopoietic precursors from the mesoderm has been elusive. We addressed this issue using a co-aggregation culture of ESC-derived Flk1+ cells with OP9 stromal cells. This culture method induced V+41-45- cells, V+41+45- cells, and CD45+ cells from Flk1+ cells. V+41+45- cells possessed potential for erythromyeloid and T-lymphoid differentiation. When Flk1+ cells were cultured in the presence of a high concentration of BMP4, the generation of V+41-45- cells was enhanced. The increase in V+41-45- cells led to the subsequent increase in V+41+45- and CD45+ cells. The addition of BMP4 also increased hematopoietic colony-forming cells of various lineages. Furthermore, BMP4 promoted the expansion of V+41+45- cells independently of the preceding V+41-45- cell stage. These results suggest that BMP4 has promotive effects on the differentiation of V+41-45- HECs from Flk1+ mesodermal cells and the subsequent proliferation of V+41+45- hematopoietic precursors. These findings may provide insights for establishing a culture system to induce definitive hematopoietic stem cells from ESCs.
Collapse
Affiliation(s)
- Mariko Tsuruda
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Saori Morino-Koga
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Minetaro Ogawa
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto, Japan.
| |
Collapse
|
10
|
Weinstein N, Mendoza L, Álvarez-Buylla ER. A Computational Model of the Endothelial to Mesenchymal Transition. Front Genet 2020; 11:40. [PMID: 32226439 PMCID: PMC7080988 DOI: 10.3389/fgene.2020.00040] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Endothelial cells (ECs) form the lining of lymph and blood vessels. Changes in tissue requirements or wounds may cause ECs to behave as tip or stalk cells. Alternatively, they may differentiate into mesenchymal cells (MCs). These processes are known as EC activation and endothelial-to-mesenchymal transition (EndMT), respectively. EndMT, Tip, and Stalk EC behaviors all require SNAI1, SNAI2, and Matrix metallopeptidase (MMP) function. However, only EndMT inhibits the expression of VE-cadherin, PECAM1, and VEGFR2, and also leads to EC detachment. Physiologically, EndMT is involved in heart valve development, while a defective EndMT regulation is involved in the physiopathology of cardiovascular malformations, congenital heart disease, systemic and organ fibrosis, pulmonary arterial hypertension, and atherosclerosis. Therefore, the control of EndMT has many promising potential applications in regenerative medicine. Despite the fact that many molecular components involved in EC activation and EndMT have been characterized, the system-level molecular mechanisms involved in this process have not been elucidated. Toward this end, hereby we present Boolean network model of the molecular involved in the regulation of EC activation and EndMT. The simulated dynamic behavior of our model reaches fixed and cyclic patterns of activation that correspond to the expected EC and MC cell types and behaviors, recovering most of the specific effects of simple gain and loss-of-function mutations as well as the conditions associated with the progression of several diseases. Therefore, our model constitutes a theoretical framework that can be used to generate hypotheses and guide experimental inquiry to comprehend the regulatory mechanisms behind EndMT. Our main findings include that both the extracellular microevironment and the pattern of molecular activity within the cell regulate EndMT. EndMT requires a lack of VEGFA and sufficient oxygen in the extracellular microenvironment as well as no FLI1 and GATA2 activity within the cell. Additionally Tip cells cannot undergo EndMT directly. Furthermore, the specific conditions that are sufficient to trigger EndMT depend on the specific pattern of molecular activation within the cell.
Collapse
Affiliation(s)
- Nathan Weinstein
- Instituto de Ecología, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elena R Álvarez-Buylla
- Instituto de Ecología, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
11
|
Menegatti S, de Kruijf M, Garcia‐Alegria E, Lacaud G, Kouskoff V. Transcriptional control of blood cell emergence. FEBS Lett 2019; 593:3304-3315. [PMID: 31432499 PMCID: PMC6916194 DOI: 10.1002/1873-3468.13585] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/09/2019] [Accepted: 08/14/2019] [Indexed: 01/06/2023]
Abstract
The haematopoietic system is established during embryonic life through a series of developmental steps that culminates with the generation of haematopoietic stem cells. Characterisation of the transcriptional network that regulates blood cell emergence has led to the identification of transcription factors essential for this process. Among the many factors wired within this complex regulatory network, ETV2, SCL and RUNX1 are the central components. All three factors are absolutely required for blood cell generation, each one controlling a precise step of specification from the mesoderm germ layer to fully functional blood progenitors. Insight into the transcriptional control of blood cell emergence has been used for devising protocols to generate blood cells de novo, either through reprogramming of somatic cells or through forward programming of pluripotent stem cells. Interestingly, the physiological process of blood cell generation and its laboratory-engineered counterpart have very little in common.
Collapse
Affiliation(s)
- Sara Menegatti
- Developmental Haematopoiesis GroupFaculty of Biology, Medicine and Healththe University of ManchesterUK
| | - Marcel de Kruijf
- Developmental Haematopoiesis GroupFaculty of Biology, Medicine and Healththe University of ManchesterUK
| | - Eva Garcia‐Alegria
- Developmental Haematopoiesis GroupFaculty of Biology, Medicine and Healththe University of ManchesterUK
| | - Georges Lacaud
- Cancer Research UK Stem Cell Biology GroupCancer Research UK Manchester InstituteThe University of ManchesterMacclesfieldUK
| | - Valerie Kouskoff
- Developmental Haematopoiesis GroupFaculty of Biology, Medicine and Healththe University of ManchesterUK
| |
Collapse
|
12
|
Kaur S, Rawal P, Siddiqui H, Rohilla S, Sharma S, Tripathi DM, Baweja S, Hassan M, Vlaic S, Guthke R, Thomas M, Dayoub R, Bihari C, Sarin SK, Weiss TS. Increased Expression of RUNX1 in Liver Correlates with NASH Activity Score in Patients with Non-Alcoholic Steatohepatitis (NASH). Cells 2019; 8:cells8101277. [PMID: 31635436 PMCID: PMC6830073 DOI: 10.3390/cells8101277] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022] Open
Abstract
Given the important role of angiogenesis in liver pathology, the current study investigated the role of Runt-related transcription factor 1 (RUNX1), a regulator of developmental angiogenesis, in the pathogenesis of non-alcoholic steatohepatitis (NASH). Quantitative RT-PCRs and a transcription factor analysis of angiogenesis-associated differentially expressed genes in liver tissues of healthy controls, patients with steatosis and NASH, indicated a potential role of RUNX1 in NASH. The gene expression of RUNX1 was correlated with histopathological attributes of patients. The protein expression of RUNX1 in liver was studied by immunohistochemistry. To explore the underlying mechanisms, in vitro studies using RUNX1 siRNA and overexpression plasmids were performed in endothelial cells (ECs). RUNX1 expression was significantly correlated with inflammation, fibrosis and NASH activity score in NASH patients. Its expression was conspicuous in liver non-parenchymal cells. In vitro, factors from steatotic hepatocytes and/or VEGF or TGF- significantly induced the expression of RUNX1 in ECs. RUNX1 regulated the expression of angiogenic and adhesion molecules in ECs, including CCL2, PECAM1 and VCAM1, which was shown by silencing or over-expression of RUNX1. Furthermore, RUNX1 increased the angiogenic activity of ECs. This study reports that steatosis-induced RUNX1 augmented the expression of adhesion and angiogenic molecules and properties in ECs and may be involved in enhancing inflammation and disease severity in NASH.
Collapse
Affiliation(s)
- Savneet Kaur
- Institute of Liver and Biliary Sciences, New Delhi-110070, India.
| | - Preety Rawal
- Gautam Buddha University, Greater Noida-201308, India.
| | - Hamda Siddiqui
- Institute of Liver and Biliary Sciences, New Delhi-110070, India.
- Gautam Buddha University, Greater Noida-201308, India.
| | | | - Shvetank Sharma
- Institute of Liver and Biliary Sciences, New Delhi-110070, India.
| | | | - Sukriti Baweja
- Institute of Liver and Biliary Sciences, New Delhi-110070, India.
| | - Mohsin Hassan
- Institute of Liver and Biliary Sciences, New Delhi-110070, India.
| | - Sebastian Vlaic
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll-Institute, 07745 Jena, Germany.
| | - Reinhard Guthke
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll-Institute, 07745 Jena, Germany.
| | - Maria Thomas
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tuebingen, 72076 Tuebingen, Germany.
| | - Rania Dayoub
- University Children Hospital (KUNO), University Hospital of Regensburg, 93053 Regensburg, Germany.
| | - Chaggan Bihari
- Institute of Liver and Biliary Sciences, New Delhi-110070, India.
| | - Shiv K Sarin
- Institute of Liver and Biliary Sciences, New Delhi-110070, India.
| | - Thomas S Weiss
- University Children Hospital (KUNO), University Hospital of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
13
|
Mevel R, Draper JE, Lie-A-Ling M, Kouskoff V, Lacaud G. RUNX transcription factors: orchestrators of development. Development 2019; 146:dev148296. [PMID: 31488508 DOI: 10.1242/dev.148296] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
RUNX transcription factors orchestrate many different aspects of biology, including basic cellular and developmental processes, stem cell biology and tumorigenesis. In this Primer, we introduce the molecular hallmarks of the three mammalian RUNX genes, RUNX1, RUNX2 and RUNX3, and discuss the regulation of their activities and their mechanisms of action. We then review their crucial roles in the specification and maintenance of a wide array of tissues during embryonic development and adult homeostasis.
Collapse
Affiliation(s)
- Renaud Mevel
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK
| | - Julia E Draper
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK
| | - Michael Lie-A-Ling
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK
| | - Valerie Kouskoff
- Division of Developmental Biology & Medicine, The University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Georges Lacaud
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK
| |
Collapse
|
14
|
Dissecting BMP signaling input into the gene regulatory networks driving specification of the blood stem cell lineage. Proc Natl Acad Sci U S A 2018; 114:5814-5821. [PMID: 28584091 DOI: 10.1073/pnas.1610615114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cells (HSCs) that sustain lifelong blood production are created during embryogenesis. They emerge from a specialized endothelial population, termed hemogenic endothelium (HE), located in the ventral wall of the dorsal aorta (DA). In Xenopus, we have been studying the gene regulatory networks (GRNs) required for the formation of HSCs, and critically found that the hemogenic potential is defined at an earlier time point when precursors to the DA express hematopoietic as well as endothelial genes, in the definitive hemangioblasts (DHs). The GRN for DH programming has been constructed and, here, we show that bone morphogenetic protein (BMP) signaling is essential for the initiation of this GRN. BMP2, -4, and -7 are the principal ligands expressed in the lineage forming the HE. To investigate the requirement and timing of all BMP signaling in HSC ontogeny, we have used a transgenic line, which inducibly expresses an inhibitor of BMP signaling, Noggin, as well as a chemical inhibitor of BMP receptors, DMH1, and described the inputs from BMP signaling into the DH GRN and the HE, as well as into primitive hematopoiesis. BMP signaling is required in at least three points in DH programming: first to initiate the DH GRN through gata2 expression, then for kdr expression to enable the DH to respond to vascular endothelial growth factor A (VEGFA) ligand from the somites, and finally for gata2 expression in the DA, but is dispensable for HE specification after hemangioblasts have been formed.
Collapse
|
15
|
Lempereur A, Canto PY, Richard C, Martin S, Thalgott J, Raymond K, Lebrin F, Drevon C, Jaffredo T. The TGFβ pathway is a key player for the endothelial-to-hematopoietic transition in the embryonic aorta. Dev Biol 2017; 434:292-303. [PMID: 29253505 DOI: 10.1016/j.ydbio.2017.12.006] [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: 10/11/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/30/2022]
Abstract
The embryonic aorta produces hematopoietic stem and progenitor cells from a hemogenic endothelium localized in the aortic floor through an endothelial to hematopoietic transition. It has been long proposed that the Bone Morphogenetic Protein (BMP)/Transforming Growth Factor ß (TGFß) signaling pathway was implicated in aortic hematopoiesis but the very nature of the signal was unknown. Here, using thorough expression analysis of the BMP/TGFß signaling pathway members in the endothelial and hematopoietic compartments of the aorta at pre-hematopoietic and hematopoietic stages, we show that the TGFß pathway is preferentially balanced with a prominent role of Alk1/TgfßR2/Smad1 and 5 on both chicken and mouse species. Functional analysis using embryonic stem cells mutated for Acvrl1 revealed an enhanced propensity to produce hematopoietic cells. Collectively, we reveal that TGFß through the Alk1/TgfßR2 receptor axis is acting on endothelial cells to produce hematopoiesis.
Collapse
Affiliation(s)
- A Lempereur
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - P Y Canto
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - C Richard
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - S Martin
- CNRS UMR 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris CEDEX 05, France; MEMOLIFE Laboratory of Excellence and Paris Sciences et Lettres Research University, France
| | - J Thalgott
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - K Raymond
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - F Lebrin
- CNRS UMR 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris CEDEX 05, France; Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands; MEMOLIFE Laboratory of Excellence and Paris Sciences et Lettres Research University, France
| | - C Drevon
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - T Jaffredo
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France.
| |
Collapse
|
16
|
Twist1 regulates embryonic hematopoietic differentiation through binding to Myb and Gata2 promoter regions. Blood Adv 2017; 1:1672-1681. [PMID: 29296814 DOI: 10.1182/bloodadvances.2017006056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
Mechanisms underlying differentiation of embryonic hematopoietic stem/progenitor cells (HSPCs) remain unclear. In mouse, intra-aortic clusters (IACs) form in the aorta-gonad-mesonephros region and acquire HSPC potential after 9.5 days postcoitum (dpc). In this study we demonstrate that Twist1 is highly expressed in c-Kit+CD31+CD34+ IACs, which are equivalent to embryonic HSPCs, compared with adult HSPCs. Progenitor activities of colony-forming unit (CFU) of granulocytes and macrophages, CFU of macrophages, burst-forming unit of erythroid, and B lymphopoiesis were impaired in IACs of Twist1-/- relative to wild-type embryos. Microarray analysis and real-time polymerase chain reaction showed downregulated expression of Myb and Gata2 transcripts in Twist1-/- IACs. Chromatin immunoprecipitation and promoter binding assays indicated that Twist1 directly binds the Myb and Gata2 promoters in 10.5-dpc IACs. We conclude that Twist1 is a novel transcriptional regulator of HSPC differentiation through direct binding to promoter regions of key regulators of the process.
Collapse
|
17
|
RUNX transcription factors at the interface of stem cells and cancer. Biochem J 2017; 474:1755-1768. [PMID: 28490659 DOI: 10.1042/bcj20160632] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 12/22/2022]
Abstract
The RUNX1 transcription factor is a critical regulator of normal haematopoiesis and its functional disruption by point mutations, deletions or translocations is a major causative factor leading to leukaemia. In the majority of cases, genetic changes in RUNX1 are linked to loss of function classifying it broadly as a tumour suppressor. Despite this, several recent studies have reported the need for a certain level of active RUNX1 for the maintenance and propagation of acute myeloid leukaemia and acute lymphoblastic leukaemia cells, suggesting an oncosupportive role of RUNX1. Furthermore, in solid cancers, RUNX1 is overexpressed compared with normal tissue, and RUNX factors have recently been discovered to promote growth of skin, oral, breast and ovarian tumour cells, amongst others. RUNX factors have key roles in stem cell fate regulation during homeostasis and regeneration of many tissues. Cancer cells appear to have corrupted these stem cell-associated functions of RUNX factors to promote oncogenesis. Here, we discuss current knowledge on the role of RUNX genes in stem cells and as oncosupportive factors in haematological malignancies and epithelial cancers.
Collapse
|
18
|
Runx transcription factors in the development and function of the definitive hematopoietic system. Blood 2017; 129:2061-2069. [PMID: 28179276 DOI: 10.1182/blood-2016-12-689109] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/29/2017] [Indexed: 01/01/2023] Open
Abstract
The Runx family of transcription factors (Runx1, Runx2, and Runx3) are highly conserved and encode proteins involved in a variety of cell lineages, including blood and blood-related cell lineages, during developmental and adult stages of life. They perform activation and repressive functions in the regulation of gene expression. The requirement for Runx1 in the normal hematopoietic development and its dysregulation through chromosomal translocations and loss-of-function mutations as found in acute myeloid leukemias highlight the importance of this transcription factor in the healthy blood system. Whereas another review will focus on the role of Runx factors in leukemias, this review will provide an overview of the normal regulation and function of Runx factors in hematopoiesis and focus particularly on the biological effects of Runx1 in the generation of hematopoietic stem cells. We will present the current knowledge of the structure and regulatory features directing lineage-specific expression of Runx genes, the models of embryonic and adult hematopoietic development that provide information on their function, and some of the mechanisms by which they affect hematopoietic function.
Collapse
|
19
|
Garcia-Alegria E, Menegatti S, Batta K, Cuvertino S, Florkowska M, Kouskoff V. Emerging concepts for the in vitro derivation of murine haematopoietic stem and progenitor cells. FEBS Lett 2016; 590:4116-4125. [PMID: 27404333 DOI: 10.1002/1873-3468.12300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/04/2016] [Accepted: 07/09/2016] [Indexed: 02/11/2024]
Abstract
Well into the second decade of the 21st century, the field of regenerative medicine is bursting with hopes and promises to heal young and old. The bespoken generation of cells is thought to offer unprecedented cures for a vast range of diseases. Haematological disorders have already benefited tremendously from stem cell therapy in the form of bone marrow transplantation. However, lack of compatible donors often means that patients remain on transplantation waiting lists for too long. The in vitro derivation of haematopoietic stem cells offers the possibility to generate tailor-made cells for the treatment of these patients. Promising approaches to generate in vitro-derived blood progenitors include the directed differentiation of pluripotent stem cells and the reprogramming of somatic cells.
Collapse
Affiliation(s)
- Eva Garcia-Alegria
- Cancer Research UK Stem Cell Haematopoiesis Group, Cancer Research UK Manchester Institute, The University of Manchester, UK
| | - Sara Menegatti
- Cancer Research UK Stem Cell Haematopoiesis Group, Cancer Research UK Manchester Institute, The University of Manchester, UK
| | - Kiran Batta
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, UK
| | - Sara Cuvertino
- Cancer Research UK Stem Cell Haematopoiesis Group, Cancer Research UK Manchester Institute, The University of Manchester, UK
| | - Magdalena Florkowska
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, UK
| | - Valerie Kouskoff
- Cancer Research UK Stem Cell Haematopoiesis Group, Cancer Research UK Manchester Institute, The University of Manchester, UK
| |
Collapse
|
20
|
Eliades A, Wareing S, Marinopoulou E, Fadlullah MZH, Patel R, Grabarek JB, Plusa B, Lacaud G, Kouskoff V. The Hemogenic Competence of Endothelial Progenitors Is Restricted by Runx1 Silencing during Embryonic Development. Cell Rep 2016; 15:2185-2199. [PMID: 27239041 PMCID: PMC4906370 DOI: 10.1016/j.celrep.2016.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 03/24/2016] [Accepted: 04/27/2016] [Indexed: 01/08/2023] Open
Abstract
It is now well-established that hematopoietic stem cells (HSCs) and progenitor cells originate from a specialized subset of endothelium, termed hemogenic endothelium (HE), via an endothelial-to-hematopoietic transition. However, the molecular mechanisms determining which endothelial progenitors possess this hemogenic potential are currently unknown. Here, we investigated the changes in hemogenic potential in endothelial progenitors at the early stages of embryonic development. Using an ETV2::GFP reporter mouse to isolate emerging endothelial progenitors, we observed a dramatic decrease in hemogenic potential between embryonic day (E)7.5 and E8.5. At the molecular level, Runx1 is expressed at much lower levels in E8.5 intra-embryonic progenitors, while Bmi1 expression is increased. Remarkably, the ectopic expression of Runx1 in these progenitors fully restores their hemogenic potential, as does the suppression of BMI1 function. Altogether, our data demonstrate that hemogenic competency in recently specified endothelial progenitors is restrained through the active silencing of Runx1 expression.
Collapse
Affiliation(s)
- Alexia Eliades
- Cancer Research UK Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Sarah Wareing
- Cancer Research UK Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Elli Marinopoulou
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Muhammad Z H Fadlullah
- Cancer Research UK Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Rahima Patel
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Joanna B Grabarek
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, UK
| | - Berenika Plusa
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, UK
| | - Georges Lacaud
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK.
| | - Valerie Kouskoff
- Cancer Research UK Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK.
| |
Collapse
|
21
|
Lelieveld SH, Schütte J, Dijkstra MJJ, Bawono P, Kinston SJ, Göttgens B, Heringa J, Bonzanni N. ConBind: motif-aware cross-species alignment for the identification of functional transcription factor binding sites. Nucleic Acids Res 2016; 44:e72. [PMID: 26721389 PMCID: PMC4856970 DOI: 10.1093/nar/gkv1518] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/23/2022] Open
Abstract
Eukaryotic gene expression is regulated by transcription factors (TFs) binding to promoter as well as distal enhancers. TFs recognize short, but specific binding sites (TFBSs) that are located within the promoter and enhancer regions. Functionally relevant TFBSs are often highly conserved during evolution leaving a strong phylogenetic signal. While multiple sequence alignment (MSA) is a potent tool to detect the phylogenetic signal, the current MSA implementations are optimized to align the maximum number of identical nucleotides. This approach might result in the omission of conserved motifs that contain interchangeable nucleotides such as the ETS motif (IUPAC code: GGAW). Here, we introduce ConBind, a novel method to enhance alignment of short motifs, even if their mutual sequence similarity is only partial. ConBind improves the identification of conserved TFBSs by improving the alignment accuracy of TFBS families within orthologous DNA sequences. Functional validation of the Gfi1b + 13 enhancer reveals that ConBind identifies additional functionally important ETS binding sites that were missed by all other tested alignment tools. In addition to the analysis of known regulatory regions, our web tool is useful for the analysis of TFBSs on so far unknown DNA regions identified through ChIP-sequencing.
Collapse
Affiliation(s)
- Stefan H Lelieveld
- Centre for Integrative Bioinformatics VU, VU University Amsterdam, Amsterdam 1081 HV, The Netherlands Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands
| | - Judith Schütte
- Department of Haematology, Wellcome Trust-MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical Research, Cambridge University, Cambridge CB2 0XY, UK Klinik für Hämatologie, Universitätsklinik Essen 45147, Germany
| | - Maurits J J Dijkstra
- Centre for Integrative Bioinformatics VU, VU University Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Punto Bawono
- Centre for Integrative Bioinformatics VU, VU University Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Sarah J Kinston
- Department of Haematology, Wellcome Trust-MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical Research, Cambridge University, Cambridge CB2 0XY, UK
| | - Berthold Göttgens
- Department of Haematology, Wellcome Trust-MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical Research, Cambridge University, Cambridge CB2 0XY, UK
| | - Jaap Heringa
- Centre for Integrative Bioinformatics VU, VU University Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Nicola Bonzanni
- Centre for Integrative Bioinformatics VU, VU University Amsterdam, Amsterdam 1081 HV, The Netherlands Computational Cancer Biology Group, Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands ENPICOM, Eindhoven 5632 CW, The Netherlands
| |
Collapse
|
22
|
Yoshikawa M, Masuda T, Kobayashi A, Senzaki K, Ozaki S, Aizawa S, Shiga T. Runx1 contributes to the functional switching of bone morphogenetic protein 4 (BMP4) from neurite outgrowth promoting to suppressing in dorsal root ganglion. Mol Cell Neurosci 2016; 72:114-22. [DOI: 10.1016/j.mcn.2016.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 02/10/2016] [Accepted: 02/11/2016] [Indexed: 10/22/2022] Open
|
23
|
Kanz D, Konantz M, Alghisi E, North TE, Lengerke C. Endothelial-to-hematopoietic transition: Notch-ing vessels into blood. Ann N Y Acad Sci 2016; 1370:97-108. [DOI: 10.1111/nyas.13030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Dirk Kanz
- Department of Stem Cell and Regenerative Biology; Harvard University; Boston Massachusetts
| | - Martina Konantz
- Department of Biomedicine; University Hospital Basel; Basel Switzerland
| | - Elisa Alghisi
- Department of Biomedicine; University Hospital Basel; Basel Switzerland
| | - Trista E. North
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston Massachusetts
- Harvard Stem Cell Institute; Cambridge Massachusetts
| | - Claudia Lengerke
- Department of Biomedicine; University Hospital Basel; Basel Switzerland
- Division of Hematology; University Hospital Basel; Basel Switzerland
| |
Collapse
|
24
|
Huang Y, Thoms JAI, Tursky ML, Knezevic K, Beck D, Chandrakanthan V, Suryani S, Olivier J, Boulton A, Glaros EN, Thomas SR, Lock RB, MacKenzie KL, Bushweller JH, Wong JWH, Pimanda JE. MAPK/ERK2 phosphorylates ERG at serine 283 in leukemic cells and promotes stem cell signatures and cell proliferation. Leukemia 2016; 30:1552-61. [PMID: 27055868 DOI: 10.1038/leu.2016.55] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 12/23/2015] [Accepted: 02/02/2016] [Indexed: 12/19/2022]
Abstract
Aberrant ERG (v-ets avian erythroblastosis virus E26 oncogene homolog) expression drives leukemic transformation in mice and high expression is associated with poor patient outcomes in acute myeloid leukemia (AML) and T-acute lymphoblastic leukemia (T-ALL). Protein phosphorylation regulates the activity of many ETS factors but little is known about ERG in leukemic cells. To characterize ERG phosphorylation in leukemic cells, we applied liquid chromatography coupled tandem mass spectrometry and identified five phosphorylated serines on endogenous ERG in T-ALL and AML cells. S283 was distinct as it was abundantly phosphorylated in leukemic cells but not in healthy hematopoietic stem and progenitor cells (HSPCs). Overexpression of a phosphoactive mutant (S283D) increased expansion and clonogenicity of primary HSPCs over and above wild-type ERG. Using a custom antibody, we screened a panel of primary leukemic xenografts and showed that ERG S283 phosphorylation was mediated by mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling and in turn regulated expression of components of this pathway. S283 phosphorylation facilitates ERG enrichment and transactivation at the ERG +85 HSPC enhancer that is active in AML and T-ALL with poor prognosis. Taken together, we have identified a specific post-translational modification in leukemic cells that promotes progenitor proliferation and is a potential target to modulate ERG-driven transcriptional programs in leukemia.
Collapse
Affiliation(s)
- Y Huang
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - J A I Thoms
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - M L Tursky
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia.,Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - K Knezevic
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - D Beck
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - V Chandrakanthan
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - S Suryani
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - J Olivier
- School of Mathematics and Statistics, UNSW Australia, Sydney, New South Wales, Australia
| | - A Boulton
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - E N Glaros
- School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - S R Thomas
- School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - R B Lock
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - K L MacKenzie
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - J H Bushweller
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - J W H Wong
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - J E Pimanda
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales, Australia.,Department of Hematology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
25
|
Souilhol C, Gonneau C, Lendinez JG, Batsivari A, Rybtsov S, Wilson H, Morgado-Palacin L, Hills D, Taoudi S, Antonchuk J, Zhao S, Medvinsky A. Inductive interactions mediated by interplay of asymmetric signalling underlie development of adult haematopoietic stem cells. Nat Commun 2016; 7:10784. [PMID: 26952187 PMCID: PMC4786750 DOI: 10.1038/ncomms10784] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 01/21/2016] [Indexed: 01/07/2023] Open
Abstract
During embryonic development, adult haematopoietic stem cells (HSCs) emerge preferentially in the ventral domain of the aorta in the aorta-gonad-mesonephros (AGM) region. Several signalling pathways such as Notch, Wnt, Shh and RA are implicated in this process, yet how these interact to regulate the emergence of HSCs has not previously been described in mammals. Using a combination of ex vivo and in vivo approaches, we report here that stage-specific reciprocal dorso-ventral inductive interactions and lateral input from the urogenital ridges are required to drive HSC development in the aorta. Our study strongly suggests that these inductive interactions in the AGM region are mediated by the interplay between spatially polarized signalling pathways. Specifically, Shh produced in the dorsal region of the AGM, stem cell factor in the ventral and lateral regions, and BMP inhibitory signals in the ventral tissue are integral parts of the regulatory system involved in the development of HSCs.
Collapse
Affiliation(s)
- Céline Souilhol
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| | - Christèle Gonneau
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| | - Javier G. Lendinez
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| | - Antoniana Batsivari
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| | - Stanislav Rybtsov
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| | - Heather Wilson
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| | - Lucia Morgado-Palacin
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| | - David Hills
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| | - Samir Taoudi
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052 Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Victoria 3052 Melbourne, Australia
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052 Melbourne, Australia
| | | | - Suling Zhao
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| | - Alexander Medvinsky
- Institute for Stem Cell Research, Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, SCRM Bioquarter, 5 Little France Drive, Edinburgh EH16 4UU, Scotland, UK
| |
Collapse
|
26
|
Martinez M, Hinojosa M, Trombly D, Morin V, Stein J, Stein G, Javed A, Gutierrez SE. Transcriptional Auto-Regulation of RUNX1 P1 Promoter. PLoS One 2016; 11:e0149119. [PMID: 26901859 PMCID: PMC4762634 DOI: 10.1371/journal.pone.0149119] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 01/27/2016] [Indexed: 12/11/2022] Open
Abstract
RUNX1 a member of the family of runt related transcription factors (RUNX), is essential for hematopoiesis. The expression of RUNX1 gene is controlled by two promoters; the distal P1 promoter and the proximal P2 promoter. Several isoforms of RUNX1 mRNA are generated through the use of both promoters and alternative splicing. These isoforms not only differs in their temporal expression pattern but also exhibit differences in tissue specificity. The RUNX1 isoforms derived from P2 are expressed in a variety of tissues, but expression of P1-derived isoform is restricted to cells of hematopoietic lineage. However, the control of hematopoietic-cell specific expression is poorly understood. Here we report regulation of P1-derived RUNX1 mRNA by RUNX1 protein. In silico analysis of P1 promoter revealed presence of two evolutionary conserved RUNX motifs, 0.6kb upstream of the transcription start site, and three RUNX motifs within 170bp of the 5'UTR. Transcriptional contribution of these RUNX motifs was studied in myeloid and T-cells. RUNX1 genomic fragment containing all sites show very low basal activity in both cell types. Mutation or deletion of RUNX motifs in the UTR enhances basal activity of the RUNX1 promoter. Chromatin immunoprecipitation revealed that RUNX1 protein is recruited to these sites. Overexpression of RUNX1 in non-hematopoietic cells results in a dose dependent activation of the RUNX1 P1 promoter. We also demonstrate that RUNX1 protein regulates transcription of endogenous RUNX1 mRNA in T-cell. Finally we show that SCL transcription factor is recruited to regions containing RUNX motifs in the promoter and the UTR and regulates activity of the RUNX1 P1 promoter in vitro. Thus, multiple lines of evidence show that RUNX1 protein regulates its own gene transcription.
Collapse
Affiliation(s)
- Milka Martinez
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Biologicas, Universidad de Concepcion, Concepcion, Chile
| | - Marcela Hinojosa
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Biologicas, Universidad de Concepcion, Concepcion, Chile
| | - Daniel Trombly
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, 01655, Massachusetts, United States of America
| | - Violeta Morin
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Biologicas, Universidad de Concepcion, Concepcion, Chile
| | - Janet Stein
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, 01655, Massachusetts, United States of America
- Department of Biochemistry and Vermont Cancer Center, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington 05405, Vermont, United States of America
| | - Gary Stein
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, 01655, Massachusetts, United States of America
- Department of Biochemistry and Vermont Cancer Center, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington 05405, Vermont, United States of America
| | - Amjad Javed
- Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama at Birmingham, Alabama, United States of America
| | - Soraya E. Gutierrez
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Biologicas, Universidad de Concepcion, Concepcion, Chile
- * E-mail:
| |
Collapse
|
27
|
|
28
|
Yzaguirre AD, Padmanabhan A, de Groh ED, Engleka KA, Li J, Speck NA, Epstein JA. Loss of neurofibromin Ras-GAP activity enhances the formation of cardiac blood islands in murine embryos. eLife 2015; 4:e07780. [PMID: 26460546 PMCID: PMC4714971 DOI: 10.7554/elife.07780] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 10/12/2015] [Indexed: 12/15/2022] Open
Abstract
Type I neurofibromatosis (NF1) is caused by mutations in the NF1 gene encoding neurofibromin. Neurofibromin exhibits Ras GTPase activating protein (Ras-GAP) activity that is thought to mediate cellular functions relevant to disease phenotypes. Loss of murine Nf1 results in embryonic lethality due to heart defects, while mice with monoallelic loss of function mutations or with tissue-specific inactivation have been used to model NF1. Here, we characterize previously unappreciated phenotypes in Nf1-/- embryos, which are inhibition of hemogenic endothelial specification in the dorsal aorta, enhanced yolk sac hematopoiesis, and exuberant cardiac blood island formation. We show that a missense mutation engineered into the active site of the Ras-GAP domain is sufficient to reproduce ectopic blood island formation, cardiac defects, and overgrowth of neural crest-derived structures seen in Nf1-/-embryos. These findings demonstrate a role for Ras-GAP activity in suppressing the hemogenic potential of the heart and restricting growth of neural crest-derived tissues. DOI:http://dx.doi.org/10.7554/eLife.07780.001 Messages are carried from the surface of a cell to the cell’s nucleus in order to regulate various processes such as how often the cell will divide. The Ras-signaling pathway carries some of these messages. A gene called Nf1 encodes a protein in this pathway that deactivates another protein called Ras when the message is no longer required. If a mutation in Nf1 prevents it from deactivating Ras, the pathway becomes hyperactivated. In humans, this results in a disorder called Neurofibromatosis type I, which is characterized by tumors that affect many parts of the body. When the expression of Nf1 is turned off in mice, the mice die as embryos because of cardiac defects. But a mouse in which Nf1 has been turned off in specific organs or tissues other than the heart can survive, and these mice are used to model Neurofibromatosis type I and to help to identify potential treatments. Yzaguirre et al. have now identified new roles for Nf1 during embryonic development. In the embryo, blood cells originate from the cells lining the blood vessels. The experiments revealed that, when the Nf1 gene was mutated in mice, fewer blood cells formed from the lining of the major blood vessel that leaves the embryonic heart. In contrast, these mutant mice formed more structures called cardiac blood islands than a normal mouse. These structures line the heart, and have the potential to generate new blood cells for the heart to pump. These results shed new light on how blood is originally formed from the lining of the heart and blood vessels, and show that Ras signaling must be tightly regulated to maintain normal blood development in the embryo. Furthermore, Yzaguirre et al. demonstrated that this excessive formation of cardiac blood islands resulted specifically from the loss of Nf1’s role in the Ras-signaling pathway. This was achieved by using gene targeting to generate a mouse that expresses Nf1 with a minor change that affects only the protein’s interaction with Ras. In the future, this new strain of mouse will be a useful tool in determining if specific aspects of Neurofibromatosis type I can be attributed to loss of Nf1’s role in Ras-signaling and could therefore be treated by medicines that target this pathway. DOI:http://dx.doi.org/10.7554/eLife.07780.002
Collapse
Affiliation(s)
- Amanda D Yzaguirre
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States.,Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| | - Arun Padmanabhan
- Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States.,Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| | - Eric D de Groh
- Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States.,Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| | - Kurt A Engleka
- Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States.,Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| | - Jun Li
- Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States.,Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| | - Nancy A Speck
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States.,Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| | - Jonathan A Epstein
- Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States.,Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
29
|
Xie J, Zhang C. Ex vivo expansion of hematopoietic stem cells. SCIENCE CHINA-LIFE SCIENCES 2015; 58:839-53. [PMID: 26246379 DOI: 10.1007/s11427-015-4895-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 06/03/2015] [Indexed: 02/03/2023]
Abstract
Ex vivo expansion of hematopoietic stem cells (HSCs) would benefit clinical applications in several aspects, to improve patient survival, utilize cord blood stem cells for adult applications, and selectively propagate stem cell populations after genetic manipulation. In this review we summarize and discuss recent advances in the culture systems of mouse and human HSCs, which include stroma/HSC co-culture, continuous perfusion and fed-batch cultures, and those supplemented with extrinsic ligands, membrane transportable transcription factors, complement components, protein modification enzymes, metabolites, or small molecule chemicals. Some of the expansion systems have been tested in clinical trials. The optimal condition for ex vivo expansion of the primitive and functional human HSCs is still under development. An improved understanding of the mechanisms for HSC cell fate determination and the HSC culture characteristics will guide development of new strategies to overcome difficulties. In the future, development of a combination treatment regimen with agents that enhance self-renewal, block differentiation, and improve homing will be critical. Methods to enhance yields and lower cost during collection and processing should be employed. The employment of an efficient system for ex vivo expansion of HSCs will facilitate the further development of novel strategies for cell and gene therapies including genome editing.
Collapse
Affiliation(s)
- JingJing Xie
- Taishan Scholar Immunology Program, Binzhou Medical University, Yantai, 264003, China
- Departments of Physiology and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, 75390, USA
| | - ChengCheng Zhang
- Departments of Physiology and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, 75390, USA.
| |
Collapse
|
30
|
Marks-Bluth J, Khanna A, Chandrakanthan V, Thoms J, Bee T, Eich C, Kang YC, Knezevic K, Qiao Q, Fitch S, Oxburgh L, Ottersbach K, Dzierzak E, de Bruijn MFTR, Pimanda JE. SMAD1 and SMAD5 Expression Is Coordinately Regulated by FLI1 and GATA2 during Endothelial Development. Mol Cell Biol 2015; 35:2165-72. [PMID: 25870111 PMCID: PMC4438244 DOI: 10.1128/mcb.00239-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/01/2015] [Accepted: 04/03/2015] [Indexed: 01/02/2023] Open
Abstract
The bone morphogenetic protein (BMP)/SMAD signaling pathway is a critical regulator of angiogenic sprouting and is involved in vascular development in the embryo. SMAD1 and SMAD5, the core mediators of BMP signaling, are vital for this activity, yet little is known about their transcriptional regulation in endothelial cells. Here, we have integrated multispecies sequence conservation, tissue-specific chromatin, in vitro reporter assay, and in vivo transgenic data to identify and validate Smad1+63 and the Smad5 promoter as tissue-specific cis-regulatory elements that are active in the developing endothelium. The activity of these elements in the endothelium was dependent on highly conserved ETS, GATA, and E-box motifs, and chromatin immunoprecipitation showed high levels of enrichment of FLI1, GATA2, and SCL at these sites in endothelial cell lines and E11 dorsal aortas in vivo. Knockdown of FLI1 and GATA2 but not SCL reduced the expression of SMAD1 and SMAD5 in endothelial cells in vitro. In contrast, CD31(+) cKit(-) endothelial cells harvested from embryonic day 9 (E9) aorta-gonad-mesonephros (AGM) regions of GATA2 null embryos showed reduced Smad1 but not Smad5 transcript levels. This is suggestive of a degree of in vivo selection where, in the case of reduced SMAD1 levels, endothelial cells with more robust SMAD5 expression have a selective advantage.
Collapse
Affiliation(s)
- Jonathon Marks-Bluth
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Anchit Khanna
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia
| | - Vashe Chandrakanthan
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia
| | - Julie Thoms
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia
| | - Thomas Bee
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia
| | - Christina Eich
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Young Chan Kang
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia
| | - Kathy Knezevic
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia
| | - Qiao Qiao
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia
| | - Simon Fitch
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Katrin Ottersbach
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Elaine Dzierzak
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands The Queen's Medical Research Institute, College of Medicine and Veterinary Medicine, Edinburgh, United Kingdom
| | - Marella F T R de Bruijn
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - John E Pimanda
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia Department of Haematology, Prince of Wales Hospital, Sydney, NSW, Australia
| |
Collapse
|
31
|
Nasrallah R, Knezevic K, Thai T, Thomas SR, Göttgens B, Lacaud G, Kouskoff V, Pimanda JE. Endoglin potentiates nitric oxide synthesis to enhance definitive hematopoiesis. Biol Open 2015; 4:819-29. [PMID: 25979706 PMCID: PMC4571086 DOI: 10.1242/bio.011494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/14/2015] [Indexed: 01/12/2023] Open
Abstract
During embryonic development, hematopoietic cells develop by a process of endothelial-to hematopoietic transition of a specialized population of endothelial cells. These hemogenic endothelium (HE) cells in turn develop from a primitive population of FLK1(+) mesodermal cells. Endoglin (ENG) is an accessory TGF-β receptor that is enriched on the surface of endothelial and hematopoietic stem cells and is also required for the normal development of hemogenic precursors. However, the functional role of ENG during the transition of FLK1(+) mesoderm to hematopoietic cells is ill defined. To address this we used a murine embryonic stem cell model that has been shown to mirror the temporal emergence of these cells in the embryo. We noted that FLK1(+) mesodermal cells expressing ENG generated fewer blast colony-forming cells but had increased hemogenic potential when compared with ENG non-expressing cells. TIE2(+)/CD117(+) HE cells expressing ENG also showed increased hemogenic potential compared with non-expressing cells. To evaluate whether high ENG expression accelerates hematopoiesis, we generated an inducible ENG expressing ES cell line and forced expression in FLK1(+) mesodermal or TIE2(+)/CD117(+) HE cells. High ENG expression at both stages accelerated the emergence of CD45(+) definitive hematopoietic cells. High ENG expression was associated with increased pSMAD2/eNOS expression and NO synthesis in hemogenic precursors. Inhibition of eNOS blunted the ENG induced increase in definitive hematopoiesis. Taken together, these data show that ENG potentiates the emergence of definitive hematopoietic cells by modulating TGF-β/pSMAD2 signalling and increasing eNOS/NO synthesis.
Collapse
Affiliation(s)
- Rabab Nasrallah
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, UNSW Australia, Sydney, NSW 2052, Australia Cancer Research UK Manchester Institute, The University of Manchester, Manchester, M20 4BX, UK
| | - Kathy Knezevic
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, UNSW Australia, Sydney, NSW 2052, Australia
| | - Thuan Thai
- Centre for Vascular Research and School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
| | - Shane R Thomas
- Centre for Vascular Research and School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK
| | - Georges Lacaud
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, M20 4BX, UK
| | - Valerie Kouskoff
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, M20 4BX, UK
| | - John E Pimanda
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, UNSW Australia, Sydney, NSW 2052, Australia Department of Haematology, Prince of Wales Hospital, Sydney, NSW 2031, Australia
| |
Collapse
|
32
|
Pearson S, Cuvertino S, Fleury M, Lacaud G, Kouskoff V. In vivo repopulating activity emerges at the onset of hematopoietic specification during embryonic stem cell differentiation. Stem Cell Reports 2015; 4:431-44. [PMID: 25660408 PMCID: PMC4375940 DOI: 10.1016/j.stemcr.2015.01.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 01/07/2015] [Accepted: 01/07/2015] [Indexed: 01/12/2023] Open
Abstract
The generation of in vivo repopulating hematopoietic cells from in vitro differentiating embryonic stem cells has remained a long-standing challenge. To date, hematopoietic engraftment has mostly been achieved through the enforced expression of ectopic transcription factors. Here, we describe serum-free culture conditions that allow the generation of in vivo repopulating hematopoietic cells in the absence of ectopically expressed factors. We show that repopulating activity arises immediately upon the commitment of mesodermal precursors to the blood program, within the first wave of hematopoietic specification. We establish that the formation of these progenitors is extremely transient and exquisitely sensitive to the cytokine milieu. Our findings define the precise differentiating stage at which hematopoietic repopulating activity first appears in vitro, and suggest that during embryonic stem cell differentiation, all hematopoietic programs are unraveled simultaneously from the mesoderm in the absence of cues that restrict the coordinated emergence of each lineage as is normally observed during embryogenesis.
Collapse
Affiliation(s)
- Stella Pearson
- Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Sara Cuvertino
- Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Maud Fleury
- Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Georges Lacaud
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK.
| | - Valerie Kouskoff
- Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK.
| |
Collapse
|
33
|
Perna F, Vu LP, Themeli M, Kriks S, Hoya-Arias R, Khanin R, Hricik T, Mansilla-Soto J, Papapetrou EP, Levine RL, Studer L, Sadelain M, Nimer SD. The polycomb group protein L3MBTL1 represses a SMAD5-mediated hematopoietic transcriptional program in human pluripotent stem cells. Stem Cell Reports 2015; 4:658-69. [PMID: 25754204 PMCID: PMC4400644 DOI: 10.1016/j.stemcr.2015.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 01/11/2023] Open
Abstract
Epigenetic regulation of key transcriptional programs is a critical mechanism that controls hematopoietic development, and, thus, aberrant expression patterns or mutations in epigenetic regulators occur frequently in hematologic malignancies. We demonstrate that the Polycomb protein L3MBTL1, which is monoallelically deleted in 20q- myeloid malignancies, represses the ability of stem cells to drive hematopoietic-specific transcriptional programs by regulating the expression of SMAD5 and impairing its recruitment to target regulatory regions. Indeed, knockdown of L3MBTL1 promotes the development of hematopoiesis and impairs neural cell fate in human pluripotent stem cells. We also found a role for L3MBTL1 in regulating SMAD5 target gene expression in mature hematopoietic cell populations, thereby affecting erythroid differentiation. Taken together, we have identified epigenetic priming of hematopoietic-specific transcriptional networks, which may assist in the development of therapeutic approaches for patients with anemia. L3MBTL1 is a chromatin-binding protein that represses SMAD5 expression Lack of L3MBTL1 primes the hematopoietic development of pluripotent stem cells L3MBTL1 regulates erythroid differentiation
Collapse
Affiliation(s)
- Fabiana Perna
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Ly P Vu
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria Themeli
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sonja Kriks
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ruben Hoya-Arias
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Raya Khanin
- Bioinformatics Core, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Todd Hricik
- Human Oncology and Pathogenesis Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jorge Mansilla-Soto
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Ross L Levine
- Human Oncology and Pathogenesis Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lorenz Studer
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michel Sadelain
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen D Nimer
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
34
|
Combining gene mutation with gene expression data improves outcome prediction in myelodysplastic syndromes. Nat Commun 2015; 6:5901. [PMID: 25574665 PMCID: PMC4338540 DOI: 10.1038/ncomms6901] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/18/2014] [Indexed: 12/15/2022] Open
Abstract
Cancer is a genetic disease, but two patients rarely have identical genotypes. Similarly, patients differ in their clinicopathological parameters, but how genotypic and phenotypic heterogeneity are interconnected is not well understood. Here we build statistical models to disentangle the effect of 12 recurrently mutated genes and 4 cytogenetic alterations on gene expression, diagnostic clinical variables and outcome in 124 patients with myelodysplastic syndromes. Overall, one or more genetic lesions correlate with expression levels of ~20% of all genes, explaining 20–65% of observed expression variability. Differential expression patterns vary between mutations and reflect the underlying biology, such as aberrant polycomb repression for ASXL1 and EZH2 mutations or perturbed gene dosage for copy-number changes. In predicting survival, genomic, transcriptomic and diagnostic clinical variables all have utility, with the largest contribution from the transcriptome. Similar observations are made on the TCGA acute myeloid leukaemia cohort, confirming the general trends reported here. The myelodysplastic syndromes (MDS) are a heterogeneous group of chronic blood cancers. Here, the authors analyse genomic and gene expression data from MDS patients to investigate how driver mutations alter gene expression, diagnostic clinical variables and survival.
Collapse
|
35
|
Cell interactions and cell signaling during hematopoietic development. Exp Cell Res 2014; 329:200-6. [DOI: 10.1016/j.yexcr.2014.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/02/2014] [Accepted: 10/05/2014] [Indexed: 12/30/2022]
|
36
|
FGF signalling restricts haematopoietic stem cell specification via modulation of the BMP pathway. Nat Commun 2014; 5:5588. [PMID: 25429520 DOI: 10.1038/ncomms6588] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 10/17/2014] [Indexed: 12/24/2022] Open
Abstract
Haematopoietic stem cells (HSCs) are produced during embryogenesis from the floor of the dorsal aorta. The localization of HSCs is dependent on the presence of instructive signals on the ventral side of the vessel. The nature of the extrinsic molecular signals that control the aortic haematopoietic niche is currently poorly understood. Here we demonstrate a novel requirement for FGF signalling in the specification of aortic haemogenic endothelium. Our results demonstrate that FGF signalling normally acts to repress BMP activity in the subaortic mesenchyme through transcriptional inhibition of bmp4, as well as through activation of two BMP antagonists, noggin2 and gremlin1a. Taken together, these findings demonstrate a key role for FGF signalling in establishment of the developmental HSC niche via its regulation of BMP activity in the subaortic mesenchyme. These results should help inform strategies to recapitulate the development of HSCs in vitro from pluripotent precursors.
Collapse
|
37
|
Inhibition of endothelial ERK signalling by Smad1/5 is essential for haematopoietic stem cell emergence. Nat Commun 2014; 5:3431. [DOI: 10.1038/ncomms4431] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 02/11/2014] [Indexed: 01/02/2023] Open
|
38
|
Endothelial Smad4 restrains the transition to hematopoietic progenitors via suppression of ERK activation. Blood 2014; 123:2161-71. [PMID: 24553180 DOI: 10.1182/blood-2013-09-526053] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In mouse mid-gestational embryos, definitive hematopoietic stem progenitor cells are derived directly from a very small proportion of the arterial endothelium. However, the physiological mechanisms restraining excessive endothelial-hematopoietic transition remain elusive. We show here that genetic deletion of Smad4 from the endothelium stage (using Tie2-Cre), but not from embryonic hematopoietic cells (using Vav-Cre), leads to a strikingly augmented emergence of intra-arterial hematopoietic clusters and an enhanced in vitro generation of hematopoietic progenitors, with no increase in the proliferation and survival of hematopoietic cluster cells. This finding indicates a temporally restricted negative effect of Smad4 on the endothelial to hematopoietic progenitor transition. Furthermore, the absence of endothelial Smad4 causes an increased expression of subaortic bone morphogenetic protein 4 and an activation of aortic extracellular signal-regulated kinase, thereby resulting in the excessive generation of blood cells. Collectively, our data for the first time identify a physiological suppressor that functions specifically during the transition of endothelial cells to hematopoietic progenitors and further suggest that endothelial Smad4 is a crucial modulator of the subaortic microenvironment that controls the hematopoietic fate of the aortic endothelium.
Collapse
|
39
|
Jaffredo T, Lempereur A, Richard C, Bollerot K, Gautier R, Canto PY, Drevon C, Souyri M, Durand C. Dorso-ventral contributions in the formation of the embryonic aorta and the control of aortic hematopoiesis. Blood Cells Mol Dis 2013; 51:232-8. [DOI: 10.1016/j.bcmd.2013.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/29/2013] [Indexed: 01/08/2023]
|
40
|
Beazley KE, Reckard S, Nurminsky D, Lima F, Nurminskaya M. Two sides of MGP null arterial disease: chondrogenic lesions dependent on transglutaminase 2 and elastin fragmentation associated with induction of adipsin. J Biol Chem 2013; 288:31400-8. [PMID: 24036114 PMCID: PMC3829453 DOI: 10.1074/jbc.m113.495556] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/02/2013] [Indexed: 01/04/2023] Open
Abstract
Mutations in matrix Gla protein (MGP) have been correlated with vascular calcification. In the mouse model, MGP null vascular disease presents as calcifying cartilaginous lesions and mineral deposition along elastin lamellae (elastocalcinosis). Here we examined the mechanisms underlying both of these manifestations. Genetic ablation of enzyme transglutaminase 2 (TG2) in Mgp(-/-) mice dramatically reduced the size of cartilaginous lesions in the aortic media, attenuated calcium accrual more than 2-fold, and doubled longevity as compared with control Mgp(-/-) animals. Nonetheless, the Mgp(-/-);Tgm2(-/-) mice still died prematurely as compared with wild-type and retained the elastocalcinosis phenotype. This pathology in Mgp(-/-) animals was developmentally preceded by extensive fragmentation of elastic lamellae and associated with elevated serine elastase activity in aortic tissue and vascular smooth muscle cells. Systematic gene expression analysis followed by an immunoprecipitation study identified adipsin as the major elastase that is induced in the Mgp(-/-) vascular smooth muscle even in the TG2 null background. These results reveal a central role for TG2 in chondrogenic transformation of vascular smooth muscle and implicate adipsin in elastin fragmentation and ensuing elastocalcinosis. The importance of elastin calcification in MGP null vascular disease is highlighted by significant residual vascular calcification and mortality in Mgp(-/-);Tgm2(-/-) mice with reduced cartilaginous lesions. Our studies identify two potential therapeutic targets in vascular calcification associated with MGP dysfunction and emphasize the need for a comprehensive approach to this multifaceted disorder.
Collapse
Affiliation(s)
- Kelly E. Beazley
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Steven Reckard
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Dmitry Nurminsky
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Florence Lima
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Maria Nurminskaya
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| |
Collapse
|
41
|
Transcriptional regulation of haematopoietic stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:187-212. [PMID: 23696358 DOI: 10.1007/978-94-007-6621-1_11] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Haematopoietic stem cells (HSCs) are a rare cell population found in the bone marrow of adult mammals and are responsible for maintaining the entire haematopoietic system. Definitive HSCs are produced from mesoderm during embryonic development, from embryonic day 10 in the mouse. HSCs seed the foetal liver before migrating to the bone marrow around the time of birth. In the adult, HSCs are largely quiescent but have the ability to divide to self-renew and expand, or to proliferate and differentiate into any mature haematopoietic cell type. Both the specification of HSCs during development and their cellular choices once formed are tightly controlled at the level of transcription. Numerous transcriptional regulators of HSC specification, expansion, homeostasis and differentiation have been identified, primarily from analysis of mouse gene knockout experiments and transplantation assays. These include transcription factors, epigenetic modifiers and signalling pathway effectors. This chapter reviews the current knowledge of these HSC transcriptional regulators, predominantly focusing on the transcriptional regulation of mouse HSCs, although transcriptional regulation of human HSCs is also mentioned where relevant. Due to the breadth and maturity of this field, we have prioritised recently identified examples of HSC transcriptional regulators. We go on to highlight additional layers of control that regulate expression and activity of HSC transcriptional regulators and discuss how chromosomal translocations that result in fusion proteins of these HSC transcriptional regulators commonly drive leukaemias through transcriptional dysregulation.
Collapse
|
42
|
Kobayashi K, Ding G, Nishikawa SI, Kataoka H. Role of Etv2-positive cells in the remodeling morphogenesis during vascular development. Genes Cells 2013; 18:704-21. [PMID: 23795570 DOI: 10.1111/gtc.12070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/02/2013] [Indexed: 01/12/2023]
Abstract
Etv2 is a critical determinant for the commitment of endothelial (EC) and hematopoietic (HPC) cells from mesoderm. Etv2 is assumed to be transiently required for EC commitment but dispensable after most ECs differentiate around E9.5. To confirm the time window of Etv2 requirement, Etv2 was ablated at different time points using ROSA26CreER mice. Unexpectedly, Etv2 ablation at E9.5 caused vascular remodeling defects in cranial and yolk sac vasculature. Immunostaining showed that Etv2+/VE-cadherin (VECAD)- cells were present around forming vasculature, mostly co-expressing Flk-1 with a small number of Etv2+/VECAD+ cells, indicating that Etv2+/Flk-1+/VECAD- cells are the major Etv2+ population promoting vascular remodeling around E9.5. Gene expression analysis showed up-regulation of Fgf proteins, Il-6, Glypican-3 and matrix metalloproteases in Etv2+/VEDAC- cells over Etv2-/VECAD+ mature ECs. Blockade of those factors caused reduced EC sprouting in ex vivo explant culture from E9.5 embryos, suggesting the functional significance of environmental factors derived from Etv2+ cells. Altogether, we propose that Etv2+/VEDAC- cells around E9.5-E10.5 provide extracellular factors to complete vascular morphogenesis in addition to becoming differentiated ECs incorporated into vessels. This insight for the new role of Ets protein in perivascular Flk-1+/VECAD-/(Etv2+) cells to induce expression of angiogenic factors may provide another strategy to control angiogenesis.
Collapse
Affiliation(s)
- Kumiko Kobayashi
- Laboratory for Stem Cell Biology, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | | | | | | |
Collapse
|
43
|
Narula J, Williams CJ, Tiwari A, Marks-Bluth J, Pimanda JE, Igoshin OA. Mathematical model of a gene regulatory network reconciles effects of genetic perturbations on hematopoietic stem cell emergence. Dev Biol 2013; 379:258-69. [PMID: 23623899 DOI: 10.1016/j.ydbio.2013.04.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/06/2013] [Accepted: 04/17/2013] [Indexed: 12/01/2022]
Abstract
Interlinked gene regulatory networks (GRNs) are vital for the spatial and temporal control of gene expression during development. The hematopoietic transcription factors (TFs) Scl, Gata2 and Fli1 form one such densely connected GRN which acts as a master regulator of embryonic hematopoiesis. This triad has been shown to direct the specification of the hemogenic endothelium and emergence of hematopoietic stem cells (HSCs) in response to Notch1 and Bmp4-Smad signaling. Here we employ previously published data to construct a mathematical model of this GRN network and use this model to systematically investigate the network dynamical properties. Our model uses a statistical-thermodynamic framework to describe the combinatorial regulation of gene expression and reconciles, mechanistically, several previously published but unexplained results from different genetic perturbation experiments. In particular, our results demonstrate how the interactions of Runx1, an essential hematopoietic TF, with components of the Bmp4 signaling pathway allow it to affect triad activation and acts as a key regulator of HSC emergence. We also explain why heterozygous deletion of this essential TF, Runx1, speeds up the network dynamics leading to accelerated HSC emergence. Taken together our results demonstrate that the triad, a master-level controller of definitive hematopoiesis, is an irreversible bistable switch whose dynamical properties are modulated by Runx1 and components of the Bmp4 signaling pathway.
Collapse
Affiliation(s)
- Jatin Narula
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | | | | | | | | |
Collapse
|
44
|
Zhang Y, Duc ACE, Rao S, Sun XL, Bilbee AN, Rhodes M, Li Q, Kappes DJ, Rhodes J, Wiest DL. Control of hematopoietic stem cell emergence by antagonistic functions of ribosomal protein paralogs. Dev Cell 2013; 24:411-25. [PMID: 23449473 DOI: 10.1016/j.devcel.2013.01.018] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 11/15/2012] [Accepted: 01/20/2013] [Indexed: 01/14/2023]
Abstract
It remains controversial whether the highly homologous ribosomal protein (RP) paralogs found in lower eukaryotes have distinct functions and this has not been explored in vertebrates. Here we demonstrate that despite ubiquitous expression, the RP paralogs, Rpl22 and Rpl22-like1 (Rpl22l1) play essential, distinct, and antagonistic roles in hematopoietic development. Knockdown of Rpl22 in zebrafish embryos selectively blocks the development of T lineage progenitors after they have seeded the thymus. In contrast, knockdown of the Rpl22 paralog, Rpl22l1, impairs the emergence of hematopoietic stem cells (HSC) in the aorta-gonad-mesonephros by abrogating Smad1 expression and the consequent induction of essential transcriptional regulator, Runx1. Indeed, despite the ability of both paralogs to bind smad1 RNA, Rpl22 and Rpl22l1 have opposing effects on Smad1 expression. Accordingly, circumstances that tip the balance of these paralogs in favor of Rpl22 (e.g., Rpl22l1 knockdown or Rpl22 overexpression) result in repression of Smad1 and blockade of HSC emergence.
Collapse
Affiliation(s)
- Yong Zhang
- Blood Cell Development and Cancer Keystone, Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Moignard V, Macaulay IC, Swiers G, Buettner F, Schütte J, Calero-Nieto FJ, Kinston S, Joshi A, Hannah R, Theis FJ, Jacobsen SE, de Bruijn M, Göttgens B. Characterization of transcriptional networks in blood stem and progenitor cells using high-throughput single-cell gene expression analysis. Nat Cell Biol 2013; 15:363-72. [PMID: 23524953 PMCID: PMC3796878 DOI: 10.1038/ncb2709] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 02/08/2013] [Indexed: 12/15/2022]
Abstract
Cellular decision-making is mediated by a complex interplay of external stimuli with the intracellular environment, in particular transcription factor regulatory networks. Here we have determined the expression of a network of 18 key haematopoietic transcription factors in 597 single primary blood stem and progenitor cells isolated from mouse bone marrow. We demonstrate that different stem/progenitor populations are characterized by distinctive transcription factor expression states, and through comprehensive bioinformatic analysis reveal positively and negatively correlated transcription factor pairings, including previously unrecognized relationships between Gata2, Gfi1 and Gfi1b. Validation using transcriptional and transgenic assays confirmed direct regulatory interactions consistent with a regulatory triad in immature blood stem cells, where Gata2 may function to modulate cross-inhibition between Gfi1 and Gfi1b. Single-cell expression profiling therefore identifies network states and allows reconstruction of network hierarchies involved in controlling stem cell fate choices, and provides a blueprint for studying both normal development and human disease.
Collapse
Affiliation(s)
- Victoria Moignard
- University of Cambridge, Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical, Cambridge, CB2 0XY, United Kingdom
| | - Iain C. Macaulay
- Haematopoietic Stem Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom
| | - Gemma Swiers
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom
| | - Florian Buettner
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Ingolstadter Landstraße 1, 85764 Neuherberg, Germany
| | - Judith Schütte
- University of Cambridge, Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical, Cambridge, CB2 0XY, United Kingdom
| | - Fernando J. Calero-Nieto
- University of Cambridge, Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical, Cambridge, CB2 0XY, United Kingdom
| | - Sarah Kinston
- University of Cambridge, Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical, Cambridge, CB2 0XY, United Kingdom
| | - Anagha Joshi
- University of Cambridge, Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical, Cambridge, CB2 0XY, United Kingdom
| | - Rebecca Hannah
- University of Cambridge, Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical, Cambridge, CB2 0XY, United Kingdom
| | - Fabian J. Theis
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Ingolstadter Landstraße 1, 85764 Neuherberg, Germany
| | - Sten Eirik Jacobsen
- Haematopoietic Stem Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom
| | - Marella de Bruijn
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom
| | - Berthold Göttgens
- University of Cambridge, Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute & Cambridge Institute for Medical, Cambridge, CB2 0XY, United Kingdom
| |
Collapse
|
46
|
Rossetti S, Sacchi N. RUNX1: A microRNA hub in normal and malignant hematopoiesis. Int J Mol Sci 2013; 14:1566-88. [PMID: 23344057 PMCID: PMC3565335 DOI: 10.3390/ijms14011566] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 12/31/2012] [Accepted: 01/04/2013] [Indexed: 12/30/2022] Open
Abstract
Hematopoietic development is orchestrated by gene regulatory networks that progressively induce lineage-specific transcriptional programs. To guarantee the appropriate level of complexity, flexibility, and robustness, these networks rely on transcriptional and post-transcriptional circuits involving both transcription factors (TFs) and microRNAs (miRNAs). The focus of this review is on RUNX1 (AML1), a master hematopoietic transcription factor which is at the center of miRNA circuits necessary for both embryonic and post-natal hematopoiesis. Interference with components of these circuits can perturb RUNX1-controlled coding and non-coding transcriptional programs in leukemia.
Collapse
Affiliation(s)
- Stefano Rossetti
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA; E-Mail:
| | - Nicoletta Sacchi
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA; E-Mail:
| |
Collapse
|
47
|
Hussin J, Sinnett D, Casals F, Idaghdour Y, Bruat V, Saillour V, Healy J, Grenier JC, de Malliard T, Busche S, Spinella JF, Larivière M, Gibson G, Andersson A, Holmfeldt L, Ma J, Wei L, Zhang J, Andelfinger G, Downing JR, Mullighan CG, Awadalla P. Rare allelic forms of PRDM9 associated with childhood leukemogenesis. Genome Res 2012; 23:419-30. [PMID: 23222848 PMCID: PMC3589531 DOI: 10.1101/gr.144188.112] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
One of the most rapidly evolving genes in humans, PRDM9, is a key determinant of the distribution of meiotic recombination events. Mutations in this meiotic-specific gene have previously been associated with male infertility in humans and recent studies suggest that PRDM9 may be involved in pathological genomic rearrangements. In studying genomes from families with children affected by B-cell precursor acute lymphoblastic leukemia (B-ALL), we characterized meiotic recombination patterns within a family with two siblings having hyperdiploid childhood B-ALL and observed unusual localization of maternal recombination events. The mother of the family carries a rare PRDM9 allele, potentially explaining the unusual patterns found. From exomes sequenced in 44 additional parents of children affected with B-ALL, we discovered a substantial and significant excess of rare allelic forms of PRDM9. The rare PRDM9 alleles are transmitted to the affected children in half the cases; nonetheless there remains a significant excess of rare alleles among patients relative to controls. We successfully replicated this latter observation in an independent cohort of 50 children with B-ALL, where we found an excess of rare PRDM9 alleles in aneuploid and infant B-ALL patients. PRDM9 variability in humans is thought to influence genomic instability, and these data support a potential role for PRDM9 variation in risk of acquiring aneuploidies or genomic rearrangements associated with childhood leukemogenesis.
Collapse
Affiliation(s)
- Julie Hussin
- Department of Biochemistry, Faculty of Medicine, University of Montreal, Montreal H3C 3J7, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Marks-Bluth J, Pimanda JE. Cell signalling pathways that mediate haematopoietic stem cell specification. Int J Biochem Cell Biol 2012; 44:2175-84. [DOI: 10.1016/j.biocel.2012.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 09/09/2012] [Accepted: 09/10/2012] [Indexed: 01/27/2023]
|
49
|
Kang YJ, Shin JW, Yoon JH, Oh IH, Lee SP, Kim SY, Park SH, Mamura M. Inhibition of erythropoiesis by Smad6 in human cord blood hematopoietic stem cells. Biochem Biophys Res Commun 2012; 423:750-6. [DOI: 10.1016/j.bbrc.2012.06.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 06/08/2012] [Indexed: 11/24/2022]
|
50
|
Ferri-Lagneau KF, Moshal KS, Grimes M, Zahora B, Lv L, Sang S, Leung T. Ginger stimulates hematopoiesis via Bmp pathway in zebrafish. PLoS One 2012; 7:e39327. [PMID: 22761764 PMCID: PMC3382625 DOI: 10.1371/journal.pone.0039327] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 05/18/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Anemia is a hematologic disorder with decreased number of erythrocytes. Erythropoiesis, the process by which red blood cells differentiate, are conserved in humans, mice and zebrafish. The only known agents available to treat pathological anemia are erythropoietin and its biologic derivatives. However, erythropoietin therapy elicits unwanted side-effects, high cost and intravenous or subcutaneous injection, warranting the development of a more cost effective and non-peptide alternative. Ginger (Zingiber officinale) has been widely used in traditional medicine; however, to date there is no scientific research documenting the potential of ginger to stimulate hematopoiesis. METHODOLOGY/PRINCIPAL FINDINGS Here, we utilized gata1:dsRed transgenic zebrafish embryos to investigate the effect of ginger extract on hematopoiesis in vivo and we identified its bioactive component, 10-gingerol. We confirmed that ginger and 10-gingerol promote the expression of gata1 in erythroid cells and increase the expression of hematopoietic progenitor markers cmyb and scl. We also demonstrated that ginger and 10-gingerol can promote the hematopoietic recovery from acute hemolytic anemia in zebrafish, by quantifying the number of circulating erythroid cells in the dorsal aorta using video microscopy. We found that ginger and 10-gingerol treatment during gastrulation results in an increase of bmp2b and bmp7a expression, and their downstream effectors, gata2 and eve1. At later stages ginger and 10-gingerol can induce bmp2b/7a, cmyb, scl and lmo2 expression in the caudal hematopoietic tissue area. We further confirmed that Bmp/Smad pathway mediates this hematopoiesis promoting effect of ginger by using the Bmp-activated Bmp type I receptor kinase inhibitors dorsomorphin, LND193189 and DMH1. CONCLUSIONS/SIGNIFICANCE Our study provides a strong foundation to further evaluate the molecular mechanism of ginger and its bioactive components during hematopoiesis and to investigate their effects in adults. Our results will provide the basis for future research into the effect of ginger during mammalian hematopoiesis to develop novel erythropoiesis promoting agents.
Collapse
Affiliation(s)
- Karine F. Ferri-Lagneau
- The Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, North Carolina, United States of America
| | - Karni S. Moshal
- The Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, North Carolina, United States of America
| | - Matthew Grimes
- The Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, North Carolina, United States of America
| | - Braden Zahora
- The Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, North Carolina, United States of America
| | - Lishuang Lv
- Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis, North Carolina, United States of America
| | - Shengmin Sang
- Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis, North Carolina, United States of America
| | - TinChung Leung
- The Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, North Carolina, United States of America
- Department of Biology, North Carolina Central University, Durham, North Carolina, United States of America
| |
Collapse
|