Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Cell Rep ; 39(11): 110957, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35705037

ABSTRACT

Hematopoietic stem cells (HSCs) express a large variety of cell surface receptors that are associated with acquisition of self-renewal and multipotent properties. Correct expression of these receptors depends on a delicate balance between cell surface trafficking, recycling, and degradation and is controlled by the microtubule network and Golgi apparatus, whose roles have hardly been explored during embryonic/fetal hematopoiesis. Here we show that, in the absence of CLASP2, a microtubule-associated protein, the overall production of HSCs is reduced, and the produced HSCs fail to self-renew and maintain their stemness throughout mouse and zebrafish development. This phenotype can be attributed to decreased cell surface expression of the hematopoietic receptor c-Kit, which originates from increased lysosomal degradation in combination with a reduction in trafficking to the plasma membrane. A dysfunctional Golgi apparatus in CLASP2-deficient HSCs seems to be the underlying cause of the c-Kit expression and signaling imbalance.


Subject(s)
Hematopoietic Stem Cells , Zebrafish , Animals , Mice , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
2.
Front Immunol ; 12: 790379, 2021.
Article in English | MEDLINE | ID: mdl-34899758

ABSTRACT

The journey of a hematopoietic stem cell (HSC) involves the passage through successive anatomical sites where HSCs are in direct contact with their surrounding microenvironment, also known as niche. These spatial and temporal cellular interactions throughout development are required for the acquisition of stem cell properties, and for maintaining the HSC pool through balancing self-renewal, quiescence and lineage commitment. Understanding the context and consequences of these interactions will be imperative for our understanding of HSC biology and will lead to the improvement of in vitro production of HSCs for clinical purposes. The aorta-gonad-mesonephros (AGM) region is in this light of particular interest since this is the cradle of HSC emergence during the embryonic development of all vertebrate species. In this review, we will focus on the developmental origin of HSCs and will discuss the novel technological approaches and recent progress made to identify the cellular composition of the HSC supportive niche and the underlying molecular events occurring in the AGM region.


Subject(s)
Genomics/trends , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Single-Cell Analysis/trends , Stem Cell Niche , Animals , Aorta/embryology , Cell Culture Techniques/trends , Cell Lineage , Cells, Cultured , Diffusion of Innovation , Gene Expression Profiling/trends , Gene Expression Regulation, Developmental , Gonads/embryology , Humans , Mesonephros/embryology , Phenotype , Proteomics/trends , Signal Transduction , Transcriptome
4.
Nat Commun ; 12(1): 3851, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34158501

ABSTRACT

Positional information driving limb muscle patterning is contained in connective tissue fibroblasts but not in myogenic cells. Limb muscles originate from somites, while connective tissues originate from lateral plate mesoderm. With cell and genetic lineage tracing we challenge this model and identify an unexpected contribution of lateral plate-derived fibroblasts to the myogenic lineage, preferentially at the myotendinous junction. Analysis of single-cell RNA-sequencing data from whole limbs at successive developmental stages identifies a population displaying a dual muscle and connective tissue signature. BMP signalling is active in this dual population and at the tendon/muscle interface. In vivo and in vitro gain- and loss-of-function experiments show that BMP signalling regulates a fibroblast-to-myoblast conversion. These results suggest a scenario in which BMP signalling converts a subset of lateral plate mesoderm-derived cells to a myogenic fate in order to create a boundary of fibroblast-derived myonuclei at the myotendinous junction that controls limb muscle patterning.


Subject(s)
Body Patterning/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Muscle, Skeletal/metabolism , Somites/metabolism , Animals , Cell Lineage/genetics , Cells, Cultured , Chick Embryo , Extremities/embryology , Fibroblasts/cytology , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Muscle Development/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Reverse Transcriptase Polymerase Chain Reaction , Somites/cytology , Somites/embryology
5.
Nat Cell Biol ; 23(4): 322-329, 2021 04.
Article in English | MEDLINE | ID: mdl-33837285

ABSTRACT

De novo blood vessel formation occurs through coalescence of endothelial cells (ECs) into a cord-like structure, followed by lumenization either through cell-1-3 or cord-hollowing4-7. Vessels generated in this manner are restricted in diameter to one or two ECs, and these models fail to explain how vasculogenesis can form large-diameter vessels. Here, we describe a model for large vessel formation that does not require a cord-like structure or a hollowing step. In this model, ECs coalesce into a network of struts in the future lumen of the vessel, a process dependent upon bone morphogenetic protein signalling. The vessel wall forms around this network and consists initially of only a few patches of ECs. To withstand external forces and to maintain the shape of the vessel, strut formation traps erythrocytes into compartments to form a rigid structure. Struts gradually prune and ECs from struts migrate into and become part of the vessel wall. Experimental severing of struts resulted in vessel collapse, disturbed blood flow and remodelling defects, demonstrating that struts enable the patency of large vessels during their formation.


Subject(s)
Blood Vessels/growth & development , Endothelial Cells/physiology , Morphogenesis/genetics , Neovascularization, Physiologic/genetics , Blood Vessels/metabolism , Endothelial Cells/metabolism , Erythrocytes/metabolism , Erythrocytes/pathology , Humans
6.
Blood ; 136(7): 831-844, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32457985

ABSTRACT

The defined location of a stem cell within a niche regulates its fate, behavior, and molecular identity via a complex extrinsic regulation that is far from being fully elucidated. To explore the molecular characteristics and key components of the aortic microenvironment, where the first hematopoietic stem cells are generated during development, we performed genome-wide RNA tomography sequencing on zebrafish, chicken, mouse, and human embryos. The resulting anterior-posterior and dorsal-ventral transcriptional maps provided a powerful resource for exploring genes and regulatory pathways active in the aortic microenvironment. By performing interspecies comparative RNA sequencing analyses and functional assays, we explored the complexity of the aortic microenvironment landscape and the fine-tuning of various factors interacting to control hematopoietic stem cell generation, both in time and space in vivo, including the ligand-receptor couple ADM-RAMP2 and SVEP1. Understanding the regulatory function of the local environment will pave the way for improved stem cell production in vitro and clinical cell therapy.


Subject(s)
Aorta/embryology , Hematopoietic Stem Cells/cytology , RNA/analysis , Stem Cell Niche/genetics , Tomography , Animals , Animals, Genetically Modified , Aorta/cytology , Cell Tracking/methods , Chick Embryo , Embryo, Mammalian , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mice , RNA/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis , Species Specificity , Tomography/methods , Tomography/veterinary , Zebrafish/embryology , Zebrafish/genetics
8.
Nat Cell Biol ; 21(11): 1334-1345, 2019 11.
Article in English | MEDLINE | ID: mdl-31685991

ABSTRACT

It is well established that haematopoietic stem and progenitor cells (HSPCs) are generated from a transient subset of specialized endothelial cells termed haemogenic, present in the yolk sac, placenta and aorta, through an endothelial-to-haematopoietic transition (EHT). HSPC generation via EHT is thought to be restricted to the early stages of development. By using experimental embryology and genetic approaches in birds and mice, respectively, we document here the discovery of a bone marrow haemogenic endothelium in the late fetus/young adult. These cells are capable of de novo producing a cohort of HSPCs in situ that harbour a very specific molecular signature close to that of aortic endothelial cells undergoing EHT or their immediate progenies, i.e., recently emerged HSPCs. Taken together, our results reveal that HSPCs can be generated de novo past embryonic stages. Understanding the molecular events controlling this production will be critical for devising innovative therapies.


Subject(s)
Bone Marrow Cells/metabolism , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Hemangioblasts/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Animals, Genetically Modified , Aorta/cytology , Aorta/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Chickens , Embryo, Mammalian , Embryo, Nonmammalian , Female , Fetus , Gene Expression Profiling , Gene Regulatory Networks , Hemangioblasts/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Heterozygote , Homozygote , Male , Mice , Pregnancy , Yolk Sac/cytology , Yolk Sac/growth & development , Yolk Sac/metabolism
9.
Adv Healthc Mater ; 8(10): e1801444, 2019 05.
Article in English | MEDLINE | ID: mdl-30941927

ABSTRACT

Niches in the bone marrow regulate hematopoietic stem and progenitor cell (HSPC) fate and behavior through cell-cell interactions and soluble factor secretion. The niche-HSPC crosstalk is a very complex process not completely elucidated yet. To aid further investigation of this crosstalk, a functional in vitro 3D model that closely represents the main supportive compartments of the bone marrow is developed. Different combinations of human stromal cells and hydrogels are tested for their potential to maintain CD34+ HSPCs. Cell viability, clonogenic hematopoietic potential, and surface marker expression are assessed over time. Optimal HSPC support is obtained in presence of adipogenic and osteogenic cells, together with progenitor derived endothelial cells. When cultured in a bioactive hydrogel, the supportive cells self-assemble into a hypoxic stromal network, stimulating CD34+ CD38+ cell formation, while maintaining the pool of CD34+ 38- HSPCs. HSPC clusters colocalize with the stromal networks, in close proximity to sinusoidal clusters of CD31+ endothelial cells. Importantly, the primary in vitro niche model supports HSPCs with no cytokine addition. Overall, the engineered primary 3D bone marrow environment provides an easy and reliable model to further investigate interactions between HSPCs and their endosteal and perivascular niches, in the context of normal hematopoiesis or blood-related diseases.


Subject(s)
Hematopoietic Stem Cells/cytology , Hydrogels/chemistry , Adipogenesis/drug effects , Alginates/chemistry , Antigens, CD34/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Collagen/chemistry , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Hydrogels/pharmacology , Laminin/chemistry , Osteogenesis/drug effects , Proteoglycans/chemistry , Stem Cell Niche
10.
Cell Rep ; 24(1): 130-141, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29972775

ABSTRACT

Microglia, the tissue-resident macrophages of the CNS, represent major targets for therapeutic intervention in a wide variety of neurological disorders. Efficient reprogramming protocols to generate microglia-like cells in vitro using patient-derived induced pluripotent stem cells will, however, require a precise understanding of the cellular and molecular events that instruct microglial cell fates. This remains a challenge since the developmental origin of microglia during embryogenesis is controversial. Here, using genetic tracing in zebrafish, we uncover primitive macrophages as the unique source of embryonic microglia. We also demonstrate that this initial population is transient, with primitive microglia later replaced by definitive microglia that persist throughout adulthood. The adult wave originates from cmyb-dependent hematopoietic stem cells. Collectively, our work challenges the prevailing model establishing erythro-myeloid progenitors as the sole and direct microglial precursor and provides further support for the existence of multiple waves of microglia, which originate from distinct hematopoietic precursors.


Subject(s)
Embryo, Nonmammalian/cytology , Macrophages/cytology , Microglia/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Embryonic Development , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Kinetics , Macrophages/metabolism
11.
Oncoimmunology ; 7(6): e1434465, 2018.
Article in English | MEDLINE | ID: mdl-29872571

ABSTRACT

Bone marrow niches support multiple myeloma, providing signals and cell-cell interactions essential for disease progression. A 3D bone marrow niche model was developed, in which supportive multipotent mesenchymal stromal cells and their osteogenic derivatives were co-cultured with endothelial progenitor cells. These co-cultured cells formed networks within the 3D culture, facilitating the survival and proliferation of primary CD138+ myeloma cells for up to 28 days. During this culture, no genetic drift was observed within the genomic profile of the primary myeloma cells, indicating a stable outgrowth of the cultured CD138+ population. The 3D bone marrow niche model enabled testing of a novel class of engineered immune cells, so called TEGs (αßT cells engineered to express a defined γδTCR) on primary myeloma cells. TEGs were engineered and tested from both healthy donors and myeloma patients. The added TEGs were capable of migrating through the 3D culture, exerting a killing response towards the primary myeloma cells in 6 out of 8 donor samples after both 24 and 48 hours. Such a killing response was not observed when adding mock transduced T cells. No differences were observed comparing allogeneic and autologous therapy. The supporting stromal microenvironment was unaffected in all conditions after 48 hours. When adding TEG therapy, the 3D model surpassed 2D models in many aspects by enabling analyses of specific homing, and both on- and off-target effects, preparing the ground for the clinical testing of TEGs. The model allows studying novel immunotherapies, therapy resistance mechanisms and possible side-effects for this incurable disease.

12.
Nat Commun ; 9(1): 2517, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29955049

ABSTRACT

Haematopoietic stem cells (HSCs) are generated from haemogenic endothelial (HE) cells via the formation of intra-aortic haematopoietic clusters (IAHCs) in vertebrate embryos. The molecular events controlling endothelial specification, endothelial-to-haematopoietic transition (EHT) and IAHC formation, as it occurs in vivo inside the aorta, are still poorly understood. To gain insight in these processes, we performed single-cell RNA-sequencing of non-HE cells, HE cells, cells undergoing EHT, IAHC cells, and whole IAHCs isolated from mouse embryo aortas. Our analysis identified the genes and transcription factor networks activated during the endothelial-to-haematopoietic switch and IAHC cell maturation toward an HSC fate. Our study provides an unprecedented complete resource to study in depth HSC generation in vivo. It will pave the way for improving HSC production in vitro to address the growing need for tailor-made HSCs to treat patients with blood-related disorders.


Subject(s)
Aorta/metabolism , Cell Lineage , Gene Expression Regulation, Developmental , Hemangioblasts/metabolism , Hematopoietic Stem Cells/metabolism , Transcriptome , Animals , Aorta/cytology , Aorta/growth & development , Cell Differentiation , Embryo, Mammalian , Female , Gene Ontology , Gene Regulatory Networks , Hemangioblasts/cytology , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL , Molecular Sequence Annotation , Single-Cell Analysis
13.
Tissue Eng Part C Methods ; 24(5): 300-312, 2018 05.
Article in English | MEDLINE | ID: mdl-29652626

ABSTRACT

The bone marrow microenvironment is the preferred location of multiple myeloma, supporting tumor growth and development. It is composed of a collection of interacting subniches, including the endosteal and perivascular niche. Current in vitro models mimic either of these subniches. By developing a model combining both niches, this study aims to further enhance the ability to culture primary myeloma cells in vitro. Also, the dependency of myeloma cells on each niche was studied. A 3D bone marrow model containing two subniches was created using 3D bioprinting technology. We used a bioprintable pasty calcium phosphate cement (CPC) scaffold with seeded osteogenic multipotent mesenchymal stromal cells (O-MSCs) to model the endosteal niche, and Matrigel containing both endothelial progenitor cells (EPCs) and MSCs to model the perivascular niche. Within the model containing one or both of the niches, primary CD138+ myeloma cells were cultured and analyzed for both survival and proliferation. The 3D bone marrow model with combined subniches significantly increasing the proliferation of CD138+ myeloma cells compared to both environments separately. The developed model showed an essential role of the perivascular niche over the endosteal niche in supporting myeloma cells. The developed model can be used to study the expansion of primary myeloma cells and their interactions with varying bone marrow subniches.


Subject(s)
Bone Marrow/blood supply , Cellular Microenvironment , Models, Biological , Multiple Myeloma/pathology , Bone Cements/pharmacology , Calcium Phosphates/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Humans , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Tissue Scaffolds/chemistry
15.
Dev Biol ; 428(2): 318-327, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28728681

ABSTRACT

Hematopoietic stem cells (HSCs) are at the origin of adult hematopoiesis, providing an organism with all blood cell types needed throughout life. During embryonic development a first wave of hematopoiesis (independent of HSCs) allows the survival and growth of the embryo until birth. A second wave of hematopoiesis that will last into adulthood depends on the production of HSCs that begins at mid-gestation in large arteries such as the aorta. HSC production occurs through a hemogenic endothelial to hematopoietic transition (EHT) process and the formation of hematopoietic clusters in most vertebrate species. Advances in understanding EHT, cluster formation and HSC production were triggered by combined progresses made in the development of in vivo assays, microscopy, imaging and fluorescence tools. Here, we review the current knowledge on developmental hematopoiesis with a focus on the first step of HSC production in the aorta and how microscopic approaches have contributed to a better understanding of the vital process of blood cell formation.


Subject(s)
Embryonic Development/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Animals , Developmental Biology/history , History, 19th Century , History, 20th Century , History, 21st Century , History, Ancient , History, Medieval , Humans , Microscopy/history , Stem Cell Research/history
16.
Development ; 144(13): 2352-2363, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28526756

ABSTRACT

Hematopoietic stem cells (HSCs), which are responsible for blood cell production, are generated during embryonic development. Human and chicken embryos share features that position the chicken as a reliable and accessible alternative model to study developmental hematopoiesis. However, the existence of HSCs has never been formally proven in chicken embryos. Here, we have established a complete cartography and quantification of hematopoietic cells in the aorta during development. We demonstrate the existence of bona fide HSCs, originating from the chicken embryo aorta (and not the yolk sac, allantois or head), through an in vivo transplantation assay. Embryos transplanted in ovo with GFP embryonic tissues on the chorio-allantoic membrane provided multilineage reconstitution in adulthood. Historically, most breakthrough discoveries in the field of developmental hematopoiesis were first made in birds and later extended to mammals. Our study sheds new light on the avian model as a valuable system to study HSC production and regulation in vivo.


Subject(s)
Cell Lineage , Chickens/metabolism , Hematopoietic Stem Cells/cytology , Animals , Aorta/cytology , Aorta/embryology , B-Lymphocytes/cytology , Cell Proliferation , Cell Survival , Chick Embryo , Chorioallantoic Membrane/transplantation , Embryonic Development , Green Fluorescent Proteins/metabolism , Hematopoiesis , Yolk Sac/embryology
18.
Nat Cell Biol ; 18(1): 21-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26619147

ABSTRACT

In vertebrates, the first haematopoietic stem cells (HSCs) with multi-lineage and long-term repopulating potential arise in the AGM (aorta-gonad-mesonephros) region. These HSCs are generated from a rare and transient subset of endothelial cells, called haemogenic endothelium (HE), through an endothelial-to-haematopoietic transition (EHT). Here, we establish the absolute requirement of the transcriptional repressors GFI1 and GFI1B (growth factor independence 1 and 1B) in this unique trans-differentiation process. We first demonstrate that Gfi1 expression specifically defines the rare population of HE that generates emerging HSCs. We further establish that in the absence of GFI1 proteins, HSCs and haematopoietic progenitor cells are not produced in the AGM, revealing the critical requirement for GFI1 proteins in intra-embryonic EHT. Finally, we demonstrate that GFI1 proteins recruit the chromatin-modifying protein LSD1, a member of the CoREST repressive complex, to epigenetically silence the endothelial program in HE and allow the emergence of blood cells.


Subject(s)
DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Hemangioblasts/metabolism , Hematopoietic Stem Cells/metabolism , Histone Demethylases/metabolism , Transcription Factors/metabolism , Animals , Aorta/cytology , Aorta/embryology , Cell Differentiation/physiology , Embryo, Mammalian/cytology , Hemangioblasts/cytology , Hematopoietic Stem Cells/cytology , Mice
19.
Cardiovasc Res ; 107(3): 352-63, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25987546

ABSTRACT

Fetal haematopoiesis is a highly regulated process in terms of time and location. It is characterized by the emergence of specific cell populations at different extra- and intraembryonic anatomical sites. Trafficking of haematopoietic stem cells (HSCs) between these supportive niches is regulated by a set of molecules, i.e. integrins and chemokine receptors, which are also described for the recruitment of differentiated innate immune cells. In this review, an overview will be given on fetal haematopoiesis as well as trafficking of HSCs during fetal life. In addition, we will focus on the appearance of the first differentiated neutrophils and monocytes in the fetal circulation and describe how they acquire the ability to roll, adhere, and transmigrate into inflamed fetal tissue. Furthermore, we will discuss other effector functions of innate immune cells evolving during fetal ontogeny.


Subject(s)
Fetal Development/immunology , Hematopoiesis , Hematopoietic Stem Cells/physiology , Monocytes/physiology , Neutrophils/physiology , Animals , Humans , Myeloid Cells/physiology
20.
Blood ; 125(3): 465-9, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25301706

ABSTRACT

Clusters of cells attached to the endothelium of the main embryonic arteries were first observed a century ago. Present in most vertebrate species, such clusters, or intraaortic hematopoietic clusters (IAHCs), derive from specialized hemogenic endothelial cells and contain the first few hematopoietic stem cells (HSCs) generated during embryonic development. However, some discrepancies remained concerning the spatio-temporal appearance and the numbers of IAHCs and HSCs. Therefore, the exact cell composition and function of IAHCs remain unclear to date. We show here that IAHCs contain pre-HSCs (or HSC precursors) that can mature into HSCs in vivo (as shown by the successful long-term multilineage reconstitution of primary neonates and secondary adult recipients). Such IAHC pre-HSCs could contribute to the HSC pool increase observed at midgestation. The novel insights in pre-HSC to HSC transition represent an important step toward generating transplantable HSCs in vitro that are needed for autologous HSC transplantation therapies.


Subject(s)
Aorta/embryology , Cell Differentiation , Hematopoietic Stem Cells/cytology , Animals , Female , Mice , Organ Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...