Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Development ; 150(23)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37840454

ABSTRACT

The emergence of definitive human haematopoietic stem cells (HSCs) from Carnegie Stage (CS) 14 to CS17 in the aorta-gonad-mesonephros (AGM) region is a tightly regulated process. Previously, we conducted spatial transcriptomic analysis of the human AGM region at the end of this period (CS16/CS17) and identified secreted factors involved in HSC development. Here, we extend our analysis to investigate the progression of dorso-ventral polarised signalling around the dorsal aorta over the entire period of HSC emergence. Our results reveal a dramatic increase in ventral signalling complexity from the CS13-CS14 transition, coinciding with the first appearance of definitive HSCs. We further observe stage-specific changes in signalling up to CS17, which may underpin the step-wise maturation of HSCs described in the mouse model. The data-rich resource is also presented in an online interface enabling in silico analysis of molecular interactions between spatially defined domains of the AGM region. This resource will be of particular interest for researchers studying mechanisms underlying human HSC development as well as those developing in vitro methods for the generation of clinically relevant HSCs from pluripotent stem cells.


Subject(s)
Hematopoietic Stem Cells , Signal Transduction , Mice , Animals , Humans , Signal Transduction/genetics , Cell Communication , Gene Expression Profiling , Aorta , Mesonephros , Gonads , Hematopoiesis/genetics
2.
Sci Transl Med ; 15(698): eabn0736, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37256934

ABSTRACT

Progressive fibrosis is a feature of aging and chronic tissue injury in multiple organs, including the kidney and heart. Glioma-associated oncogene 1 expressing (Gli1+) cells are a major source of activated fibroblasts in multiple organs, but the links between injury, inflammation, and Gli1+ cell expansion and tissue fibrosis remain incompletely understood. We demonstrated that leukocyte-derived tumor necrosis factor (TNF) promoted Gli1+ cell proliferation and cardiorenal fibrosis through induction and release of Indian Hedgehog (IHH) from renal epithelial cells. Using single-cell-resolution transcriptomic analysis, we identified an "inflammatory" proximal tubular epithelial (iPT) population contributing to TNF- and nuclear factor κB (NF-κB)-induced IHH production in vivo. TNF-induced Ubiquitin D (Ubd) expression was observed in human proximal tubular cells in vitro and during murine and human renal disease and aging. Studies using pharmacological and conditional genetic ablation of TNF-induced IHH signaling revealed that IHH activated canonical Hedgehog signaling in Gli1+ cells, which led to their activation, proliferation, and fibrosis within the injured and aging kidney and heart. These changes were inhibited in mice by Ihh deletion in Pax8-expressing cells or by pharmacological blockade of TNF, NF-κB, or Gli1 signaling. Increased amounts of circulating IHH were associated with loss of renal function and higher rates of cardiovascular disease in patients with chronic kidney disease. Thus, IHH connects leukocyte activation to Gli1+ cell expansion and represents a potential target for therapies to inhibit inflammation-induced fibrosis.


Subject(s)
Hedgehog Proteins , Renal Insufficiency, Chronic , Animals , Humans , Mice , Fibrosis , Hedgehog Proteins/metabolism , Inflammation , NF-kappa B , Tumor Necrosis Factors , Zinc Finger Protein GLI1
3.
Cell Stem Cell ; 27(5): 822-839.e8, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32946788

ABSTRACT

Hematopoietic stem cells (HSCs) first emerge in the embryonic aorta-gonad-mesonephros (AGM) region. Studies of model organisms defined intersecting signaling pathways that converge to promote HSC emergence predominantly in the ventral domain of the dorsal aorta. Much less is known about mechanisms driving HSC development in humans. Here, to identify secreted signals underlying human HSC development, we combined spatial transcriptomics analysis of dorsoventral polarized signaling in the aorta with gene expression profiling of sorted cell populations and single cells. Our analysis revealed a subset of aortic endothelial cells with a downregulated arterial signature and a predicted lineage relationship with the emerging HSC/progenitor population. Analysis of the ventrally polarized molecular landscape identified endothelin 1 as an important secreted regulator of human HSC development. The obtained gene expression datasets will inform future studies on mechanisms of HSC development in vivo and on generation of clinically relevant HSCs in vitro.


Subject(s)
Endothelial Cells , Transcriptome , Gonads , Hematopoiesis , Hematopoietic Stem Cells , Humans , Mesonephros , Transcriptome/genetics
4.
Stem Cell Reports ; 12(5): 1056-1068, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30956115

ABSTRACT

Definitive hematopoietic stem cells (HSCs) first emerge in the aorta-gonad-mesonephros (AGM) region in both mice and humans. An ex vivo culture approach has enabled recapitulation and analysis of murine HSC development. Knowledge of early human HSC development is hampered by scarcity of tissue: analysis of both CFU-C and HSC development in the human embryo is limited. Here, we characterized the spatial distribution and temporal kinetics of CFU-C development within early human embryonic tissues. We then sought to adapt the murine ex vivo culture system to recapitulate human HSC development. We show robust expansion of CFU-Cs and maintenance, but no significant expansion, of human HSCs in culture. Furthermore, we demonstrate that HSCs emerge predominantly in the middle section of the dorsal aorta in our culture system. We conclude that there are important differences between early mouse and human hematopoiesis, which currently hinder the quest to recapitulate human HSC development ex vivo.


Subject(s)
Aorta/cytology , Embryo, Mammalian/cytology , Hematopoietic Stem Cells/cytology , Spatio-Temporal Analysis , Stem Cells/cytology , Animals , Cell Proliferation/physiology , Cells, Cultured , Colony-Forming Units Assay , Embryo, Mammalian/embryology , Gonads/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation/methods , Heterografts , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
5.
Stem Cell Reports ; 11(3): 784-794, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30208304

ABSTRACT

Hematopoietic stem cells (HSCs) develop in the embryonic aorta-gonad-mesonephros (AGM) region and subsequently relocate to fetal liver. Runx1 transcription factor is essential for HSC development, but is largely dispensable for adult HSCs. Here, we studied tamoxifen-inducible Runx1 inactivation in vivo. Induction at pre-liver stages (up to embryonic day 10.5) reduced erythromyeloid progenitor numbers, but surprisingly did not block the appearance of Runx1-null HSCs in liver. By contrast, ex vivo analysis showed an absolute Runx1 dependency of HSC development in the AGM region. We found that, contrary to current beliefs, significant Cre-inducing tamoxifen activity persists in mouse blood for at least 72 hr after injection. This deferred recombination can hit healthy HSCs, which escaped early Runx1 ablation and result in appearance of Runx1-null HSCs in liver. Such extended recombination activity in vivo is a potential source of misinterpretation, particularly in analysis of dynamic developmental processes during embryogenesis.


Subject(s)
Aorta/embryology , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoietic Stem Cells/cytology , Liver/embryology , Mesonephros/embryology , Animals , Aorta/cytology , Core Binding Factor Alpha 2 Subunit/metabolism , Female , Gene Deletion , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Liver/cytology , Mesonephros/cytology , Mice, Inbred C57BL , Mice, Transgenic
6.
BMC Dev Biol ; 15: 35, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26453549

ABSTRACT

BACKGROUND: Pluripotent cells are present in early embryos until the levels of the pluripotency regulator Oct4 drop at the beginning of somitogenesis. Elevating Oct4 levels in explanted post-pluripotent cells in vitro restores their pluripotency. Cultured pluripotent cells can participate in normal development when introduced into host embryos up to the end of gastrulation. In contrast, pluripotent cells efficiently seed malignant teratocarcinomas in adult animals. In humans, extragonadal teratomas and teratocarcinomas are most frequently found in the sacrococcygeal region of neonates, suggesting that these tumours originate from cells in the posterior of the embryo that either reactivate or fail to switch off their pluripotent status. However, experimental models for the persistence or reactivation of pluripotency during embryonic development are lacking. METHODS: We manually injected embryonic stem cells into conceptuses at E9.5 to test whether the presence of pluripotent cells at this stage correlates with teratocarcinoma formation. We then examined the effects of reactivating embryonic Oct4 expression ubiquitously or in combination with Nanog within the primitive streak (PS)/tail bud (TB) using a transgenic mouse line and embryo chimeras carrying a PS/TB-specific heterologous gene expression cassette respectively. RESULTS: Here, we show that pluripotent cells seed teratomas in post-gastrulation embryos. However, at these stages, induced ubiquitous expression of Oct4 does not lead to restoration of pluripotency (indicated by Nanog expression) and tumour formation in utero, but instead causes a severe phenotype in the extending anteroposterior axis. Use of a more restricted T(Bra) promoter transgenic system enabling inducible ectopic expression of Oct4 and Nanog specifically in the posteriorly-located primitive streak (PS) and tail bud (TB) led to similar axial malformations to those induced by Oct4 alone. These cells underwent induction of pluripotency marker expression in Epiblast Stem Cell (EpiSC) explants derived from somitogenesis-stage embryos, but no teratocarcinoma formation was observed in vivo. CONCLUSIONS: Our findings show that although pluripotent cells with teratocarcinogenic potential can be produced in vitro by the overexpression of pluripotency regulators in explanted somitogenesis-stage somatic cells, the in vivo induction of these genes does not yield tumours. This suggests a restrictive regulatory role of the embryonic microenvironment in the induction of pluripotency.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Teratoma/metabolism , Teratoma/pathology , Animals , Embryo, Mammalian/pathology , Fetal Proteins/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Tail/embryology
7.
Development ; 141(17): 3319-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25139854

ABSTRACT

Haematopoiesis in adult animals is maintained by haematopoietic stem cells (HSCs), which self-renew and can give rise to all blood cell lineages. The AGM region is an important intra-embryonic site of HSC development and a wealth of evidence indicates that HSCs emerge from the endothelium of the embryonic dorsal aorta and extra-embryonic large arteries. This, however, is a stepwise process that occurs through sequential upregulation of CD41 and CD45 followed by emergence of fully functional definitive HSCs. Although largely dispensable at later stages, the Runx1 transcription factor is crucially important during developmental maturation of HSCs; however, exact points of crucial involvement of Runx1 in this multi-step developmental maturation process remain unclear. Here, we have investigated requirements for Runx1 using a conditional reversible knockout strategy. We report that Runx1 deficiency does not preclude formation of VE-cad+CD45-CD41+ cells, which are phenotypically equivalent to precursors of definitive HSCs (pre-HSC Type I) but blocks transition to the subsequent CD45+ stage (pre-HSC Type II). These data emphasise that developmental progression of HSCs during a very short period of time is regulated by precise stage-specific molecular mechanisms.


Subject(s)
Cell Lineage , Core Binding Factor Alpha 2 Subunit/metabolism , Embryo, Mammalian/cytology , Hematopoietic Stem Cells/cytology , Platelet Membrane Glycoprotein IIb/metabolism , Animals , Core Binding Factor Alpha 2 Subunit/deficiency , Hematopoietic Stem Cells/metabolism , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Blood ; 122(14): 2338-45, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23863896

ABSTRACT

During mouse development, definitive hematopoietic stem cells (dHSCs) emerge by late E10.5 to E11 in several hematopoietic sites. Of them, the aorta-gonad-mesonephros (AGM) region drew particular attention owing to its capacity to autonomously initiate and expand dHSCs in culture, indicating its key role in HSC development. The dorsal aorta contains characteristic hematopoietic clusters and is the initial site of dHSC emergence, where they mature through vascular endothelial (VE)-cadherin(+)CD45(-)CD41(low) (type 1 pre-HSCs) and VE-cadherin(+)CD45(+) (type 2 pre-HSCs) intermediates. Although dHSCs were also found in other embryonic niches (placenta, yolk sac, and extraembryonic vessels), attempts to detect their HSC initiating potential have been unsuccessful to date. Extraembryonic arterial vessels contain hematopoietic clusters, suggesting that they develop HSCs, but functional evidence for this has been lacking. Here we show that umbilical cord and vitelline arteries (VAs), but not veins, contain pre-HSCs capable of maturing into dHSCs in the presence of exogenous interleukin 3, although in fewer numbers than the AGM region, and that pre-HSC activity in VAs increases with proximity to the embryo proper. Our functional data strongly suggest that extraembryonic arteries can actively contribute to adult hematopoiesis.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Umbilical Arteries/cytology , Vitelline Duct/cytology , Animals , Flow Cytometry , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Arteries/embryology , Vitelline Duct/blood supply , Vitelline Duct/embryology
9.
Proc Natl Acad Sci U S A ; 110(16): 6542-7, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23576749

ABSTRACT

Tissue progenitor cells are an attractive target for regenerative therapy. In various organs, bone marrow cell (BMC) therapy has shown promising preliminary results, but to date no definite mechanism has been demonstrated to account for the observed benefit in organ regeneration. Tissue injury and regeneration is invariably accompanied by macrophage infiltration, but their influence upon the progenitor cells is incompletely understood, and direct signaling pathways may be obscured by the multiple roles of macrophages during organ injury. We therefore examined a model without injury; a single i.v. injection of unfractionated BMCs in healthy mice. This induced ductular reactions (DRs) in healthy mice. We demonstrate that macrophages within the unfractionated BMCs are responsible for the production of DRs, engrafting in the recipient liver and localizing to the DRs. Engrafted macrophages produce the cytokine TWEAK (TNF-like weak inducer of apoptosis) in situ. We go on to show that recombinant TWEAK activates DRs and that BMC mediated DRs are TWEAK dependent. DRs are accompanied by liver growth, occur in the absence of liver tissue injury and hepatic progenitor cells can be isolated from the livers of mice with DRs. Overall these results reveal a hitherto undescribed mechanism linking macrophage infiltration to DRs in the liver and highlight a rationale for macrophage derived cell therapy in regenerative medicine.


Subject(s)
Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/growth & development , Bone Marrow Transplantation/methods , Macrophages/metabolism , Regenerative Medicine/methods , Signal Transduction/physiology , Tumor Necrosis Factors/metabolism , Animals , Colony-Forming Units Assay , Cytokine TWEAK , Flow Cytometry , Immunohistochemistry , In Situ Hybridization, Fluorescence , Macrophages/physiology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Real-Time Polymerase Chain Reaction
10.
Stem Cells ; 30(2): 150-60, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22084016

ABSTRACT

Hematopoietic differentiation of embryonic stem cells (ESCs) in vitro has been used as a model to study early hematopoietic development, and it is well documented that hematopoietic differentiation can be enhanced by overexpression of HOXB4. HOXB4 is expressed in hematopoietic progenitor cells (HPCs) where it promotes self-renewal, but it is also expressed in the primitive streak of the gastrulating embryo. This led us to hypothesize that HOXB4 might modulate gene expression in prehematopoietic mesoderm and that this property might contribute to its prohematopoietic effect in differentiating ESCs. To test our hypothesis, we developed a conditionally activated HOXB4 expression system using the mutant estrogen receptor (ER(T2)) and showed that a pulse of HOXB4 prior to HPC emergence in differentiating ESCs led to an increase in hematopoietic differentiation. Expression profiling revealed an increase in the expression of genes associated with paraxial mesoderm that gives rise to the hematopoietic niche. Therefore, we considered that HOXB4 might modulate the formation of the hematopoietic niche as well as the production of hematopoietic cells per se. Cell mixing experiments supported this hypothesis demonstrating that HOXB4 activation can generate a paracrine as well as a cell autonomous effect on hematopoietic differentiation. We provide evidence to demonstrate that this activity is partly mediated by the secreted protein FRZB.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Stem Cell Niche , Transcription Factors/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Glycoproteins/genetics , Glycoproteins/metabolism , Hematopoiesis , Homeodomain Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Mice , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Paracrine Communication , Transcription Factors/genetics , beta Catenin/metabolism
11.
BMC Biol ; 9: 88, 2011 Dec 28.
Article in English | MEDLINE | ID: mdl-22204560

ABSTRACT

The close association between hematopoietic and endothelial cells during embryonic development led to the proposal that they may originate from a common ancestor--the hemangioblast. Due to a lack of unique specific markers for in vivo cell fate tracking studies, evidence supporting this theory derives mainly from in vitro differentiation studies. Teixeira and colleagues describe a novel enhancer that drives specific eGFP expression in blood islands of the electroporated chick embryo, thereby presenting a tool potentially suitable for analysis of hemangioblast differentiation and development of blood islands.


Subject(s)
Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Animals , Cell Differentiation , Chick Embryo , Electroporation , Hematopoiesis
12.
Stem Cells Dev ; 19(11): 1687-98, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20184433

ABSTRACT

Hematopoietic differentiation of embryonic stem (ES) cells can be enhanced by co-culture with stromal cells derived from hematopoietic tissues and by overexpression of the transcription factor HOXB4. In this study, we compare the hematopoietic inductive effects of stromal cell lines derived from different subregions of the embryonic aorta-gonad-mesonephros tissue with the commonly used OP9 stromal cell line and with HOXB4 activation. We show that stromal cell lines derived from the aorta and surrounding mesenchyme (AM) act at an earlier stage of the differentiation process compared with the commonly used OP9 stromal cells. AM stromal cells were able to promote the further differentiation of isolated brachyury-GFP(+) mesodermal cells into hematopoietic progenitors, whereas the OP9 stromal cells could not support the differentiation of these cells. Co-culture and analyses of individual embryoid bodies support the hypothesis that the AM stromal cell lines could enhance the de novo production of hematopoietic progenitors, lending support to the idea that AM stromal cells might act on prehematopoietic mesoderm. The induction level observed for AM stromal cells was comparable to HOXB4 activation, but no additive effect was observed when these 2 inductive strategies were combined. Addition of a γ-secretase inhibitor reduced the inductive effects of both the stromal cell line and HOXB4, providing clues to possible shared molecular mechanisms.


Subject(s)
Aorta/cytology , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Hematopoiesis/physiology , Homeodomain Proteins/metabolism , Mesoderm/cytology , Stromal Cells/physiology , Transcription Factors/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Aorta/embryology , Biomarkers/metabolism , Cell Line , Coculture Techniques , Embryo, Mammalian/cytology , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/cytology , Fetal Proteins/genetics , Fetal Proteins/metabolism , Homeodomain Proteins/genetics , Humans , Mesoderm/embryology , Mesonephros/cytology , Mesonephros/embryology , Mice , Stromal Cells/cytology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics
13.
Exp Cell Res ; 312(18): 3595-603, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16952354

ABSTRACT

We investigated whether the in vitro differentiation of ES cells into haematopoietic progenitors could be enhanced by exposure to the aorta-gonadal-mesonephros (AGM) microenvironment that is involved in the generation of haematopoietic stem cells (HSC) during embryonic development. We established a co-culture system that combines the requirements for primary organ culture and differentiating ES cells and showed that exposure of differentiating ES cells to the primary AGM region results in a significant increase in the number of ES-derived haematopoietic progenitors. Co-culture of ES cells on the AM20-1B4 stromal cell line derived from the AGM region also increases haematopoietic activity. We conclude that factors promoting the haematopoietic activity of differentiating ES cells present in primary AGM explants are partially retained in the AM20.1B4 stromal cell line and that these factors are likely to be different to those required for adult HSC maintenance.


Subject(s)
Aorta/metabolism , Cell Differentiation/physiology , Coculture Techniques , Embryonic Stem Cells/physiology , Gonads/metabolism , Hematopoiesis/physiology , Mesonephros/metabolism , Animals , Aorta/cytology , Cells, Cultured , Embryonic Stem Cells/cytology , Environment , Gonads/cytology , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Mesonephros/cytology , Mice , Mice, Inbred C57BL , Stromal Cells/cytology , Stromal Cells/physiology , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...