1
|
Clements WK, Khoury H. The molecular and cellular hematopoietic stem cell specification niche. Exp Hematol 2024; 136:104280. [PMID: 39009276 PMCID: PMC11338702 DOI: 10.1016/j.exphem.2024.104280] [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: 04/24/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
Hematopoietic stem cells (HSCs) are a population of tissue-specific stem cells that reside in the bone marrow of adult mammals, where they self-renew and continuously regenerate the adult hematopoietic lineages over the life of the individual. Prominence as a stem cell model and clinical usefulness have driven interest in understanding the physiologic processes that lead to the specification of HSCs during embryonic development. High-efficiency directed differentiation of HSCs by the instruction of defined progenitor cells using sequentially defined instructive molecules and conditions remains impossible, indicating that comprehensive knowledge of the complete set of precursor intermediate identities and required inductive inputs remains incompletely understood. Recently, interest in the molecular and cellular microenvironment where HSCs are specified from endothelial precursors-the "specification niche"-has increased. Here we review recent progress in understanding these niche spaces across vertebrate phyla, as well as how a better characterization of the origin and molecular phenotypes of the niche cell populations has helped inform and complicate previous understanding of signaling required for HSC emergence and maturation.
Collapse
Affiliation(s)
- Wilson K Clements
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
| | - Hanane Khoury
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
2
|
Liu W, Ding Y, Shen Z, Xu C, Yi W, Wang D, Zhou Y, Zon LI, Liu JX. BF170 hydrochloride enhances the emergence of hematopoietic stem and progenitor cells. Development 2024; 151:dev202476. [PMID: 38940293 DOI: 10.1242/dev.202476] [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: 10/24/2023] [Accepted: 05/14/2024] [Indexed: 06/29/2024]
Abstract
Generation of hematopoietic stem and progenitor cells (HSPCs) ex vivo and in vivo, especially the generation of safe therapeutic HSPCs, still remains inefficient. In this study, we have identified compound BF170 hydrochloride as a previously unreported pro-hematopoiesis molecule, using the differentiation assays of primary zebrafish blastomere cell culture and mouse embryoid bodies (EBs), and we demonstrate that BF170 hydrochloride promoted definitive hematopoiesis in vivo. During zebrafish definitive hematopoiesis, BF170 hydrochloride increases blood flow, expands hemogenic endothelium (HE) cells and promotes HSPC emergence. Mechanistically, the primary cilia-Ca2+-Notch/NO signaling pathway, which is downstream of the blood flow, mediated the effects of BF170 hydrochloride on HSPC induction in vivo. Our findings, for the first time, reveal that BF170 hydrochloride is a compound that enhances HSPC induction and may be applied to the ex vivo expansion of HSPCs.
Collapse
Affiliation(s)
- WenYe Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - YuYan Ding
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zheng Shen
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Cong Xu
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - William Yi
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ding Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yi Zhou
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Leonard I Zon
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute/Children's Hospital, 300 Longwood Avenue, Karp 8, Boston, MA 02115, USA
| | - Jing-Xia Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
3
|
Abstract
Formation of the vasculature is a critical step within the developing embryo and its disruption causes early embryonic lethality. This complex process is driven by a cascade of signaling events that controls differentiation of mesodermal progenitors into primordial endothelial cells and their further specification into distinct subtypes (arterial, venous, hemogenic) that are needed to generate a blood circulatory network. Hemogenic endothelial cells give rise to hematopoietic stem and progenitor cells that generate all blood cells in the body during embryogenesis and postnatally. We focus our discussion on the regulation of endothelial cell differentiation, and subsequent hemogenic specification, and highlight many of the signaling pathways involved in these processes, which are conserved across vertebrates. Gaining a better understanding of the regulation of these processes will yield insights needed to optimize the treatment of vascular and hematopoietic disease and generate human stem cell-derived vascular and hematopoietic cells for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Jordon W Aragon
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
- Departments of Medicine and Genetics, Yale University School of Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut 06520, USA
| |
Collapse
|
4
|
Sun XY, Ju XC, Li Y, Zeng PM, Wu J, Zhou YY, Shen LB, Dong J, Chen Y, Luo ZG. Generation of vascularized brain organoids to study neurovascular interactions. eLife 2022; 11:76707. [PMID: 35506651 PMCID: PMC9246368 DOI: 10.7554/elife.76707] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/01/2022] [Indexed: 12/05/2022] Open
Abstract
Brain organoids have been used to recapitulate the processes of brain development and related diseases. However, the lack of vasculatures, which regulate neurogenesis and brain disorders, limits the utility of brain organoids. In this study, we induced vessel and brain organoids, respectively, and then fused two types of organoids together to obtain vascularized brain organoids. The fused brain organoids were engrafted with robust vascular network-like structures and exhibited increased number of neural progenitors, in line with the possibility that vessels regulate neural development. Fusion organoids also contained functional blood–brain barrier-like structures, as well as microglial cells, a specific population of immune cells in the brain. The incorporated microglia responded actively to immune stimuli to the fused brain organoids and showed ability of engulfing synapses. Thus, the fusion organoids established in this study allow modeling interactions between the neuronal and non-neuronal components in vitro, particularly the vasculature and microglia niche. Understanding how the organs form and how their cells behave is essential to finding the causes and treatment for developmental disorders, as well as understanding certain diseases. However, studying most organs in live animals or humans is technically difficult, expensive and invasive. To address this issue, scientists have developed models called ‘organoids’ that recapitulate the development of organs using stem cells in the lab. These models are easier to study and manipulate than the live organs. Brain organoids have been used to recapitulate brain formation as well as developmental, degenerative and psychiatric brain conditions such as microcephaly, autism and Alzheimer’s disease. However, these brain organoids lack the vasculature (the network of blood vessels) that supplies a live brain with nutrients and regulates its development, and which has important roles in brain disorders. Partly due to this lack of blood vessels, brain organoids also do not develop a blood brain barrier, the structure that prevents certain contents of the blood, including pathogens, toxins and even certain drugs from entering the brain. These characteristics limit the utility of existing brain organoids. To overcome these limitations, Sun, Ju et al. developed brain organoids and blood vessel organoids independently, and then fused them together to obtain vascularized brain organoids. These fusion organoids developed a robust network of blood vessels that was well integrated with the brain cells, and produced more neural cell precursors than brain organoids that had not been fused. This result is consistent with the idea that blood vessels can regulate brain development. Analyzing the fusion organoids revealed that they contain structures similar to the blood-brain barrier, as well as microglial cells (immune cells specific to the brain). When exposed to lipopolysaccharide – a component of the cell wall of certain bacteria – these cells responded by initiating an immune response in the fusion organoids. Notably, the microglial cells were also able to engulf connections between brain cells, a process necessary for the brain to develop the correct structures and work normally. Sun, Ju et al. have developed a new organoid system that will be of broad interest to researchers studying interactions between the brain and the circulatory system. The development of brain-blood-barrier-like structures in the fusion organoids could also facilitate the development of drugs that can cross this barrier, making it easier to treat certain conditions that affect the brain. Refining this model to allow the fusion organoids to grow for longer times in the lab, and adding blood flow to the system will be the next steps to establish this system.
Collapse
Affiliation(s)
- Xin-Yao Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiang-Chun Ju
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Yang Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Peng-Ming Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jian Wu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ying-Ying Zhou
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Li-Bing Shen
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Jian Dong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuejun Chen
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Zhen-Ge Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
5
|
Ghersi JJ, Mahony CB, Bertrand JY. bif1, a new BMP signaling inhibitor, regulates embryonic hematopoiesis in the zebrafish. Development 2019; 146:dev.164103. [PMID: 30837221 DOI: 10.1242/dev.164103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/22/2019] [Indexed: 01/17/2023]
Abstract
Hematopoiesis maintains the entire blood system, and dysregulation of this process can lead to malignancies (leukemia), immunodeficiencies or red blood cell diseases (anemia, polycythemia vera). We took advantage of the zebrafish model that shares most of the genetic program involved in hematopoiesis with mammals to characterize a new gene of unknown function, si:ch73-299h12.2, which is expressed in the erythroid lineage during primitive, definitive and adult hematopoiesis. This gene, required during primitive and definitive erythropoiesis, encodes a C2H2 zinc-finger protein that inhibits BMP signaling. We therefore named this gene blood-inducing factor 1 and BMP inhibitory factor 1 (bif1). We identified a bif1 ortholog in Sinocyclocheilus rhinocerous, another fish, and in the mouse genome. Both genes also inhibit BMP signaling when overexpressed in zebrafish. In conclusion, we have deorphanized a new zebrafish gene of unknown function: bif1 codes for a zinc-finger protein that inhibits BMP signaling and also regulates primitive erythropoiesis and definitive hematopoiesis.
Collapse
Affiliation(s)
- Joey J Ghersi
- University of Geneva, School of Medicine, Department of Pathology and Immunology, CMU, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Christopher B Mahony
- University of Geneva, School of Medicine, Department of Pathology and Immunology, CMU, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Julien Y Bertrand
- University of Geneva, School of Medicine, Department of Pathology and Immunology, CMU, University of Geneva, CH-1211 Geneva 4, Switzerland
| |
Collapse
|
6
|
Fukui H, Miyazaki T, Chow RWY, Ishikawa H, Nakajima H, Vermot J, Mochizuki N. Hippo signaling determines the number of venous pole cells that originate from the anterior lateral plate mesoderm in zebrafish. eLife 2018; 7:29106. [PMID: 29809141 PMCID: PMC5995544 DOI: 10.7554/elife.29106] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 05/26/2018] [Indexed: 12/11/2022] Open
Abstract
The differentiation of the lateral plate mesoderm cells into heart field cells constitutes a critical step in the development of cardiac tissue and the genesis of functional cardiomyocytes. Hippo signaling controls cardiomyocyte proliferation, but the role of Hippo signaling during early cardiogenesis remains unclear. Here, we show that Hippo signaling regulates atrial cell number by specifying the developmental potential of cells within the anterior lateral plate mesoderm (ALPM), which are incorporated into the venous pole of the heart tube and ultimately into the atrium of the heart. We demonstrate that Hippo signaling acts through large tumor suppressor kinase 1/2 to modulate BMP signaling and the expression of hand2, a key transcription factor that is involved in the differentiation of atrial cardiomyocytes. Collectively, these results demonstrate that Hippo signaling defines venous pole cardiomyocyte number by modulating both the number and the identity of the ALPM cells that will populate the atrium of the heart.
Collapse
Affiliation(s)
- Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Takahiro Miyazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Renee Wei-Yan Chow
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Hiroyuki Ishikawa
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,AMED-Core Research for Evolutional Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| |
Collapse
|
7
|
Perens EA, Garavito-Aguilar ZV, Guio-Vega GP, Peña KT, Schindler YL, Yelon D. Hand2 inhibits kidney specification while promoting vein formation within the posterior mesoderm. eLife 2016; 5:19941. [PMID: 27805568 PMCID: PMC5132343 DOI: 10.7554/elife.19941] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/01/2016] [Indexed: 12/29/2022] Open
Abstract
Proper organogenesis depends upon defining the precise dimensions of organ progenitor territories. Kidney progenitors originate within the intermediate mesoderm (IM), but the pathways that set the boundaries of the IM are poorly understood. Here, we show that the bHLH transcription factor Hand2 limits the size of the embryonic kidney by restricting IM dimensions. The IM is expanded in zebrafish hand2 mutants and is diminished when hand2 is overexpressed. Within the posterior mesoderm, hand2 is expressed laterally adjacent to the IM. Venous progenitors arise between these two territories, and hand2 promotes venous development while inhibiting IM formation at this interface. Furthermore, hand2 and the co-expressed zinc-finger transcription factor osr1 have functionally antagonistic influences on kidney development. Together, our data suggest that hand2 functions in opposition to osr1 to balance the formation of kidney and vein progenitors by regulating cell fate decisions at the lateral boundary of the IM. DOI:http://dx.doi.org/10.7554/eLife.19941.001 The human body is made up of many different types of cells, yet they are all descended from one single fertilized egg cell. The process by which cells specialize into different types is complex and has many stages. At each step of the process, the selection of cell types that a cell can eventually become is increasingly restricted. The entire system is controlled by switching different genes on and off in different groups of cells. Balancing the activity of these genes ensures that enough cells of each type are made in order to build a complete and healthy body. Upsetting this balance can result in organs that are too large, too small or even missing altogether. The cells that form the kidneys and bladder originate within a tissue called the intermediate mesoderm. Controlling the size of this tissue is an important part of building working kidneys. Perens et al. studied how genes control the size of the intermediate mesoderm of zebrafish embryos, which is very similar to the intermediate mesoderm of humans. The experiments revealed that a gene called hand2, which is switched on in cells next to the intermediate mesoderm, restricts the size of this tissue in order to determine the proper size of the kidney. Switching off the hand2 gene resulted in zebrafish with abnormally large kidneys. Loss of hand2 also led to the loss of a different type of cell that forms veins. These findings suggest that cells with an active hand2 gene are unable to become intermediate mesoderm cells and instead go on to become part of the veins. These experiments also demonstrated that a gene called osr1 works in opposition to hand2 to determine the right number of cells that are needed to build the kidneys. Further work will reveal how hand2 prevents cells from joining the intermediate mesoderm and how its role is balanced by the activity of osr1. Understanding how the kidneys form could eventually help to diagnose or treat several genetic diseases and may make it possible to grow replacement kidneys from unspecialized cells. DOI:http://dx.doi.org/10.7554/eLife.19941.002
Collapse
Affiliation(s)
- Elliot A Perens
- Division of Biological Sciences, University of California, San Diego, San Diego, United States.,Department of Pediatrics, School of Medicine, University of California, San Diego, San Diego, United States
| | - Zayra V Garavito-Aguilar
- Division of Biological Sciences, University of California, San Diego, San Diego, United States.,Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Gina P Guio-Vega
- Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Karen T Peña
- Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Yocheved L Schindler
- Division of Biological Sciences, University of California, San Diego, San Diego, United States
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, San Diego, United States
| |
Collapse
|
8
|
Crisan M, Solaimani Kartalaei P, Neagu A, Karkanpouna S, Yamada-Inagawa T, Purini C, Vink CS, van der Linden R, van Ijcken W, Chuva de Sousa Lopes SM, Monteiro R, Mummery C, Dzierzak E. BMP and Hedgehog Regulate Distinct AGM Hematopoietic Stem Cells Ex Vivo. Stem Cell Reports 2016; 6:383-95. [PMID: 26923823 PMCID: PMC4788785 DOI: 10.1016/j.stemcr.2016.01.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 11/19/2022] Open
Abstract
Hematopoietic stem cells (HSC), the self-renewing cells of the adult blood differentiation hierarchy, are generated during embryonic stages. The first HSCs are produced in the aorta-gonad-mesonephros (AGM) region of the embryo through endothelial to a hematopoietic transition. BMP4 and Hedgehog affect their production and expansion, but it is unknown whether they act to affect the same HSCs. In this study using the BRE GFP reporter mouse strain that identifies BMP/Smad-activated cells, we find that the AGM harbors two types of adult-repopulating HSCs upon explant culture: One type is BMP-activated and the other is a non-BMP-activated HSC type that is indirectly controlled by Hedgehog signaling through the VEGF pathway. Transcriptomic analyses demonstrate that the two HSC types express distinct but overlapping genetic programs. These results revealing the bifurcation in HSC types at early embryonic stages in the AGM explant model suggest that their development is dependent upon the signaling molecules in the microenvironment. AGM explants contain two HSC types, BMP-activated and non-BMP-activated Non-BMP-activated HSCs are dependent on Hedgehog/VEGF Changes in the microenvironment ex vivo contribute to novel HSC composition
Collapse
Affiliation(s)
- Mihaela Crisan
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; BHF Centre for Cardiovascular Science, Scottish Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Parham Solaimani Kartalaei
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Alex Neagu
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Sofia Karkanpouna
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Tomoko Yamada-Inagawa
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Caterina Purini
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Chris S Vink
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Reinier van der Linden
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Wilfred van Ijcken
- Center for Biomics, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | | | - Rui Monteiro
- Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Elaine Dzierzak
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
9
|
Abstract
While zebrafish have for some time been regarded as a powerful model organism with which to study early events in hematopoiesis, recent evidence suggests that it also ideal for unraveling the molecular requirements for T cell development in the thymus. Like mammals, zebrafish possess an adaptive immune system, comprising B lymphocytes as well as both the γδ and αβ lineages of T cells, which develop in the thymus. Moreover, the molecular processes underlying T cell development in zebrafish appear to be remarkably conserved. Thus, findings in the zebrafish model will be of high relevance to the equivalent processes in mammals. Finally, molecular processes can be interrogated in zebrafish far more rapidly than is possible in mammals because the zebrafish possesses many unique advantages. These unique attributes, and the methods by which they can be exploited to investigate the role of novel genes in T cell development, are described here.
Collapse
Affiliation(s)
- Yong Zhang
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Avenue, R364, Reimann Building, Philadelphia, PA, 19111, USA,
| | | |
Collapse
|
10
|
Xu Q, Cai C, Hu X, Liu Y, Guo Y, Hu P, Chen Z, Peng S, Zhang D, Jiang S, Wu Z, Chan J, Chen L. Evolutionary suppression of erythropoiesis via the modulation of TGF-β signalling in an Antarctic icefish. Mol Ecol 2015; 24:4664-78. [PMID: 26268413 DOI: 10.1111/mec.13344] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 07/17/2015] [Accepted: 08/08/2015] [Indexed: 01/20/2023]
Abstract
The Antarctic icefish, a family (Channichthyidae) of teleosts within the perciform suborder Notothenioidei, are the only known vertebrates without oxygen-transporting haemoglobins and that are largely devoid of circulating erythrocytes. To elucidate the evo-devo mechanisms underpinning the suppressed erythropoiesis in the icefish, we conducted comparative studies on the transcriptomes and microRNAomes of the primary haematopoietic tissues between an icefish (Chionodraco hamatus) and two red-blooded notothenioids (Trematomus bernacchii and Gymnodraco acuticeps). We identified substantial remodelling of the haematopoietic programs in the icefish through which erythropoiesis is selectively suppressed. Experimental verification showed that erythropoietic suppression in the icefish may be attributable to the upregulation of TGF-β signalling, which coincides with reductions in multiple transcription factors essential for erythropoiesis and the upregulation of hundreds of microRNAs, the majority (> 80%) of which potentially target erythropoiesis regulating factors. Of the six microRNAs selected for verification, three miRNAs (miR-152, miR-1388 and miR-16b) demonstrated suppressive functions on GATA1 and ALAS2, which are two factors important for erythroid differentiation, resulting in reduced numbers of erythroids in microinjected zebra fish embryos. Codon substitution analyses of the genes of the TGF-β superfamily revealed signs of positive selection in TGF-β1 and endoglin in the lineages leading to Antarctic notothenioids. Both genes are previously known to function in erythropoietic suppression. These findings implied a general trend of erythropoietic suppression in the cold-adapted notothenioid lineages through evolutionary modulation of the multi-functional TGF-β signalling pathway. This trend is more pronounced in the haemoglobin-less icefish, which may pre-emptively hinder the otherwise defective erythroids from production.
Collapse
Affiliation(s)
- Qianghua Xu
- Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, Ministry of Education, College of Marine Sciences, Shanghai Ocean University, Shanghai, 201306, China.,Collaborative Innovation Center for Distant-water Fisheries, Shanghai, 201306, China
| | - Chang Cai
- Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, Ministry of Education, College of Marine Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Xingxing Hu
- Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, Ministry of Education, College of Marine Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Yun Liu
- Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, Ministry of Education, College of Marine Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Yanan Guo
- Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, Ministry of Education, College of Marine Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Peng Hu
- Key Laboratory of Aquaculture Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Zuozhou Chen
- Key Laboratory of Aquaculture Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Sihua Peng
- Key Laboratory of Aquaculture Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Dongsheng Zhang
- Key Laboratory of Aquaculture Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Shouwen Jiang
- Key Laboratory of Aquaculture Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Zhichao Wu
- Key Laboratory of Aquaculture Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Jiulin Chan
- Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, Ministry of Education, College of Marine Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Liangbiao Chen
- Key Laboratory of Aquaculture Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| |
Collapse
|
11
|
Lee Y, Manegold JE, Kim AD, Pouget C, Stachura DL, Clements WK, Traver D. FGF signalling specifies haematopoietic stem cells through its regulation of somitic Notch signalling. Nat Commun 2014; 5:5583. [PMID: 25428693 PMCID: PMC4271318 DOI: 10.1038/ncomms6583] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 10/16/2014] [Indexed: 01/07/2023] Open
Abstract
Hematopoietic stem cells (HSCs) derive from hemogenic endothelial cells of the primitive dorsal aorta (DA) during vertebrate embryogenesis. The molecular mechanisms governing this unique endothelial to hematopoietic transition remain unclear. Here, we demonstrate a novel requirement for fibroblast growth factor (FGF) signaling in HSC emergence. This requirement is non-cell-autonomous, and acts within the somite to bridge the Wnt and Notch signaling pathways. We previously demonstrated that Wnt16 regulates the somitic expression of two Notch ligands, deltaC (dlc) and deltaD (dld), whose combined function is required for HSC fate. How Wnt16 connects to Notch function has remained an open question. Our current studies demonstrate that FGF signaling, via FGF receptor 4 (Fgfr4), mediates a signal transduction pathway between Wnt16 and Dlc, but not Dld, to regulate HSC specification. Our findings demonstrate that FGF signaling acts as a key molecular relay within the developmental HSC niche to instruct HSC fate.
Collapse
Affiliation(s)
- Yoonsung Lee
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Jennifer E Manegold
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Albert D Kim
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Claire Pouget
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - David L Stachura
- 1] Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA [2] Department of Biological Sciences, California State University, Chico, California 95929, USA
| | - Wilson K Clements
- 1] Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA [2] Department of Hematology, St Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - David Traver
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
12
|
Kim JD, Lee HW, Jin SW. Diversity is in my veins: role of bone morphogenetic protein signaling during venous morphogenesis in zebrafish illustrates the heterogeneity within endothelial cells. Arterioscler Thromb Vasc Biol 2014; 34:1838-45. [PMID: 25060789 DOI: 10.1161/atvbaha.114.303219] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endothelial cells are a highly diverse group of cells which display distinct cellular responses to exogenous stimuli. Although the aptly named vascular endothelial growth factor-A signaling pathway is hailed as the most important signaling input for endothelial cells, additional factors also participate in regulating diverse aspects of endothelial behaviors and functions. Given this heterogeneity, these additional factors seem to play a critical role in creating a custom-tailored environment to regulate behaviors and functions of distinct subgroups of endothelial cells. For instance, molecular cues that modulate morphogenesis of arterial vascular beds can be distinct from those that govern morphogenesis of venous vascular beds. Recently, we have found that bone morphogenetic protein signaling selectively promotes angiogenesis from venous vascular beds without eliciting similar responses from arterial vascular beds in zebrafish, indicating that bone morphogenetic protein signaling functions as a context-dependent regulator during vascular morphogenesis. In this review, we will provide an overview of the molecular mechanisms that underlie proangiogenic effects of bone morphogenetic protein signaling on venous vascular beds in the context of endothelial heterogeneity and suggest a more comprehensive picture of the molecular mechanisms of vascular morphogenesis during development.
Collapse
Affiliation(s)
- Jun-Dae Kim
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.)
| | - Heon-Woo Lee
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.)
| | - Suk-Won Jin
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.).
| |
Collapse
|
13
|
Tsao KC, Tu CF, Lee SJ, Yang RB. Zebrafish scube1 (signal peptide-CUB (complement protein C1r/C1s, Uegf, and Bmp1)-EGF (epidermal growth factor) domain-containing protein 1) is involved in primitive hematopoiesis. J Biol Chem 2012; 288:5017-26. [PMID: 23271740 DOI: 10.1074/jbc.m112.375196] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
scube1 (signal peptide-CUB (complement protein C1r/C1s, Uegf, and Bmp1)-EGF domain-containing protein 1), the founding member of a novel secreted and cell surface SCUBE protein family, is expressed predominantly in various developing tissues in mice. However, its function in primitive hematopoiesis remains unknown. In this study, we identified and characterized zebrafish scube1 and analyzed its function by injecting antisense morpholino-oligonucleotide into embryos. Whole-mount in situ hybridization revealed that zebrafish scube1 mRNA is maternally expressed and widely distributed during early embryonic development. Knockdown of scube1 by morpholino-oligonucleotide down-regulated the expression of marker genes associated with early primitive hematopoietic precursors (scl) and erythroid (gata1 and hbbe1), as well as early (pu.1) and late (mpo and l-plastin) myelomonocytic lineages. However, the expression of an early endothelial marker fli1a and vascular morphogenesis appeared normal in scube1 morphants. Overexpression of bone morphogenetic protein (bmp) rescued the expression of scl in the posterior lateral mesoderm during early primitive hematopoiesis in scube1 morphants. Biochemical and molecular analysis revealed that Scube1 could be a BMP co-receptor to augment BMP signaling. Our results suggest that scube1 is critical for and functions at the top of the regulatory hierarchy of primitive hematopoiesis by modulating BMP activity during zebrafish embryogenesis.
Collapse
Affiliation(s)
- Ku-Chi Tsao
- Institute of Biomedical Sciences, Taiwan International Graduate Program, Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | | | | | | |
Collapse
|
14
|
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]
|
15
|
Belmokhtar K, Bourguignon T, Worou ME, Khamis G, Bonnet P, Domenech J, Eder V. Regeneration of three layers vascular wall by using BMP2-treated MSC involving HIF-1α and Id1 expressions through JAK/STAT pathways. Stem Cell Rev Rep 2012; 7:847-59. [PMID: 21472453 DOI: 10.1007/s12015-011-9254-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Engineering living, multilayered blood vessels to form in vivo arteries is a promising alternative to peripheral artery bypass using acellular grafts restricted by thrombosis and occlusion at long term. Bone Morphogenetic Protein 2 (BMP2) is a growth factor determining in the early vascular embryonic development. The aim of the present study was evaluate the collaborative effect of recombinant human--BMP2 and Bone marrow--Mesenchymal stem cells (BM-MSCs) seeded on vascular patch to regenerate a vascular arterial wall in a rat model. BM-MSCs expressing green fluorescent protein (GFP) seeded on vascular patch were cultured in presence of recombinant human-BMP2 [100 ng/mL] during 1 week before their implantation on the abdominal aorta of Wistar rats. We observed after 2 weeks under physiological arterial flow a regeneration of a three layers adult-like arterial wall with a middle layer expressing smooth muscle proteins and a border layer expressing endothelial marker. In vitro study, using Matrigel assay and co-culture of BM-MSCs with endothelial cells demonstrated that rh-BMP2 promoted tube-like formation even at long term (90 days) allowing the organization of thick rails. We demonstrated using inhibitors and siRNAs that rh-BMP2 enhanced the expression of HIF-1α and Id1 through, at least in part, the stimulation of JAK2/STAT3/STAT5 signaling pathways. Rh-BMP2 by mimicking embryological conditions allowed vascular BM-MSCs differentiation.
Collapse
Affiliation(s)
- Karim Belmokhtar
- Faculty of Medicine, University François Rabelais, Labpart EA 3852, IFR 135 Tours, France.
| | | | | | | | | | | | | |
Collapse
|
16
|
Garriock RJ, Mikawa T. Early arterial differentiation and patterning in the avian embryo model. Semin Cell Dev Biol 2011; 22:985-92. [PMID: 22020129 DOI: 10.1016/j.semcdb.2011.09.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 09/26/2011] [Accepted: 09/29/2011] [Indexed: 01/04/2023]
Abstract
Of the many models to study vascular biology the avian embryo remains an informative and powerful model system that has provided important insights into endothelial cell recruitment, assembly and remodeling during development of the circulatory system. This review highlights several discoveries in the avian system that show how arterial patterning is regulated using the model of dorsal aortae development along the embryo midline during gastrulation and neurulation. These discoveries were made possible through spatially and temporally controlled gain-of-function experiments that provided direct evidence that BMP signaling plays a pivotal role in vascular recruitment, patterning and remodeling and that Notch-signaling recruits vascular precursor cells to the dorsal aortae. Importantly, BMP ligands are broadly expressed throughout embryos but BMP signaling activation region is spatially defined by precisely regulated expression of BMP antagonists. These discoveries provide insight into how signaling, both positive and negative, regulate vascular patterning. This review also illustrates similarities of early arterial patterning along the embryonic midline in amniotes both avian and mammalians including human, evolutionarily specialized from non-amniotes such as fish and frog.
Collapse
|
17
|
Zape JP, Zovein AC. Hemogenic endothelium: origins, regulation, and implications for vascular biology. Semin Cell Dev Biol 2011; 22:1036-47. [PMID: 22001113 DOI: 10.1016/j.semcdb.2011.10.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 10/03/2011] [Accepted: 10/03/2011] [Indexed: 12/31/2022]
Abstract
The study of endothelial development has been intertwined with hematopoiesis since the early 20th century when a bi-potential cell (hemangioblast) was noted to produce both endothelial and hematopoietic cells. Since then, ideas regarding the nature of connection between the vascular and hematopoietic systems have ranged from a tenuous association to direct lineage origination. In this review, historical data that spans hematopoietic development is examined within the context of hemogenic endothelium. Hemogenic endothelium, a specialized endothelial population capable of hematopoiesis, is an emerging theory that has recently gained momentum. Evidence across species and decades are reviewed, as are the possible modulators of the phenomenon, which include pathways that specify definitive hematopoiesis (Runx1), arterial identity (Notch1), as well as physiological and developmental factors.
Collapse
Affiliation(s)
- Joan P Zape
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143, USA
| | | |
Collapse
|
18
|
Blank U, Karlsson S. The role of Smad signaling in hematopoiesis and translational hematology. Leukemia 2011; 25:1379-88. [PMID: 21566654 DOI: 10.1038/leu.2011.95] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hematopoietic stem cells (HSCs) reside in the bone marrow (BM) of adult individuals and function to produce and regenerate the entire blood and immune system over the course of an individual's lifetime. Historically, HSCs are among the most thoroughly characterized tissue-specific stem cells. Despite this, the regulation of fate options, such as self-renewal and differentiation, has remained elusive, partly because of the expansive plethora of factors and signaling cues that govern HSC behavior in vivo. In the BM, HSCs are housed in specialized niches that dovetail the behavior of HSCs with the need of the organism. The Smad-signaling pathway, which operates downstream of the transforming growth factor-β (TGF-β) superfamily of ligands, regulates a diverse set of biological processes, including proliferation, differentiation and apoptosis, in many different organ systems. Much of the function of Smad signaling in hematopoiesis has remained nebulous due to early embryonic lethality of most knockout mouse models. However, recently new data have been uncovered, suggesting that the Smad-signaling circuitry is intimately linked to HSC regulation. In this review, we bring the Smad-signaling pathway into focus, chronicling key concepts and recent advances with respect to TGF-β-superfamily signaling in normal and leukemic hematopoiesis.
Collapse
Affiliation(s)
- U Blank
- Division of Molecular Medicine and Gene Therapy, Laboratory Medicine, Lund Stem Cell Center, Lund University Hospital, Lund, Sweden.
| | | |
Collapse
|
19
|
Laux DW, Febbo JA, Roman BL. Dynamic analysis of BMP-responsive smad activity in live zebrafish embryos. Dev Dyn 2011; 240:682-94. [PMID: 21337466 DOI: 10.1002/dvdy.22558] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2010] [Indexed: 11/06/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are critical players in development and disease, regulating such diverse processes as dorsoventral patterning, palate formation, and ossification. These ligands are classically considered to signal via BMP receptor-specific Smad proteins 1, 5, and 8. To determine the spatiotemporal pattern of Smad1/5/8 activity and thus canonical BMP signaling in the developing zebrafish embryo, we generated a transgenic line expressing EGFP under the control of a BMP-responsive element. EGFP is expressed in many established BMP signaling domains and is responsive to alterations in BMP type I receptor activity and smad1 and smad5 expression. This transgenic Smad1/5/8 reporter line will be useful for determining ligand and receptor requirements for specific domains of BMP activity, as well as for genetic and pharmacological screens aimed at identifying enhancers or suppressors of canonical BMP signaling.
Collapse
|
20
|
Chan J, Mably JD. Dissection of cardiovascular development and disease pathways in zebrafish. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 100:111-53. [PMID: 21377626 DOI: 10.1016/b978-0-12-384878-9.00004-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The use of animal models in medicine has contributed significantly to the development of drug treatments and surgical procedures for the last century, in particular for cardiovascular disease. In order to model human disease in an animal, an appreciation of the strengths and limitations of the system are required to interpret results and design the logical sequence of steps toward clinical translation. As the world's population ages, cardiovascular disease will become even more prominent and further progress will be essential to stave off what seems destined to become a massive public health issue. Future treatments will require the imaginative application of current models as well as the generation of new ones. In this review, we discuss the resources available for modeling cardiovascular disease in zebrafish and the varied attributes of this system. We then discuss current zebrafish disease models and their potential that has yet to be exploited.
Collapse
Affiliation(s)
- Joanne Chan
- Vascular Biology Program, Department of Surgery, Children's Hospital Boston, and Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
21
|
Mueller RL, Huang C, Ho RK. Spatio-temporal regulation of Wnt and retinoic acid signaling by tbx16/spadetail during zebrafish mesoderm differentiation. BMC Genomics 2010; 11:492. [PMID: 20828405 PMCID: PMC2996988 DOI: 10.1186/1471-2164-11-492] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 09/09/2010] [Indexed: 01/02/2023] Open
Abstract
Background A complex network of signaling pathways and transcription factors regulates vertebrate mesoderm development. Zebrafish mutants provide a powerful tool for examining the roles of individual genes in such a network. spadetail (spt) is a mutant with a lesion in tbx16, a T-box transcription factor involved in mesoderm development; the mutant phenotype includes disrupted primitive red blood cell formation as well as disrupted somitogenesis. Despite much recent progress, the downstream targets of tbx16 remain incompletely understood. The current study was carried out to test whether any of the five major signaling pathways are regulated by tbx16 during two specific stages of mesoderm development: primitive red blood cell formation in the intermediate mesoderm and somite formation in the tail paraxial mesoderm. This test was performed using Gene Set Enrichment Analysis, which identifies coordinated changes in expression among a priori sets of genes associated with biological features or processes. Results Our Gene Set Enrichment Analysis results identify Wnt and retinoic acid signaling as likely downstream targets of tbx16 in the developing zebrafish intermediate mesoderm, the site of primitive red blood cell formation. In addition, such results identify retinoic acid signaling as a downstream target of tbx16 in the developing zebrafish posterior somites. Finally, using candidate gene identification and in situ hybridization, we provide expression domain information for 25 additional genes downstream of tbx16 that are outside of both pathways; 23 were previously unknown downstream targets of tbx16, and seven had previously uncharacterized expression in zebrafish. Conclusions Our results suggest that (1) tbx16 regulates Wnt signaling in the developing zebrafish intermediate mesoderm, the site of primitive red blood cell formation, and (2) tbx16 regulates retinoic acid signaling at two distinct embryonic locations and developmental stages, which may imply ongoing spatio-temporal regulation throughout mesoderm development.
Collapse
Affiliation(s)
- Rachel Lockridge Mueller
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
22
|
Canonical BMP signaling is dispensable for hematopoietic stem cell function in both adult and fetal liver hematopoiesis, but essential to preserve colon architecture. Blood 2010; 115:4689-98. [DOI: 10.1182/blood-2009-05-220988] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Abstract
Numerous publications have described the importance of bone morphogenetic protein (BMP) signaling in the specification of hematopoietic tissue in developing embryos. Here we investigate the full role of canonical BMP signaling in both adult and fetal liver hematopoiesis using conditional knockout strategies because conventional disruption of components of the BMP signaling pathway result in early death of the embryo. By targeting both Smad1 and Smad5, we have generated a double-knockout mouse with complete disruption of canonical BMP signaling. Interestingly, concurrent deletion of Smad1 and Smad5 results in death because of extrahematopoietic pathologic changes in the colon. However, Smad1/Smad5-deficient bone marrow cells can compete normally with wild-type cells and display unaffected self-renewal and differentiation capacity when transplanted into lethally irradiated recipients. Moreover, although BMP receptor expression is increased in fetal liver, fetal liver cells deficient in both Smad1 and Smad5 remain competent to long-term reconstitute lethally irradiated recipients in a multilineage manner. In conclusion, canonical BMP signaling is not required to maintain either adult or fetal liver hematopoiesis, despite its crucial role in the initial patterning of hematopoiesis in early embryonic development.
Collapse
|
23
|
Dudley B, Palumbo C, Nalepka J, Molyneaux K. BMP signaling controls formation of a primordial germ cell niche within the early genital ridges. Dev Biol 2010; 343:84-93. [PMID: 20417197 DOI: 10.1016/j.ydbio.2010.04.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 04/13/2010] [Accepted: 04/14/2010] [Indexed: 11/17/2022]
Abstract
Stem cells are necessary to maintain tissue homeostasis and the microenvironment (a.k.a. the niche) surrounding these cells controls their ability to self-renew or differentiate. For many stem cell populations it remains unclear precisely what cells and signals comprise a niche. Here we identify a possible PGC niche in the mouse genital ridges. Conditional ablation of Bmpr1a was used to demonstrate that BMP signaling is required for PGC survival and migration as these cells colonize the genital ridges. Reduced BMP signaling within the genital ridges led to increased somatic cell death within the mesonephric mesenchyme. Loss of these supporting cells correlated with decreased levels of the mesonephric marker, Pax2, as well as a reduction in genes expressed in the coelomic epithelium including the putative PGC chemo-attractants Kitl and Sdf1a. We propose that BMP signaling promotes mesonephric cell survival within the genital ridges and that these cells support correct development of the coelomic epithelium, the target of PGC migration. Loss of BMP signaling leads to the loss of the PGC target resulting in reduced PGC numbers and disrupted PGC migration.
Collapse
Affiliation(s)
- Brian Dudley
- Department of Genetics, Case Western Reserve University, Cleveland, OH, USA
| | | | | | | |
Collapse
|
24
|
Söderberg SS, Karlsson G, Karlsson S. Complex and context dependent regulation of hematopoiesis by TGF-beta superfamily signaling. Ann N Y Acad Sci 2009; 1176:55-69. [PMID: 19796233 DOI: 10.1111/j.1749-6632.2009.04569.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The transforming growth factor (TGF)-beta superfamily of growth factors, including the TGF-betas, activins, and bone morphogenetic proteins (BMPs), provide cells with a broad spectrum of regulatory signals through the intracellular Smad pathway. Since loss-of-function studies of a majority of the TGF-beta superfamily members result in embryonic lethality, much of our current knowledge of the TGF-beta superfamily's role in hematopoiesis is generated from studies performed in vitro, or in very early stages of embryonic development. TGF-beta is well documented as a potent inhibitor of hematopoietic stem cell (HSC) proliferation in vitro, while its role in vivo is largely unknown. BMP signaling is crucial for the initiation of hematopoiesis in the developing embryo, although its role in adult hematopoiesis remains elusive. More recently we and others have used conditional knockout models to unravel the role of several components of TGF-beta family signaling in adult hematopoiesis. Here we review the currently known functions for the major factors of this signaling family in embryonic and adult hematopoietic regulation and discuss the context dependency and complexity that permeate this regulation.
Collapse
Affiliation(s)
- Sofie Singbrant Söderberg
- Department of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, Lund, Sweden
| | | | | |
Collapse
|
25
|
Frost V, Grocott T, Eccles MR, Chantry A. Self-RegulatedPaxGene Expression and Modulation by the TGFβ Superfamily. Crit Rev Biochem Mol Biol 2009; 43:371-91. [DOI: 10.1080/10409230802486208] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
26
|
Soroldoni D, Hogan BM, Oates AC. Simple and efficient transgenesis with meganuclease constructs in zebrafish. Methods Mol Biol 2009; 546:117-30. [PMID: 19378101 DOI: 10.1007/978-1-60327-977-2_8] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the past, microinjection of plasmid DNA into early embryos represented the state of the art to generate transgenic zebrafish. However, this approach suffers significant drawbacks (mosaic distribution of the injected transgene, late transgene integration at high copy numbers, low transgenesis frequency), making the generation of transgenic lines a laborious task. Coinjection of I-SceI meganuclease with a reporter construct flanked by I-SceI sites overcomes these problems by earlier transgene integration into the host genome. Here, we provide an optimized protocol for I-SceI meganuclease-mediated transgenesis in zebrafish. This simple protocol provides a reliable method to transiently test tissue-specific reporter expression of meganuclease constructs in injected embryos (F0). Furthermore, it substantially facilitates the generation of multiple stable transgenic lines increasing transgenesis frequencies up to 45%, compared with 5% without I-SceI. The reliable reporter activity in F0 and the improved transgenesis frequency make this protocol a powerful tool for use in gain- and loss-of-function, cell tracing, and cell labeling experiments.
Collapse
Affiliation(s)
- Daniele Soroldoni
- Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Pfotenhauerstrasse 108, 01307, Dresden, Germany.
| | | | | |
Collapse
|
27
|
McReynolds LJ, Tucker J, Mullins MC, Evans T. Regulation of hematopoiesis by the BMP signaling pathway in adult zebrafish. Exp Hematol 2008; 36:1604-1615. [PMID: 18973974 DOI: 10.1016/j.exphem.2008.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2007] [Revised: 08/12/2008] [Accepted: 08/21/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The zebrafish is an established model system for studying the embryonic emergence of tissues and organs, including the hematopoietic system. We hypothesized that key signaling pathways controlling embryonic hematopoiesis continue to be important in the adult, and we sought to develop approaches to test this in zebrafish, focused on the bone morphogenetic protein (BMP) signaling pathway. Functions for this pathway in adult hematopoiesis have been challenging to probe in other models. MATERIALS AND METHODS Several approaches tested the function of BMP signaling during adult zebrafish hematopoiesis. First, we evaluated steady-state hematopoiesis in adult fish that are heterozygous for mutant alleles of Smad5, or are homozygous for mutant alleles, and rescued to adulthood by injection of RNA encoding Smad5. Second, we tested the relative ability of smad5 mutant fish to recover from hemolytic anemia. Third, we generated a transgenic line that targets the expression of a dominant-negative BMP receptor to adult-stage Gata1+ progenitor cells. RESULTS Adult fish with a strong mutant smad5 allele are anemic at steady state and, in addition, respond to hemolytic anemia with kinetics that are altered compared to wild-type fish. Fish expressing a mutant BMP receptor in early Gata1+ definitive progenitors generate excessive eosinophils. CONCLUSIONS Our study provides proof of principle that regulation of adult hematopoiesis can be studied in zebrafish by altering specific pathways. We show that the BMP signaling pathway is relevant for adult hematopoiesis to maintain steady state erythropoiesis, control the erythropoietic response following stress anemia, and to generate normal numbers of eosinophils.
Collapse
Affiliation(s)
- Lisa J McReynolds
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
28
|
Abstract
The hemangioblast hypothesis was proposed a century ago. The existence of hemangioblasts is now demonstrated in mouse and human embryonic stem cell (ESC)-derived embryoid bodies (EBs), in the mouse and zebrafish gastrula, and in adults. The hemangioblast is believed to derive from mesodermal cells, and is enriched in the Bry+Flk1+ and Flk1+Scl+ cell populations in EBs and in the posterior primitive streak of the mouse gastrula and in the ventral mesoderm of the zebrafish gastrula. However, recent studies suggest that the hemangioblast does not give rise to all endothelial and hematopoietic lineages in mouse and zebrafish embryos. Although several signaling pathways are known to involve the generation of hemangioblasts, it remains largely unknown how the hemangioblast is formed and what are the master genes controlling hemangioblast development. This review will summarize our current knowledge, challenges, and future directions on molecular and developmental aspects of the hemangioblast.
Collapse
Affiliation(s)
- Jing-Wei Xiong
- The Nephrology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 01219, USA.
| |
Collapse
|
29
|
Marschallinger J, Steinbacher P, Haslett JR, Sänger AM, Rescan PY, Stoiber W. Patterns of angiogenic and hematopoietic gene expression during brown trout embryogenesis. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2008; 310:479-91. [DOI: 10.1002/jez.b.21220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
30
|
Mudumana SP, Hentschel D, Liu Y, Vasilyev A, Drummond IA. odd skipped related1 reveals a novel role for endoderm in regulating kidney versus vascular cell fate. Development 2008; 135:3355-67. [PMID: 18787069 DOI: 10.1242/dev.022830] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The kidney and vasculature are intimately linked both functionally and during development, when nephric and blood/vascular progenitor cells occupy adjacent bands of mesoderm in zebrafish and frog embryos. Developmental mechanisms that underlie the differentiation of kidney versus blood/vascular lineages remain unknown. The odd skipped related1 (osr1) gene encodes a zinc-finger transcription factor that is expressed in the germ ring mesendoderm and subsequently in the endoderm and intermediate mesoderm, prior to the expression of definitive kidney or blood/vascular markers. Knockdown of osr1 in zebrafish embryos resulted in a complete, segment-specific loss of anterior kidney progenitors and a compensatory increase in the number of angioblast cells in the same trunk region. Histology revealed a subsequent absence of kidney tubules, an enlarged cardinal vein and expansion of the posterior venous plexus. Altered kidney versus vascular development correlated with expanded endoderm development in osr1 knockdowns. Combined osr1 loss of function and blockade of endoderm development by knockdown of sox32/casanova rescued anterior kidney development. The results indicate that osr1 activity is required to limit endoderm differentiation from mesendoderm; in the absence of osr1, excess endoderm alters mesoderm differentiation, shifting the balance from kidney towards vascular development.
Collapse
Affiliation(s)
- Sudha P Mudumana
- Nephrology Division, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | | | | | | | |
Collapse
|
31
|
Zhang Y, Bai XT, Zhu KY, Jin Y, Deng M, Le HY, Fu YF, Chen Y, Zhu J, Look AT, Kanki J, Chen Z, Chen SJ, Liu TX. In vivo interstitial migration of primitive macrophages mediated by JNK-matrix metalloproteinase 13 signaling in response to acute injury. THE JOURNAL OF IMMUNOLOGY 2008; 181:2155-64. [PMID: 18641354 DOI: 10.4049/jimmunol.181.3.2155] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Interstitial cell migration through extracellular matrix is a hallmark of the inflammation response, tumor invasion, and metastasis. We have established a stable zebrafish transgenic line expressing enhanced GFP under the lysozyme C promoter for visualizing and measuring primitive macrophage migration in vivo. We show that tissue-resident primitive macrophages migrate rapidly through extracellular matrix to the site of acute injury induced by tail transection. Mechanistically, the specific inhibition of JNK, but not p38 and ERK, dramatically abolished the chemotactic migration in a dose-dependent manner, suppressing the trauma-induced recruitment of phosphorylated C-Jun transcription factor to proximal AP-1 sites in the promoter of matrix metalloproteinase 13 (mmp13), a gene specifically expressed in primitive macrophages during embryogenesis and required for the interstitial migration. Furthermore, dexamethasone suppressed the trauma-induced JNK phosphorylation and macrophage migration accompanied by simultaneous up-regulation of mkp-1, a well-known phosphatase capable of inactivating phosphorylated JNK. The results indicate that the JNK-Mmp13 signaling pathway plays an essential role in regulating the innate immune cell migration in response to severe injury in vivo.
Collapse
Affiliation(s)
- Yong Zhang
- Laboratory of Development and Diseases and State Key Laboratory for Medical Genomics and Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Le HY, Zhang Y, Liu H, Ma LH, Jin Y, Huang QH, Chen Y, Deng M, Chen Z, Chen SJ, Liu TX. eena Promotes Myeloid Proliferation through Stimulating ERK1/2 Phosphorylation in Zebrafish. J Biol Chem 2008; 283:17652-61. [DOI: 10.1074/jbc.m710517200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
33
|
Porayette P, Paulson RF. BMP4/Smad5 dependent stress erythropoiesis is required for the expansion of erythroid progenitors during fetal development. Dev Biol 2008; 317:24-35. [PMID: 18374325 PMCID: PMC2409149 DOI: 10.1016/j.ydbio.2008.01.047] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 01/27/2008] [Accepted: 01/29/2008] [Indexed: 11/24/2022]
Abstract
The rapid growth of the embryo places severe demands on the ability of the cardiovascular system to deliver oxygen to cells. To meet this need, erythroid progenitors rapidly expand in the fetal liver microenvironment such that by E14.5, erythropoiesis predominates in the fetal liver. In this report we show that the BMP4/Smad5 dependent stress erythropoiesis pathway plays a key role in the expansion of erythroid progenitors in the fetal liver. These data show that the fetal liver contains two populations of erythroid progenitors. One population resembles the steady state erythroid progenitors found in the adult bone marrow. While the second population exhibits the properties of stress erythroid progenitors found in adult spleen. Here we demonstrate that defects in BMP4/Smad5 signaling preferentially affect the expansion of the stress erythroid progenitors in the fetal liver leading to fetal anemia. These data suggest that steady state erythropoiesis is unable to generate sufficient erythrocytes to maintain the rapid growth of the embryo leading to the induction of the BMP4 dependent stress erythropoiesis pathway. These observations underscore the similarities between fetal erythropoiesis and stress erythropoiesis.
Collapse
Affiliation(s)
- Prashanth Porayette
- The Huck Institutes of the Life Sciences, Molecular Medicine Option at The Pennsylvania State University, University Park, PA 16802
| | - Robert F. Paulson
- The Department of Veterinary and Biomedical Sciences, Molecular Medicine Option at The Pennsylvania State University, University Park, PA 16802
- The Center for Molecular, Immunology and Infectious Disease, Molecular Medicine Option at The Pennsylvania State University, University Park, PA 16802
- The Huck Institutes of the Life Sciences, Molecular Medicine Option at The Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
34
|
Lohmann F, Bieker JJ. Activation of Eklf expression during hematopoiesis by Gata2 and Smad5 prior to erythroid commitment. Development 2008; 135:2071-82. [PMID: 18448565 DOI: 10.1242/dev.018200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The hierarchical progression of stem and progenitor cells to their more-committed progeny is mediated through cell-to-cell signaling pathways and intracellular transcription factor activity. However, the mechanisms that govern the genetic networks underlying lineage fate decisions and differentiation programs remain poorly understood. Here we show how integration of Bmp4 signaling and Gata factor activity controls the progression of hematopoiesis, as exemplified by the regulation of Eklf during establishment of the erythroid lineage. Utilizing transgenic reporter assays in differentiating mouse embryonic stem cells as well as in the murine fetal liver, we demonstrate that Eklf expression is initiated prior to erythroid commitment during hematopoiesis. Applying phylogenetic footprinting and in vivo binding studies in combination with newly developed loss-of-function technology in embryoid bodies, we find that Gata2 and Smad5 cooperate to induce Eklf in a progenitor population, followed by a switch to Gata1-controlled regulation of Eklf transcription upon erythroid commitment. This stage- and lineage-dependent control of Eklf expression defines a novel role for Eklf as a regulator of lineage fate decisions during hematopoiesis.
Collapse
Affiliation(s)
- Felix Lohmann
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, Box 1020, 1 Gustave Levy Place, New York, NY 10029, USA
| | | |
Collapse
|
35
|
Baldessari D, Mione M. How to create the vascular tree? (Latest) help from the zebrafish. Pharmacol Ther 2008; 118:206-30. [PMID: 18439684 DOI: 10.1016/j.pharmthera.2008.02.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 02/19/2008] [Indexed: 12/22/2022]
Abstract
The cardiovascular system provides oxygen, nutrients and hormones to organs, it directs traffic of metabolites and it maintains tissue homeostasis. It is one of the first organs assembled during vertebrate development and it is essential to life from early stages to adult. For these reasons, the process of vessel formation has being studied for more than a century, but it is only in the late eighties that there has been an explosion of research in the field with the employment of various in vitro and in vivo model systems. The zebrafish (Danio rerio) offers several advantages for in vivo studies; it played a fundamental role in new discoveries and helped to refine our knowledge of the vascular system. This review recapitulates the zebrafish data on vasculogenesis and angiogenesis, including the specification of the haemangioblasts from the mesoderm, their migration to form the vascular cord followed by axial vessels specification, the primary and secondary sprouting of intersomitic vessels, the formation of the lumen, the arterial versus venous specification and patterning. To emphasize the strengths of the zebrafish system in the vascular field, we summarize main tools, such as gene expression and mutagenesis screens, knock down technologies, transgenic lines and imaging, which played a major role in the development of the field and allowed significant discoveries, for instance the recent visualization of the lymphatic system in zebrafish. This information contributes to the prospective of drug discovery to cure human diseases linked to angiogenesis, not last tumours.
Collapse
Affiliation(s)
- Danila Baldessari
- IFOM-IEO Campus (FIRC Institute of Molecular Oncology Foundation-European Institute of Oncology), Via Adamello 16, 20139 Milan, Italy.
| | | |
Collapse
|
36
|
Abstract
The use of stem cells in regenerative medicine is a promising approach to the treatment of disease and injury. Natural and synthetic small molecules have been shown to be useful chemical tools for controlling and manipulating the fates of cells. Small molecules can target signaling transduction pathways (for example, tyrosine kinase receptors) and affect DNA replication, cell differentiation, tumor metastasis and apoptosis. Stem cells share many properties with cancer cells and these similarities can provide insights to control and direct cell behavior; small molecules are already standard chemotherapeutics in the treatment of cancer. Libraries of small molecules have been examined for anticancer behavior (especially apoptosis), and, more recently, for stem cell self-renewal and differentiation capabilities in potential approaches to regenerative medicine. Differentiation therapy for cancer is based on the idea that cancer cells are undifferentiated embryonic-like cells and proposes to promote the differentiation and hence block cell proliferation. For example, retinoids have a role in stem cell differentiation to several lineages and have also been used to promote differentiation of acute promyeloic leukemic cells. Small molecules are also important tools for understanding mechanistic and developmental processes. Strategies for generating functional small molecule libraries have been outlined previously. In this review, we will look at several small molecules that have been described in the recent literature as effectors of stem cell self-renewal or differentiation as associated with the Wnt, Hedgehog or NF-kappaB pathways.
Collapse
|
37
|
Dalgin G, Goldman DC, Donley N, Ahmed R, Eide CA, Christian JL. GATA-2 functions downstream of BMPs and CaM KIV in ectodermal cells during primitive hematopoiesis. Dev Biol 2007; 310:454-69. [PMID: 17850784 PMCID: PMC2049090 DOI: 10.1016/j.ydbio.2007.08.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2006] [Revised: 07/26/2007] [Accepted: 08/06/2007] [Indexed: 01/12/2023]
Abstract
In Xenopus, primitive blood originates from the mesoderm, but extrinsic signals from the ectoderm are required during gastrulation to enable these cells to differentiate as erythrocytes. The nature of these signals, and how they are transcriptionally regulated, is not well understood. We have previously shown that bone morphogenetic proteins (BMPs) are required to signal to ectodermal cells to generate secondary non-cell-autonomous signal(s) necessary for primitive erythropoiesis, and that calmodulin-dependent protein kinase IV (CaM KIV) antagonizes BMP signaling. The current studies demonstrate that Gata-2 functions downstream of BMP receptor activation in these same cells, and is a direct target for antagonism by CaM KIV. We show, using loss of function analysis in whole embryos and in explants, that ectodermal Gata-2 is required for primitive erythropoiesis, and that BMP signals cannot rescue blood defects caused by ectoderm removal or loss of ectodermal GATA-2. Furthermore, we provide evidence that acetylation of GATA-2 is required for its function in primitive blood formation in vivo. Our data support a model in which Gata-2 is a transcriptional target downstream of BMPs within ectodermal cells, while activation of the CaM KIV signaling pathway alters GATA-2 function posttranslationally, by inhibiting its acetylation.
Collapse
Affiliation(s)
- Gokhan Dalgin
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Devorah C. Goldman
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Nathan Donley
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Riffat Ahmed
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Christopher A. Eide
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Jan L. Christian
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| |
Collapse
|
38
|
McReynolds LJ, Gupta S, Figueroa ME, Mullins MC, Evans T. Smad1 and Smad5 differentially regulate embryonic hematopoiesis. Blood 2007; 110:3881-90. [PMID: 17761518 PMCID: PMC2200801 DOI: 10.1182/blood-2007-04-085753] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The bone morphogenetic protein (BMP) signaling pathway regulates multiple steps of hematopoiesis, mediated through receptor-regulated Smads, including Smad1 and Smad5. Here, we use loss-of-function approaches in zebrafish to compare the roles of Smad1 and Smad5 during embryonic hematopoiesis. We show that knockdown of Smad1 or Smad5 generates distinct and even opposite hematopoietic phenotypes. Embryos depleted for Smad1 have an increased number of primitive erythrocytes, but fail to produce mature embryonic macrophages. In contrast, Smad5-depleted embryos are defective in primitive erythropoiesis, yet have normal numbers of macrophages. Loss of either Smad1 or Smad5 causes a failure in the generation of definitive hematopoietic progenitors. To investigate the mechanism behind these phenotypes, we used rescue experiments and found that Smad5 is unable to rescue the Smad1 loss-of-function phenotype, indicating that the 2 highly related proteins have inherently distinct activities. Microarray experiments revealed that the 2 proteins redundantly regulate the key initiators of the hemato-vascular program, including scl, lmo2, and gfi1. However, each also regulates a remarkably distinct genetic program, with Smad5 uniquely regulating the BMP signaling pathway itself. Our results suggest that specificity of BMP signaling output, with respect to hematopoiesis, can be explained by differential functions of Smad1 and Smad5.
Collapse
Affiliation(s)
- Lisa J McReynolds
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|
39
|
Jin SW, Herzog W, Santoro MM, Mitchell TS, Frantsve J, Jungblut B, Beis D, Scott IC, D'Amico LA, Ober EA, Verkade H, Field HA, Chi NC, Wehman AM, Baier H, Stainier DYR. A transgene-assisted genetic screen identifies essential regulators of vascular development in vertebrate embryos. Dev Biol 2007; 307:29-42. [PMID: 17531218 PMCID: PMC2695512 DOI: 10.1016/j.ydbio.2007.03.526] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 03/30/2007] [Accepted: 03/30/2007] [Indexed: 11/18/2022]
Abstract
Formation of a functional vasculature during mammalian development is essential for embryonic survival. In addition, imbalance in blood vessel growth contributes to the pathogenesis of numerous disorders. Most of our understanding of vascular development and blood vessel growth comes from investigating the Vegf signaling pathway as well as the recent observation that molecules involved in axon guidance also regulate vascular patterning. In order to take an unbiased, yet focused, approach to identify novel genes regulating vascular development, we performed a three-step ENU mutagenesis screen in zebrafish. We first screened live embryos visually, evaluating blood flow in the main trunk vessels, which form by vasculogenesis, and the intersomitic vessels, which form by angiogenesis. Embryos that displayed reduced or absent circulation were fixed and stained for endogenous alkaline phosphatase activity to reveal blood vessel morphology. All putative mutants were then crossed into the Tg(flk1:EGFP)(s843) transgenic background to facilitate detailed examination of endothelial cells in live and fixed embryos. We screened 4015 genomes and identified 30 mutations affecting various aspects of vascular development. Specifically, we identified 3 genes (or loci) that regulate the specification and/or differentiation of endothelial cells, 8 genes that regulate vascular tube and lumen formation, 8 genes that regulate vascular patterning, and 11 genes that regulate vascular remodeling, integrity and maintenance. Only 4 of these genes had previously been associated with vascular development in zebrafish illustrating the value of this focused screen. The analysis of the newly defined loci should lead to a greater understanding of vascular development and possibly provide new drug targets to treat the numerous pathologies associated with dysregulated blood vessel growth.
Collapse
Affiliation(s)
- Suk-Won Jin
- Department of Biochemistry and Biophysics, Genetics and Human Genetics, and Cardiovascular Research Institute, University of California San Francisco, CA 94158, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pimanda JE, Donaldson IJ, de Bruijn MFTR, Kinston S, Knezevic K, Huckle L, Piltz S, Landry JR, Green AR, Tannahill D, Göttgens B. The SCL transcriptional network and BMP signaling pathway interact to regulate RUNX1 activity. Proc Natl Acad Sci U S A 2007; 104:840-5. [PMID: 17213321 PMCID: PMC1783401 DOI: 10.1073/pnas.0607196104] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Indexed: 11/18/2022] Open
Abstract
Hematopoietic stem cell (HSC) development is regulated by several signaling pathways and a number of key transcription factors, which include Scl/Tal1, Runx1, and members of the Smad family. However, it remains unclear how these various determinants interact. Using a genome-wide computational screen based on the well characterized Scl +19 HSC enhancer, we have identified a related Smad6 enhancer that also targets expression to blood and endothelial cells in transgenic mice. Smad6, Bmp4, and Runx1 transcripts are concentrated along the ventral aspect of the E10.5 dorsal aorta in the aorta-gonad-mesonephros region from which HSCs originate. Moreover, Smad6, an inhibitor of Bmp4 signaling, binds and inhibits Runx1 activity, whereas Smad1, a positive mediator of Bmp4 signaling, transactivates the Runx1 promoter. Taken together, our results integrate three key determinants of HSC development; the Scl transcriptional network, Runx1 activity, and the Bmp4/Smad signaling pathway.
Collapse
Affiliation(s)
- John E. Pimanda
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 2XY, United Kingdom
| | - Ian J. Donaldson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 2XY, United Kingdom
| | - Marella F. T. R. de Bruijn
- Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom; and
| | - Sarah Kinston
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 2XY, United Kingdom
| | - Kathy Knezevic
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 2XY, United Kingdom
| | - Liz Huckle
- The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, United Kingdom
| | - Sandie Piltz
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 2XY, United Kingdom
| | - Josette-Renée Landry
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 2XY, United Kingdom
| | - Anthony R. Green
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 2XY, United Kingdom
| | - David Tannahill
- The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, United Kingdom
| | - Berthold Göttgens
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 2XY, United Kingdom
| |
Collapse
|
41
|
Naito AT, Shiojima I, Akazawa H, Hidaka K, Morisaki T, Kikuchi A, Komuro I. Developmental stage-specific biphasic roles of Wnt/beta-catenin signaling in cardiomyogenesis and hematopoiesis. Proc Natl Acad Sci U S A 2006; 103:19812-7. [PMID: 17170140 PMCID: PMC1750922 DOI: 10.1073/pnas.0605768103] [Citation(s) in RCA: 365] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although Wingless (Wg)/Wnt signaling has been implicated in heart development of multiple organisms, conflicting results have been reported regarding the role of Wnt/beta-catenin pathway in cardiac myogenesis: Wg/armadillo signaling promotes heart development in Drosophila, whereas activation of Wnt/beta-catenin signaling inhibits heart formation in avians and amphibians. Using an in vitro system of mouse ES cell differentiation into cardiomyocytes, we show here that Wnt/beta-catenin signaling exhibits developmental stage-specific, biphasic, and antagonistic effects on cardiomyogenesis and hematopoiesis/vasculogenesis. Activation of the Wnt/beta-catenin pathway in the early phase during embryoid body (EB) formation enhances ES cell differentiation into cardiomyocytes while suppressing the differentiation into hematopoietic and vascular cell lineages. In contrast, activation of Wnt/beta-catenin signaling in the late phase after EB formation inhibits cardiomyocyte differentiation and enhances the expression of hematopoietic/vascular marker genes through suppression of bone morphogenetic protein signaling. Thus, Wnt/beta-catenin signaling exhibits biphasic and antagonistic effects on cardiomyogenesis and hematopoiesis/vasculogenesis, depending on the stage of development.
Collapse
Affiliation(s)
- Atsuhiko T. Naito
- *Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Ichiro Shiojima
- *Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Hiroshi Akazawa
- *Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Kyoko Hidaka
- Department of Bioscience, National Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan; and
| | - Takayuki Morisaki
- Department of Bioscience, National Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan; and
| | - Akira Kikuchi
- Department of Biochemistry, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Issei Komuro
- *Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
42
|
Little SC, Mullins MC. Extracellular modulation of BMP activity in patterning the dorsoventral axis. ACTA ACUST UNITED AC 2006; 78:224-42. [PMID: 17061292 DOI: 10.1002/bdrc.20079] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Signaling via bone morphogenetic proteins (BMPs) regulates a vast array of diverse biological processes in the developing embryo and in postembryonic life. Many insights into BMP signaling derive from studies of the BMP signaling gradients that pattern cell fates along the embryonic dorsal-ventral (DV) axis of both vertebrates and invertebrates. This review examines recent developments in the field of DV patterning by BMP signaling, focusing on extracellular modulation as a key mechanism in the formation of BMP signaling gradients in Drosophila, Xenopus, and zebrafish.
Collapse
Affiliation(s)
- Shawn C Little
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6058, USA
| | | |
Collapse
|