Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
EMBO J ; 43(9): 1722-1739, 2024 May.
Article in English | MEDLINE | ID: mdl-38580775

ABSTRACT

Understanding the regulatory mechanisms facilitating hematopoietic stem cell (HSC) specification during embryogenesis is important for the generation of HSCs in vitro. Megakaryocyte emerged from the yolk sac and produce platelets, which are involved in multiple biological processes, such as preventing hemorrhage. However, whether megakaryocytes regulate HSC development in the embryonic aorta-gonad-mesonephros (AGM) region is unclear. Here, we use platelet factor 4 (PF4)-Cre;Rosa-tdTomato+ cells to report presence of megakaryocytes in the HSC developmental niche. Further, we use the PF4-Cre;Rosa-DTA (DTA) depletion model to reveal that megakaryocytes control HSC specification in the mouse embryos. Megakaryocyte deficiency blocks the generation and maturation of pre-HSCs and alters HSC activity at the AGM. Furthermore, megakaryocytes promote endothelial-to-hematopoietic transition in a OP9-DL1 coculture system. Single-cell RNA-sequencing identifies megakaryocytes positive for the cell surface marker CD226 as the subpopulation with highest potential in promoting the hemogenic fate of endothelial cells by secreting TNFSF14. In line, TNFSF14 treatment rescues hematopoietic cell function in megakaryocyte-depleted cocultures. Taken together, megakaryocytes promote production and maturation of pre-HSCs, acting as a critical microenvironmental control factor during embryonic hematopoiesis.


Subject(s)
Hematopoietic Stem Cells , Megakaryocytes , Animals , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Cell Differentiation , Hematopoiesis/physiology , Mesonephros/embryology , Mesonephros/metabolism , Mesonephros/cytology , Endothelial Cells/metabolism , Endothelial Cells/cytology , Coculture Techniques
2.
Nat Cell Biol ; 24(4): 579-589, 2022 04.
Article in English | MEDLINE | ID: mdl-35414020

ABSTRACT

Intercellular communication orchestrates a multitude of physiologic and pathologic conditions. Algorithms to infer cell-cell communication and predict downstream signalling and regulatory networks are needed to illuminate mechanisms of stem cell differentiation and tissue development. Here, to fill this gap, we developed and applied CellComm to investigate how the aorta-gonad-mesonephros microenvironment dictates haematopoietic stem and progenitor cell emergence. We identified key microenvironmental signals and transcriptional networks that regulate haematopoietic development, including Stat3, Nr0b2, Ybx1 and App, and confirmed their roles using zebrafish, mouse and human models. Notably, CellComm revealed extensive crosstalk among signalling pathways and convergence on common transcriptional regulators, indicating a resilient developmental programme that ensures dynamic adaptation to changes in the embryonic environment. Our work provides an algorithm and data resource for the scientific community.


Subject(s)
Hematopoietic Stem Cells , Zebrafish , Animals , Cell Differentiation , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Mesonephros/metabolism , Mice , Zebrafish/genetics
3.
Blood ; 139(3): 343-356, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34517413

ABSTRACT

In vitro generation and expansion of hematopoietic stem cells (HSCs) holds great promise for the treatment of any ailment that relies on bone marrow or blood transplantation. To achieve this, it is essential to resolve the molecular and cellular pathways that govern HSC formation in the embryo. HSCs first emerge in the aorta-gonad-mesonephros (AGM) region, where a rare subset of endothelial cells, hemogenic endothelium (HE), undergoes an endothelial-to-hematopoietic transition (EHT). Here, we present full-length single-cell RNA sequencing (scRNA-seq) of the EHT process with a focus on HE and dorsal aorta niche cells. By using Runx1b and Gfi1/1b transgenic reporter mouse models to isolate HE, we uncovered that the pre-HE to HE continuum is specifically marked by angiotensin-I converting enzyme (ACE) expression. We established that HE cells begin to enter the cell cycle near the time of EHT initiation when their morphology still resembles endothelial cells. We further demonstrated that RUNX1 AGM niche cells consist of vascular smooth muscle cells and PDGFRa+ mesenchymal cells and can functionally support hematopoiesis. Overall, our study provides new insights into HE differentiation toward HSC and the role of AGM RUNX1+ niche cells in this process. Our expansive scRNA-seq datasets represents a powerful resource to investigate these processes further.


Subject(s)
Embryo, Mammalian/embryology , Hemangioblasts/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Animals , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Hemangioblasts/metabolism , Hematopoietic Stem Cells/metabolism , Mesonephros/cytology , Mesonephros/embryology , Mesonephros/metabolism , Mice , Single-Cell Analysis , Transcriptome , Zebrafish
4.
Acta Histochem ; 123(6): 151760, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34303296

ABSTRACT

Participation of molecular determinants of endocytosis in the processes of glomerular filtration and tubular reabsorption of albumin and lysozyme in the mesonephros of grass frogs (Rana temporaria L.), lake frogs (Rana ridibunda P.), and newts (Triturus vulgaris L.) is investigated. In all studied species, the constitutive expression of endocytic receptors in proximal tubule (PT) cells is established using immunofluorescence microscopy and immunoblotting. The certain stages of lysozyme and albumin endocytosis involving megalin/LRP2, cubilin, clathrin and protein Rab11 are detailed, and the central role of ligand-induced megalin/LRP2 activity in this process is shown. Increased ligand-induced expression for clathrin and Rab11was also found. In grass frogs, the different patterns of endocytic receptors and both absorbed proteins in the initial parts of proximal tubules suggest the proximo-distal specialization of absorptive processes along these tubule segments, similar to this in more complex mammalian nephrons. This data, as well as the revealed peculiarities of ligand-receptor interactions during intracellular trafficking of proteins prove that megalin is mainly involved in the absorption of lysozyme. At the same time, albumin absorption is mediated by both receptors, or cubilin contributes the most. The detection of endocytic receptor in glomerular structural elements in frogs and newts suggests the participation of filtration barrier components in endocytosis of filterable proteins. The results represent a new contribution to the study of the fundamental mechanisms of renal protein uptake in the amphibian mesonephros as a more primitive kidney compared to mammalian metanephros.


Subject(s)
Amphibian Proteins/metabolism , Endocytosis , Kidney Tubules, Proximal/metabolism , Mesonephros/metabolism , Animals , Protein Transport , Rana ridibunda , Rana temporaria , Triturus
5.
Biochem Biophys Res Commun ; 558: 161-167, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33930817

ABSTRACT

Current understanding of hematopoietic stem cell (HSC) development comes from mouse models is considered to be evolutionarily conserved in human. However, the cross-species comparison of the transcriptomic profiles of developmental HSCs at single-cell level is still lacking. Here, we performed integrative transcriptomic analysis of a series of key cell populations during HSC development in human and mouse, including HSC-primed hemogenic endothelial cells and pre-HSCs in mid-gestational aorta-gonad-mesonephros (AGM) region, and mature HSCs in fetal liver and adult bone marrow. We demonstrated the general similarity of transcriptomic characteristics between corresponding cell populations of the two species. Of note, one of the previously transcriptomically defined hematopoietic stem progenitor cell (HSPC) populations with certain arterial characteristics in AGM region of human embryos showed close transcriptomic similarity to pre-HSCs in mouse embryos. On the other hand, the other two HSPC populations in human AGM region displayed molecular similarity with fetal liver HSPCs, suggesting the maturation in AGM before HSCs colonizing the fetal liver in human, which was different to that in mouse. Finally, we re-clustered cells based on the integrated dataset and illustrated the evolutionarily conserved molecular signatures of major cell populations. Our results revealed transcriptomic conservation of critical cell populations and molecular characteristics during HSC development between human and mouse, providing a resource and theoretic basis for future studies on mammalian HSC development and regeneration by using mouse models.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Transcriptome , Animals , Cell Differentiation/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Hemangioblasts/cytology , Hemangioblasts/metabolism , Hematopoiesis/genetics , Humans , Mesonephros/cytology , Mesonephros/metabolism , Mice , Multigene Family , Single-Cell Analysis/methods , Species Specificity
6.
Pathol Res Pract ; 220: 153388, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33647867

ABSTRACT

Mesonephric remnants are embryonic vestiges of the mesonephric (Wolffian) ducts which regress during normal development. These remnants have been uncommonly reported in the female and male reproductive tract as a spectrum of morphologic lesions that can be misdiagnosed as carcinoma. One case of mesonephric remnant hyperplasia of the jejunal mesentery incidentally found in a 47-year-old man is herein reported. This is the first description of mesonephric hyperplasia arisen in the mesentery. The presence of ducts, tubules, and cysts lined by bland, epithelial, cuboidal cells with scant cytoplasm, and diffuse pseudoinfiltrative growth pattern can raise the possibility of neoplasia. Immunohistochemically, mesonephric epithelia have a characteristic staining. CD10 highlights the apical-luminal aspect of the cells. Besides, intense reactivity is showed for high-molecular-weight cytokeratin (CK), CK7, bcl2, and vimentin. The main differential diagnosis includes mesothelial hyperplasia, epithelial mesothelioma, well-differentiated neuroendocrine tumor, and infiltration due to acinar adenocarcinoma of the prostate. However, a detailed microscopic study with the aid of immunohistochemistry helps separate mesonephric remnants from malignant processes. The mesonephric hyperplasia of the mesentery we have reported adds to the spectrum of mesonephric remnants a new location. Familiarity with this lesion is indispensable to avoid overdiagnosis.


Subject(s)
Incidental Findings , Jejunum/pathology , Mesentery/pathology , Mesonephros/metabolism , Wolffian Ducts/pathology , Biomarkers/analysis , Biopsy , Diagnosis, Differential , Humans , Hyperplasia , Immunohistochemistry , Jejunum/chemistry , Jejunum/surgery , Male , Mesentery/chemistry , Mesentery/surgery , Mesonephros/chemistry , Mesonephros/surgery , Middle Aged , Predictive Value of Tests , Wolffian Ducts/chemistry , Wolffian Ducts/surgery
7.
IUBMB Life ; 72(1): 45-52, 2020 01.
Article in English | MEDLINE | ID: mdl-31634421

ABSTRACT

Runx1 is an important haematopoietic transcription factor as stressed by its involvement in a number of haematological malignancies. Furthermore, it is a key regulator of the emergence of the first haematopoietic stem cells (HSCs) during development. The transcription factor Gata3 has also been linked to haematological disease and was shown to promote HSC production in the embryo by inducing the secretion of important niche factors. Both proteins are expressed in several different cell types within the aorta-gonads-mesonephros (AGM) region, in which the first HSCs are generated; however, a direct interaction between these two key transcription factors in the context of embryonic HSC production has not formally been demonstrated. In this current study, we have detected co-localisation of Runx1 and Gata3 in rare sub-aortic mesenchymal cells in the AGM. Furthermore, the expression of Runx1 is reduced in Gata3 -/- embryos, which also display a shift in HSC emergence. Using an AGM-derived cell line as a model for the stromal microenvironment in the AGM and performing ChIP-Seq and ChIP-on-chip experiments, we demonstrate that Runx1, together with other key niche factors, is a direct target gene of Gata3. In addition, we can pinpoint Gata3 binding to the Runx1 locus at specific enhancer elements which are active in the microenvironment. These results reveal a direct interaction between Gata3 and Runx1 in the niche that supports embryonic HSCs and highlight a dual role for Runx1 in driving the transdifferentiation of haemogenic endothelial cells into HSCs as well as in the stromal cells that support this process.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Embryo, Mammalian/cytology , Embryonic Development , Endothelium, Vascular/cytology , GATA3 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Animals , Aorta/cytology , Aorta/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Embryo, Mammalian/metabolism , Endothelium, Vascular/metabolism , Female , GATA3 Transcription Factor/genetics , Gonads/cytology , Gonads/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Mesonephros/cytology , Mesonephros/metabolism , Mice , Mice, Inbred C57BL
8.
J Genet Genomics ; 46(10): 489-498, 2019 10 20.
Article in English | MEDLINE | ID: mdl-31776062

ABSTRACT

The functional heterogeneity of hematopoietic stem cells (HSCs) has been comprehensively investigated by single-cell transplantation assay. However, the heterogeneity regarding their physiological contribution remains an open question, especially for those with life-long hematopoietic fate of rigorous self-renewing and balanced differentiation capacities. In this study, we revealed that Procr expression was detected principally in phenotypical vascular endothelium co-expressing Dll4 and CD44 in the mid-gestation mouse embryos, and could enrich all the HSCs of the embryonic day 11.5 (E11.5) aorta-gonad-mesonephros (AGM) region. We then used a temporally restricted genetic tracing strategy to irreversibly label the Procr-expressing cells at E9.5. Interestingly, most labeled mature HSCs in multiple sites (such as AGM) around E11.5 were functionally categorized as lymphomyeloid-balanced HSCs assessed by direct transplantation. Furthermore, the labeled cells contributed to an average of 7.8% of immunophenotypically defined HSCs in E14.5 fetal liver (FL) and 6.9% of leukocytes in peripheral blood (PB) during one-year follow-up. Surprisingly, in aged mice of 24 months, the embryonically tagged cells displayed constant contribution to leukocytes with no bias to myeloid or lymphoid lineages. Altogether, we demonstrated, for the first time, the existence of a subtype of physiologically long-lived balanced HSCs as hypothesized, whose precise embryonic origin and molecular identity await further characterization.


Subject(s)
Endothelial Protein C Receptor/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Embryo, Mammalian , Endothelial Protein C Receptor/genetics , Female , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Male , Mesonephros/cytology , Mesonephros/metabolism , Mice , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
9.
Biochem Soc Trans ; 47(2): 591-601, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30902922

ABSTRACT

The first definitive blood cells during embryogenesis are derived from endothelial cells in a highly conserved process known as endothelial-to-haematopoietic transition (EHT). This conversion involves activation of a haematopoietic transcriptional programme in a subset of endothelial cells in the major vasculature of the embryo, followed by major morphological changes that result in transitioning cells rounding up, breaking the tight junctions to neighbouring endothelial cells and adopting a haematopoietic fate. The whole process is co-ordinated by a complex interplay of key transcription factors and signalling pathways, with additional input from surrounding tissues. Diverse model systems, including mouse, chick and zebrafish embryos as well as differentiation of pluripotent cells in vitro, have contributed to the elucidation of the details of the EHT, which was greatly accelerated in recent years by sophisticated live imaging techniques and advances in transcriptional profiling, such as single-cell RNA-Seq. A detailed knowledge of these developmental events is required in order to be able to apply it to the generation of haematopoietic stem cells from pluripotent stem cells in vitro - an achievement which is of obvious clinical importance. The aim of this review is to summarise the latest findings and describe how these may have contributed towards achieving this goal.


Subject(s)
Endothelium/cytology , Animals , Aorta/cytology , Aorta/metabolism , Endothelium/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Gonads/cytology , Gonads/metabolism , Hematopoiesis/genetics , Hematopoiesis/physiology , Humans , Mesonephros/cytology , Mesonephros/metabolism
10.
PLoS One ; 14(3): e0214130, 2019.
Article in English | MEDLINE | ID: mdl-30901367

ABSTRACT

Cells on the surface of the mesonephros give rise to replicating Gonadal Ridge Epithelial-Like (GREL) cells, the first somatic cells of the gonadal ridge. Later germ cells associate with the GREL cells in the ovigerous cords, and the GREL cells subsequently give rise to the granulosa cells in follicles. To examine these events further, 27 bovine fetal ovaries of different gestational ages were collected and prepared for immunohistochemical localisation of collagen type I and Ki67 to identify regions of the ovary and cell proliferation, respectively. The non-stromal cortical areas (collagen-negative) containing GREL cells and germ cells and later in development, the follicles with oocytes and granulosa cells, were analysed morphometrically. Another set of ovaries (n = 17) were collected and the expression of genes associated with germ cell lineages and GREL/granulosa cells were quantitated by RT-PCR. The total volume of non-stromal areas in the cortex increased significantly and progressively with ovarian development, plateauing at the time the surface epithelium developed. However, the proportion of non-stromal areas in the cortex declined significantly and progressively throughout gestation, largely due to a cessation in growth of the non-stroma cells and the continued growth of stroma. The proliferation index in the non-stromal area was very high initially and then declined substantially at the time follicles formed. Thereafter, it remained low. The numerical density of the non-stromal cells was relatively constant throughout ovarian development. The expression levels of a number of genes across gestation either increased (AMH, FSHR, ESR1, INHBA), declined (CYP19A1, ESR2, ALDH1A1, DSG2, OCT4, LGR5) or showed no particular pattern (CCND2, CTNNB1, DAZL, FOXL2, GATA4, IGFBP3, KRT19, NR5A1, RARRES1, VASA, WNT2B). Many of the genes whose expression changed across gestation, were positively or negatively correlated with each other. The relationships between these genes may reflect their roles in the important events such as the transition of ovigerous cords to follicles, oogonia to oocytes or GREL cells to granulosa cells.


Subject(s)
Cattle/embryology , Gene Expression Regulation, Developmental , Ovary/embryology , Animals , Cattle/genetics , Female , Germ Cells/cytology , Germ Cells/metabolism , Granulosa Cells/cytology , Granulosa Cells/metabolism , Mesonephros/cytology , Mesonephros/embryology , Mesonephros/metabolism , Ovary/cytology , Ovary/metabolism
11.
Semin Cell Dev Biol ; 91: 86-93, 2019 07.
Article in English | MEDLINE | ID: mdl-30172050

ABSTRACT

The intermediate mesoderm is located between the somites and the lateral plate mesoderm and gives rise to renal progenitors that contribute to the three mammalian kidney types (pronephros, mesonephros and metanephros). In this review, focusing largely on murine kidney development, we examine how the intermediate mesoderm forms during gastrulation/axis elongation and how it progressively gives rise to distinct renal progenitors along the rostro-caudal axis. We highlight some of the potential signalling cues and core transcription factor circuits that direct these processes, up to the point of early metanephric kidney formation.


Subject(s)
Kidney/embryology , Mesoderm/embryology , Mesonephros/embryology , Somites/embryology , Animals , Body Patterning/genetics , Gene Expression Regulation, Developmental , Kidney/metabolism , Mesoderm/metabolism , Mesonephros/metabolism , Mice , Organogenesis/genetics , Somites/metabolism , Transcription Factors/genetics
12.
Thromb Haemost ; 118(8): 1370-1381, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29991091

ABSTRACT

The development of new strategies based on cell therapy approaches to correct haemophilia A (HA) requires further insights into new cell populations capable of producing coagulation factor VIII (FVIII) and presenting stable engraftment potential. The major producers of FVIII in the adult are liver sinusoidal endothelial cells (LSECs) and in a lesser degree bone marrow-derived cells, both of which have been shown to ameliorate the bleeding phenotype in adult HA mice after transplantation. We have previously shown that cells from the foetal liver (FL) and the aorta-gonads-mesonephros (AGM) haematopoietic locations possess higher LSEC engraftment potential in newborn mice compared with adult-derived LSECs, constituting likely therapeutic targets for the treatment of HA in neonates. However, less is known about the production of FVIII in embryonic locations. Quantitative polymerase chain reaction and Western blot analysis were performed to assess the relative level of FVIII production in different embryonic tissues and at various developmental stages, identifying the FL and AGM region from day 12 (E12) as prominent sources of FVIII. Furthermore, FL-derived VE-cad+CD45-Lyve1+/- endothelial/endothelial progenitor cells, presenting vascular engraftment potential, produced high levels of F8 ribonucleic acid compared with CD45+ blood progenitors or Dlk1+ hepatoblasts. In addition, we show that the E11 AGM explant cultures expanded cells with LSEC repopulation activity, instrumental to further understand signals for in vitro generation of LSECs. Taking into account the capacity for FVIII expression, culture expansion and newborn engraftment potential, these results support the use of cells with foetal characteristics for correction of FVIII deficiency in young individuals.


Subject(s)
Aorta/metabolism , Endothelial Progenitor Cells/metabolism , Factor VIII/metabolism , Gonads/metabolism , Hemophilia A/metabolism , Liver/metabolism , Mesonephros/metabolism , Animals , Aorta/embryology , Aorta/transplantation , Cell Differentiation , Endothelial Progenitor Cells/transplantation , Factor VIII/genetics , Gene Expression Regulation, Developmental , Gestational Age , Gonads/embryology , Gonads/transplantation , Hemophilia A/genetics , Hemophilia A/surgery , Liver/embryology , Mesonephros/embryology , Mesonephros/transplantation , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cell Transplantation/methods , Tissue Culture Techniques
13.
Am J Physiol Renal Physiol ; 315(1): F130-F137, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29561184

ABSTRACT

The mesonephros of mammals is a transient renal structure that contributes to various aspects of mammalian fetal development, including the male reproductive system, hematopoietic stem cells, and vascular endothelial cells. The mesonephros develops from the intermediate mesoderm and forms tubules that are segmented in a similar way to the nephrons of the permanent kidney (but lacking loops of Henle). Early studies have suggested that the mesonephros in marsupials and some placental mammals may perform an excretory function, but these studies have not directly shown active transport of organic anions and cations. Excretory function in the rodent mesonephros has not been investigated. Functional characterization of the earliest stages of mammalian renal development is important for our understanding of congenital disease and may help to inform the growing field of renal tissue engineering. Here, we use live uptake and efflux assays in vitro to show that the murine mesonephros is able to transport organic anions and cations through specific transporters from early in its development. Transcript analysis suggests that there are subtle differences between the transporters involved in uptake and efflux by the murine permanent metanephric tubules and by the mesonephric tubules. These data suggest that the mammalian mesonephros can provide an excretory function for the early developing embryo, in addition to the excretory function provided by the placenta.


Subject(s)
Mesonephros/metabolism , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Animals , Biological Transport , Female , Gene Expression Regulation, Developmental , Gestational Age , Mice , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Pregnancy , Tissue Culture Techniques
14.
Mol Hum Reprod ; 24(5): 233-243, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29528446

ABSTRACT

STUDY QUESTION: Which set of antibodies can be used to identify migratory and early post-migratory human primordial germ cells (hPGCs)? STUDY FINDING: We validated the specificity of 33 antibodies for 31 markers, including POU5F1, NANOG, PRDM1 and TFAP2C as specific markers of hPGCs at 4.5 weeks of development of Carnegie stage (CS12-13), whereas KIT and SOX17 also marked the intra-aortic hematopoietic stem cell cluster in the aorta-gonad-mesonephros (AGM). WHAT IS KNOWN ALREADY: The dynamics of gene expression during germ cell development in mice is well characterized and this knowledge has proved crucial to allow the development of protocols for the in vitro derivation of functional gametes. Although there is a great interest in generating human gametes in vitro, it is still unclear which markers are expressed during the early stages of hPGC development and many studies use markers described in mouse to benchmark differentiation of human PGC-like cells (hPGCLCs). Early post-implantation development differs significantly between mice and humans, and so some germ cells markers, including SOX2, SOX17, IFITM3 and ITGA6 may not identify mPGCs and hPGCs equally well. STUDY DESIGN, SIZE, DURATION: This immunofluorescence study investigated the expression of putative hPGC markers in the caudal part of a single human embryo at 4.5 weeks of development. PARTICIPANTS/MATERIALS, SETTING, METHODS: We have investigated by immunofluorescence the expression of a set of 33 antibodies for 31 markers, including pluripotency, germ cell, adhesion, migration, surface, mesenchymal and epigenetic markers on paraffin sections of the caudal part, including the AGM region, of a single human embryo (CS12-13). The human material used was anonymously donated with informed consent from elective abortions without medical indication. MAIN RESULTS AND THE ROLE OF CHANCE: We observed germ cell specific expression of NANOG, TFAP2C and PRDM1 in POU5F1+ hPGCs in the AGM. The epigenetic markers H3K27me3 and 5mC were sufficient to distinguish hPGCs from the surrounding somatic cells. Some mPGC-markers were not detected in hPGCs, but marked other tissues; whereas other markers, such as ALPL, SOX17, KIT, TUBB3, ITGA6 marked both POU5F1+ hPGCs and other cells in the AGM. We used a combination of multiple markers, immunostaining different cellular compartments when feasible, to decrease the chance of misidentifying hPGCs. LARGE SCALE DATA: Non-applicable. LIMITATIONS REASONS FOR CAUTION: Material to study early human development is unique and very rare thus restricting the sample size. We have used a combination of antibodies limited by the number of paraffin sections available. WIDER IMPLICATIONS OF THE FINDINGS: Most of our knowledge on early gametogenesis has been obtained from model organisms such as mice and is extrapolated to humans. However, since there is a dedicated effort to produce human artificial gametes in vitro, it is of great importance to determine the expression and specificity of human-specific germ cell markers. We provide a systematic analysis of the expression of 31 different markers in paraffin sections of a CS12-13 embryo. Our results will help to set up a toolbox of markers to evaluate protocols to induce hPGCLCs in vitro. STUDY FUNDING AND COMPETING INTEREST(S): M.G.F. was funded by Fundação para a Ciência e Tecnologia (FCT) [SFRH/BD/78689/2011] and S.M.C.S.L. was funded by the Interuniversity Attraction Poles (IAP, P7/07) and the European Research Council Consolidator (ERC-CoG-725722-OVOGROWTH). The authors declare no conflict of interest.


Subject(s)
Aorta/cytology , Gametogenesis/physiology , Germ Cells/cytology , Gonads/cytology , Mesonephros/cytology , Aorta/embryology , Aorta/metabolism , Biomarkers/metabolism , Cell Adhesion/physiology , Cell Movement/physiology , Germ Cells/metabolism , Gonads/embryology , Gonads/metabolism , Humans , Mesonephros/embryology , Mesonephros/metabolism
15.
Exp Cell Res ; 365(1): 145-155, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29458175

ABSTRACT

The aorta-gonad-mesonephros region, from which definitive hematopoiesis first arises in midgestation mouse embryos, has intra-aortic hematopoietic clusters (IAHCs) containing hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). We previously reported expression of the transcription factor Sox17 in IAHCs, and overexpression of Sox17 in CD45lowc-KIThigh cells comprising IAHCs maintains the formation of cell clusters and their multipotency in vitro over multiple passages. Here, we demonstrate the importance of NOTCH1 in IAHC formation and maintenance of the HSC/HPC phenotype. We further show that Notch1 expression is positively regulated by SOX17 via direct binding to its gene promoter. SOX17 and NOTCH1 were both found to be expressed in vivo in cells of IAHCs by whole mount immunostaining. We found that cells transduced with the active form of NOTCH1 or its downstream target, Hes1, maintained their multipotent colony-forming capacity in semisolid medium. Moreover, cells stimulated by NOTCH1 ligand, Jagged1, or Delta-like protein 1, had the capacity to form multilineage colonies. Conversely, knockdown of Notch1 and Hes1 led to a reduction of their multipotent colony-forming capacity. These results suggest that the Sox17-Notch1-Hes1 pathway is critical for maintaining the undifferentiated state of IAHCs.


Subject(s)
Aorta/metabolism , HMGB Proteins/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Receptor, Notch1/metabolism , SOXF Transcription Factors/metabolism , Transcription Factor HES-1/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Fetus/metabolism , Gonads/metabolism , Mesonephros/metabolism , Mice , Mice, Inbred ICR , Promoter Regions, Genetic/physiology
16.
J Exp Med ; 214(12): 3731-3751, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-29093060

ABSTRACT

In the developing embryo, hematopoietic stem cells (HSCs) emerge from the aorta-gonad-mesonephros (AGM) region, but the molecular regulation of this process is poorly understood. Recently, the progression from E9.5 to E10.5 and polarity along the dorso-ventral axis have been identified as clear demarcations of the supportive HSC niche. To identify novel secreted regulators of HSC maturation, we performed RNA sequencing over these spatiotemporal transitions in the AGM region and supportive OP9 cell line. Screening several proteins through an ex vivo reaggregate culture system, we identify BMPER as a novel positive regulator of HSC development. We demonstrate that BMPER is associated with BMP signaling inhibition, but is transcriptionally induced by BMP4, suggesting that BMPER contributes to the precise control of BMP activity within the AGM region, enabling the maturation of HSCs within a BMP-negative environment. These findings and the availability of our transcriptional data through an accessible interface should provide insight into the maintenance and potential derivation of HSCs in culture.


Subject(s)
Aorta/metabolism , Cell Differentiation , Gonads/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mesonephros/metabolism , Animals , Aorta/embryology , Bone Morphogenetic Proteins/metabolism , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cluster Analysis , Feedback, Physiological , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gonads/embryology , Mesoderm/metabolism , Mesonephros/embryology , Mice, Inbred C57BL , Signal Transduction , Smad Proteins/metabolism , Stem Cell Niche/genetics , Time Factors
17.
J Exp Med ; 214(11): 3347-3360, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-28931624

ABSTRACT

T lymphocytes are key cellular components of the adaptive immune system and play a central role in cell-mediated immunity in vertebrates. Despite their heterogeneities, it is believed that all different types of T lymphocytes are generated exclusively via the differentiation of hematopoietic stem cells (HSCs). Using temporal-spatial resolved fate-mapping analysis and time-lapse imaging, here we show that the ventral endothelium in the zebrafish aorta-gonad-mesonephros and posterior blood island, the hematopoietic tissues previously known to generate HSCs and erythromyeloid progenitors, respectively, gives rise to a transient wave of T lymphopoiesis independent of HSCs. This HSC-independent T lymphopoiesis occurs early and generates predominantly CD4 Tαß cells in the larval but not juvenile and adult stages, whereas HSC-dependent T lymphopoiesis emerges late and produces various subtypes of T lymphocytes continuously from the larval stage to adulthood. Our study unveils the existence, origin, and ontogeny of HSC-independent T lymphopoiesis in vivo and reveals the complexity of the endothelial-hematopoietic transition of the aorta.


Subject(s)
Aorta/cytology , Embryo, Nonmammalian/cytology , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/cytology , Lymphopoiesis , T-Lymphocytes/cytology , Animals , Animals, Genetically Modified , Aorta/embryology , Aorta/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Gonads/cytology , Gonads/embryology , Gonads/metabolism , Hematopoietic Stem Cells/metabolism , In Situ Hybridization , Mesonephros/cytology , Mesonephros/embryology , Mesonephros/metabolism , Microscopy, Confocal , T-Lymphocytes/metabolism , Time-Lapse Imaging/methods , Zebrafish
18.
Stem Cell Reports ; 8(6): 1549-1562, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28479304

ABSTRACT

During development, hematopoietic stem cells (HSCs) emerge in the aorta-gonad-mesonephros (AGM) region through a process of multi-step maturation and expansion. While proliferation of adult HSCs is implicated in the balance between self-renewal and differentiation, very little is known about the proliferation status of nascent HSCs in the AGM region. Using Fucci reporter mice that enable in vivo visualization of cell-cycle status, we detect increased proliferation during pre-HSC expansion followed by a slowing down of cycling once cells start to acquire a definitive HSC state, similar to fetal liver HSCs. We observe time-specific changes in intra-aortic hematopoietic clusters corresponding to HSC maturation stages. The proliferative architecture of the clusters is maintained in an orderly anatomical manner with slowly cycling cells at the base and more actively proliferating cells at the more apical part of the cluster, which correlates with c-KIT expression levels, thus providing an anatomical basis for the role of SCF in HSC maturation.


Subject(s)
Aorta/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Genes, Reporter , Gonads/metabolism , Hematopoietic Stem Cells/cytology , Leukocyte Common Antigens/metabolism , Mesonephros/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Platelet Membrane Glycoprotein IIb/metabolism , Proto-Oncogene Proteins c-kit/metabolism
19.
Dev Biol ; 424(2): 208-220, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28274610

ABSTRACT

Retinoic acid (RA) is a potent inducer of cell differentiation and plays an essential role in sex-specific germ cell development in the mammalian gonad. RA is essential for male gametogenesis and hence fertility. However, RA can also disrupt sexual cell fate in somatic cells of the testis, promoting transdifferentiation of male Sertoli cells to female granulosa-like cells when the male sexual regulator Dmrt1 is absent. The feminizing ability of RA in the Dmrt1 mutant somatic testis suggests that RA might normally play a role in somatic cell differentiation or cell fate maintenance in the ovary. To test for this possibility we disrupted RA signaling in somatic cells of the early fetal ovary using three genetic strategies and one pharmaceutical approach. We found that deleting all three RA receptors (RARs) in the XX somatic gonad at the time of sex determination did not significantly affect ovarian differentiation, follicle development, or female fertility. Transcriptome analysis of adult triple mutant ovaries revealed remarkably little effect on gene expression in the absence of somatic RAR function. Likewise, deletion of three RA synthesis enzymes (Aldh1a1-3) at the time of sex determination did not masculinize the ovary. A dominant-negative RAR transgene altered granulosa cell proliferation, likely due to interference with a non-RA signaling pathway, but did not prevent granulosa cell specification and oogenesis or abolish fertility. Finally, culture of fetal XX gonads with an RAR antagonist blocked germ cell meiotic initiation but did not disrupt sex-biased gene expression. We conclude that RA signaling, although crucial in the ovary for meiotic initiation, is not required for granulosa cell specification, differentiation, or reproductive function.


Subject(s)
Ovary/embryology , Ovary/metabolism , Signal Transduction/drug effects , Tretinoin/pharmacology , Aldehyde Dehydrogenase 1 Family , Animals , Cell Lineage/drug effects , Female , Fetus/embryology , Fetus/metabolism , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Gene Knockout Techniques , Genes, Dominant , Isoenzymes/metabolism , Male , Mammals , Meiosis/drug effects , Mesonephros/drug effects , Mesonephros/embryology , Mesonephros/metabolism , Mice , Ovary/drug effects , Receptors, Retinoic Acid/metabolism , Retinal Dehydrogenase/metabolism , Retinoids/pharmacology , Sex Determination Processes/drug effects , Tissue Culture Techniques
20.
Cytokine ; 95: 35-42, 2017 07.
Article in English | MEDLINE | ID: mdl-28235674

ABSTRACT

In the midgestation mouse embryo, hematopoietic cell clusters containing hematopoietic stem/progenitor cells arise in the aorta-gonad-mesonephros (AGM) region. We have previously reported that forced expression of the Sox17 transcription factor in CD45lowc-Kithigh AGM cells, which are the hematopoietic cellular component of the cell clusters, and subsequent coculture with OP9 stromal cells in the presence of three cytokines, stem cell factor (SCF), interleukin-3 (IL-3), and thrombopoietin (TPO), led to the formation and the maintenance of cell clusters with cells at an undifferentiated state in vitro. In this study, we investigated the role of each cytokine in the formation of hematopoietic cell clusters. We cultured Sox17-transduced AGM cells with each of the 7 possible combinations of the three cytokines. The size and the number of Sox17-transduced cell clusters in the presence of TPO, either alone or in combination, were comparable to that observed with the complete set of the three cytokines. Expression of TPO receptor, c-Mpl was almost ubiquitously expressed and maintained in Sox17-transduced hematopoietic cell clusters. In addition, the expression level of c-Mpl was highest in the CD45lowc-Kithigh cells among the Sox17-transduced cell clusters. Moreover, c-Mpl protein was highly expressed in the intra-aortic hematopoietic cell clusters in comparison with endothelial cells of dorsal aorta. Finally, stimulation of the endothelial cells prepared from the AGM region by TPO induced the production of hematopoietic cells. These results suggest that TPO contributes to the formation and the maintenance of hematopoietic cell clusters in the AGM region.


Subject(s)
Aorta/cytology , Gonads/cytology , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Mesonephros/cytology , Thrombopoietin/physiology , Animals , Aorta/embryology , Aorta/metabolism , Cells, Cultured , Gonads/embryology , Gonads/metabolism , Interleukin-3/physiology , Mesonephros/metabolism , Mice, Inbred C57BL , Mice, Inbred ICR , Receptors, Thrombopoietin/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Signal Transduction , Stem Cell Factor/physiology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...