Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Sci Immunol ; 8(85): eabo4365, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37450574

ABSTRACT

Vγ9Vδ2 T cells are the largest population of γδ T cells in adults and can play important roles in providing effective immunity against cancer and infection. Many studies have suggested that peripheral Vγ9Vδ2 T cells are derived from the fetal liver and thymus and that the postnatal thymus plays little role in the development of these cells. More recent evidence suggested that these cells may also develop postnatally in the thymus. Here, we used high-dimensional flow cytometry, transcriptomic analysis, functional assays, and precursor-product experiments to define the development pathway of Vγ9Vδ2 T cells in the postnatal thymus. We identify three distinct stages of development for Vγ9Vδ2 T cells in the postnatal thymus that are defined by the progressive acquisition of functional potential and major changes in the expression of transcription factors, chemokines, and other surface markers. Furthermore, our analysis of donor-matched thymus and blood revealed that the molecular requirements for the development of functional Vγ9Vδ2 T cells are delivered predominantly by the postnatal thymus and not in the periphery. Tbet and Eomes, which are required for IFN-γ and TNFα expression, are up-regulated as Vγ9Vδ2 T cells mature in the thymus, and mature thymic Vγ9Vδ2 T cells rapidly express high levels of these cytokines after stimulation. Similarly, the postnatal thymus programs Vγ9Vδ2 T cells to express the cytolytic molecules, perforin, granzyme A, and granzyme K. This study provides a greater understanding of how Vγ9Vδ2 T cells develop in humans and may lead to opportunities to manipulate these cells to treat human diseases.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets , Adult , Humans , Thymus Gland , Gene Expression Profiling
2.
Stem Cell Res ; 69: 103109, 2023 06.
Article in English | MEDLINE | ID: mdl-37150143

ABSTRACT

We describe the generation and characterisation of five human induced pluripotent stem cell (iPSC) lines derived from peripheral blood mononuclear cells (PBMCs) of healthy adult individuals. The PBMCs were reprogrammed using non-integrating Sendai viruses containing the reprogramming factors POU5F1 (OCT4), SOX2, KLF4 and MYC. The iPSC lines exhibited a normal karyotype, and pluripotency was validated by flow cytometry and immunofluorescence of pluripotency markers, and their differentiation into cells representative of the three embryonic germ layers. These iPSC lines can be used as controls in studying disease mechanisms.


Subject(s)
Induced Pluripotent Stem Cells , Adult , Humans , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , Kruppel-Like Factor 4 , Cell Differentiation , Cell Line , Cellular Reprogramming
3.
Proc Natl Acad Sci U S A ; 120(19): e2211510120, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37126720

ABSTRACT

Chondrocytes and osteoblasts differentiated from induced pluripotent stem cells (iPSCs) will provide insights into skeletal development and genetic skeletal disorders and will generate cells for regenerative medicine applications. Here, we describe a method that directs iPSC-derived sclerotome to chondroprogenitors in 3D pellet culture then to articular chondrocytes or, alternatively, along the growth plate cartilage pathway to become hypertrophic chondrocytes that can transition to osteoblasts. Osteogenic organoids deposit and mineralize a collagen I extracellular matrix (ECM), mirroring in vivo endochondral bone formation. We have identified gene expression signatures at key developmental stages including chondrocyte maturation, hypertrophy, and transition to osteoblasts and show that this system can be used to model genetic cartilage and bone disorders.


Subject(s)
Cartilage , Induced Pluripotent Stem Cells , Humans , Cartilage/metabolism , Chondrocytes/metabolism , Cell Differentiation , Osteoblasts , Induced Pluripotent Stem Cells/metabolism
4.
Cell Rep ; 42(5): 112322, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37105170

ABSTRACT

Crosstalk between cardiac cells is critical for heart performance. Here we show that vascular cells within human cardiac organoids (hCOs) enhance their maturation, force of contraction, and utility in disease modeling. Herein we optimize our protocol to generate vascular populations in addition to epicardial, fibroblast, and cardiomyocyte cells that self-organize into in-vivo-like structures in hCOs. We identify mechanisms of communication between endothelial cells, pericytes, fibroblasts, and cardiomyocytes that ultimately contribute to cardiac organoid maturation. In particular, (1) endothelial-derived LAMA5 regulates expression of mature sarcomeric proteins and contractility, and (2) paracrine platelet-derived growth factor receptor ß (PDGFRß) signaling from vascular cells upregulates matrix deposition to augment hCO contractile force. Finally, we demonstrate that vascular cells determine the magnitude of diastolic dysfunction caused by inflammatory factors and identify a paracrine role of endothelin driving dysfunction. Together this study highlights the importance and role of vascular cells in organoid models.


Subject(s)
Endothelial Cells , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Pericytes/metabolism , Signal Transduction , Organoids/metabolism
5.
Immunol Rev ; 315(1): 154-170, 2023 05.
Article in English | MEDLINE | ID: mdl-36939073

ABSTRACT

Lymphoid cells encompass the adaptive immune system, including T and B cells and Natural killer T cells (NKT), and innate immune cells (ILCs), including Natural Killer (NK) cells. During adult life, these lineages are thought to derive from the differentiation of long-term hematopoietic stem cells (HSCs) residing in the bone marrow. However, during embryogenesis and fetal development, the ontogeny of lymphoid cells is both complex and multifaceted, with a large body of evidence suggesting that lymphoid lineages arise from progenitor cell populations antedating the emergence of HSCs. Recently, the application of single cell RNA-sequencing technologies and pluripotent stem cell-based developmental models has provided new insights into lymphoid ontogeny during embryogenesis. Indeed, PSC differentiation platforms have enabled de novo generation of lymphoid immune cells independently of HSCs, supporting conclusions drawn from the study of hematopoiesis in vivo. Here, we examine lymphoid development from non-HSC progenitor cells and technological advances in the differentiation of human lymphoid cells from pluripotent stem cells for clinical translation.


Subject(s)
Pluripotent Stem Cells , Adult , Humans , Cell Differentiation , Hematopoietic Stem Cells , Killer Cells, Natural , Hematopoiesis
6.
Cell Rep ; 40(11): 111339, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36103836

ABSTRACT

Precursors of the adult hematopoietic system arise from the aorta-gonad-mesonephros (AGM) region shortly after the embryonic circulation is established. Here, we develop a microfluidic culture system to mimic the primitive embryonic circulation and address the hypothesis that circulatory flow and shear stress enhance embryonic blood development. Embryonic (HOXA+) hematopoiesis was derived from human pluripotent stem cells and induced from mesoderm by small-molecule manipulation of TGF-ß and WNT signaling (SB/CHIR). Microfluidic and orbital culture promoted the formation of proliferative CD34+RUNX1C-GFP+SOX17-mCHERRY+ precursor cells that were released into the artificial circulation from SOX17+ arterial-like structures. Single-cell transcriptomic analysis delineated extra-embryonic (yolk sac) and HOXA+ embryonic blood differentiation pathways. SB/CHIR and circulatory flow enhance hematopoiesis by the formation of proliferative HOXA+RUNX1C+CD34+ precursor cells that differentiate into monocyte/macrophage, granulocyte, erythrocyte, and megakaryocyte progenitors.


Subject(s)
Hematopoiesis , Mesonephros , Adult , Antigens, CD34 , Cell Differentiation , Hematopoietic Stem Cells , Humans , Yolk Sac
7.
Stem Cell Reports ; 17(9): 2156-2166, 2022 09 13.
Article in English | MEDLINE | ID: mdl-35985333

ABSTRACT

Human macrophages are a natural host of many mycobacterium species, including Mycobacterium abscessus (M. abscessus), an emerging pathogen affecting immunocompromised and cystic fibrosis patients with few available treatments. The search for an effective treatment is hindered by the lack of a tractable in vitro intracellular infection model. Here, we established a reliable model for M. abscessus infection using human pluripotent stem cell-derived macrophages (hPSC-macrophages). hPSC differentiation permitted reproducible generation of functional macrophages that were highly susceptible to M. abscessus infection. Electron microscopy demonstrated that M. abscessus was present in the hPSC-macrophage vacuoles. RNA sequencing analysis revealed a time-dependent host cell response, with differing gene and protein expression patterns post-infection. Engineered tdTOMATO-expressing hPSC-macrophages with GFP-expressing mycobacteria enabled rapid image-based high-throughput analysis of intracellular infection and quantitative assessment of antibiotic efficacy. Our study describes the first to our knowledge hPSC-based model for M. abscessus infection, representing a novel and accessible system for studying pathogen-host interaction and drug discovery.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Mycobacterium , Pluripotent Stem Cells , Humans , Macrophages/metabolism , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium Infections, Nontuberculous/microbiology
8.
Front Immunol ; 13: 846281, 2022.
Article in English | MEDLINE | ID: mdl-35371075

ABSTRACT

Thymic epithelium is critical for the structural integrity of the thymus and for T cell development. Within the fully formed thymus, large numbers of hematopoietic cells shape the thymic epithelium into a scaffold-like structure which bears little similarity to classical epithelial layers, such as those observed in the skin, intestine or pancreas. Here, we show that human thymic epithelial cells (TECs) possess an epithelial identity that also incorporates the expression of mesenchymal cell associated genes, whose expression levels vary between medullary and cortical TECs (m/cTECs). Using pluripotent stem cell (PSC) differentiation systems, we identified a unique population of cells that co-expressed the master TEC transcription factor FOXN1, as well as the epithelial associated marker EPCAM and the mesenchymal associated gene CD90. Using the same serum free culture conditions, we also observed co-expression of EPCAM and CD90 on cultured TECs derived from neonatal human thymus in vitro. Single cell RNA-sequencing revealed these cultured TECs possessed an immature mTEC phenotype and expressed epithelial and mesenchymal associated genes, such as EPCAM, CLDN4, CD90 and COL1A1. Importantly, flow cytometry and single cell RNA-sequencing analysis further confirmed the presence of an EPCAM+CD90+ population in the CD45- fraction of neonatal human thymic stromal cells in vivo. Using the human thymus cell atlas, we found that cTECs displayed more pronounced mesenchymal characteristics than mTECs during embryonic development. Collectively, these results suggest human TECs possess a hybrid gene expression program comprising both epithelial and mesenchymal elements, and provide a basis for the further exploration of thymus development from primary tissues and from the in vitro differentiation of PSCs.


Subject(s)
Epithelial Cells , RNA , Cell Differentiation , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cells/metabolism , Epithelium , Humans , RNA/metabolism , Thy-1 Antigens/metabolism , Thymus Gland
9.
Front Cell Dev Biol ; 10: 835321, 2022.
Article in English | MEDLINE | ID: mdl-35372371

ABSTRACT

Many clinical trials are in progress using cells derived from induced pluripotent stem cells (iPSC) for immunotherapies and regenerative medicine. The success of these new therapies is underpinned by the quality of the cell population used to create the iPSC lines, along with the creation of iPSCs in a fully Good Manufacturing Practice (GMP)-compliant environment such that they can be used safely and effectively in the clinical setting. Umbilical cord blood (CB) from public cord blood banks is an excellent source of starting material for creation of iPSCs. All CB units are manufactured under GMP-conditions, have been screened for infectious diseases, with known family and medical history of the donor. Furthermore, the HLA tissue typing is known, thereby allowing identification of CB units with homozygous HLA haplotypes. CB cells are naïve with less exposure to environmental insults and iPSC can be generated with high efficiency. We describe a protocol that can be adopted by those seeking to create clinical-grade iPSC from banked CB. This protocol uses a small volume of thawed CB buffy to first undergo ex-vivo expansion towards erythroid progenitor cells, which are then used for reprogramming using the CytoTune™-iPS 2.0 Sendai Reprogramming Kit. Resultant iPSC lines are tested to confirm pluripotency, genomic integrity, and stability. Cells are maintained in a feeder-free, xeno-free environment, using fully defined, commercially available reagents. Adoption of this protocol, with heed given to tips provided, allows efficient and robust creation of clinical-grade iPSC cell lines from small volumes of cryopreserved CB.

10.
Elife ; 112022 03 16.
Article in English | MEDLINE | ID: mdl-35293863

ABSTRACT

Organ fibroblasts are essential components of homeostatic and diseased tissues. They participate in sculpting the extracellular matrix, sensing the microenvironment, and communicating with other resident cells. Recent studies have revealed transcriptomic heterogeneity among fibroblasts within and between organs. To dissect the basis of interorgan heterogeneity, we compare the gene expression of murine fibroblasts from different tissues (tail, skin, lung, liver, heart, kidney, and gonads) and show that they display distinct positional and organ-specific transcriptome signatures that reflect their embryonic origins. We demonstrate that expression of genes typically attributed to the surrounding parenchyma by fibroblasts is established in embryonic development and largely maintained in culture, bioengineered tissues and ectopic transplants. Targeted knockdown of key organ-specific transcription factors affects fibroblast functions, in particular genes involved in the modulation of fibrosis and inflammation. In conclusion, our data reveal that adult fibroblasts maintain an embryonic gene expression signature inherited from their organ of origin, thereby increasing our understanding of adult fibroblast heterogeneity. The knowledge of this tissue-specific gene signature may assist in targeting fibrotic diseases in a more precise, organ-specific manner.


Subject(s)
Fibroblasts , Transcriptome , Animals , Fibroblasts/metabolism , Fibrosis , Lung/metabolism , Mice , Skin/metabolism
11.
Cell Mol Gastroenterol Hepatol ; 13(5): 1530-1553.e4, 2022.
Article in English | MEDLINE | ID: mdl-35032693

ABSTRACT

BACKGROUND & AIMS: Pancreatic islet ß-cells are factories for insulin production; however, ectopic expression of insulin also is well recognized. The gallbladder is a next-door neighbor to the developing pancreas. Here, we wanted to understand if gallbladders contain functional insulin-producing cells. METHODS: We compared developing and adult mouse as well as human gallbladder epithelial cells and islets using immunohistochemistry, flow cytometry, enzyme-linked immunosorbent assays, RNA sequencing, real-time polymerase chain reaction, chromatin immunoprecipitation, and functional studies. RESULTS: We show that the epithelial lining of developing, as well as adult, mouse and human gallbladders naturally contain interspersed cells that retain the capacity to actively transcribe, translate, package, and release insulin. We show that human gallbladders also contain functional insulin-secreting cells with the potential to naturally respond to glucose in vitro and in situ. Notably, in a non-obese diabetic (NOD) mouse model of type 1 diabetes, we observed that insulin-producing cells in the gallbladder are not targeted by autoimmune cells. Interestingly, in human gallbladders, insulin splice variants are absent, although insulin splice forms are observed in human islets. CONCLUSIONS: In summary, our biochemical, transcriptomic, and functional data in mouse and human gallbladder epithelial cells collectively show the evolutionary and developmental similarities between gallbladder and the pancreas that allow gallbladder epithelial cells to continue insulin production in adult life. Understanding the mechanisms regulating insulin transcription and translation in gallbladder epithelial cells would help guide future studies in type 1 diabetes therapy.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans , Animals , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Epithelial Cells/metabolism , Gallbladder/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Inbred NOD
13.
Blood ; 139(7): 1080-1097, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34695195

ABSTRACT

In an effort to identify novel drugs targeting fusion-oncogene-induced acute myeloid leukemia (AML), we performed high-resolution proteomic analysis. In AML1-ETO (AE)-driven AML, we uncovered a deregulation of phospholipase C (PLC) signaling. We identified PLCgamma 1 (PLCG1) as a specific target of the AE fusion protein that is induced after AE binding to intergenic regulatory DNA elements. Genetic inactivation of PLCG1 in murine and human AML inhibited AML1-ETO dependent self-renewal programs, leukemic proliferation, and leukemia maintenance in vivo. In contrast, PLCG1 was dispensable for normal hematopoietic stem and progenitor cell function. These findings are extended to and confirmed by pharmacologic perturbation of Ca++-signaling in AML1-ETO AML cells, indicating that the PLCG1 pathway poses an important therapeutic target for AML1-ETO+ leukemic stem cells.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/metabolism , Phospholipase C gamma/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Animals , Cell Self Renewal , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Phospholipase C gamma/genetics , Proteome , RUNX1 Translocation Partner 1 Protein/genetics , Transcriptome , Translocation, Genetic
14.
Stem Cell Res ; 56: 102547, 2021 10.
Article in English | MEDLINE | ID: mdl-34619644

ABSTRACT

Insulin expressing beta cells and glucagon expressing alpha cells are the two most abundant endocrine cell types of the human pancreatic islet. Both alpha and beta cells can be generated in vitro via the differentiation of pluripotent stem cells (PSCs), affording the opportunity to study their ontogeny and to examine their developmental inter-relationship. To aid these analyses, we have generated a PSC line in which insulin expression is reported by GFP and glucagon expression is reported by mCherry. This cell line enables viable isolation of cells expressing each hormone and optimisation of methods that lead to their generation.


Subject(s)
Endocrine Cells , Islets of Langerhans , Pluripotent Stem Cells , Cell Differentiation , Glucagon , Green Fluorescent Proteins , Humans , Insulin , Islets of Langerhans/cytology , Luminescent Proteins , Pancreas , Red Fluorescent Protein
15.
Exp Hematol ; 103: 30-39.e2, 2021 11.
Article in English | MEDLINE | ID: mdl-34437953

ABSTRACT

Exogenous growth factors play an important role in mediating hematopoietic differentiation of human pluripotent stem cells. We explored the role of different factors in early human blood cell production using blast colony formation in methylcellulose as a surrogate assay for yolk sac hematopoiesis. A reporter cell line that read out endothelial (SOX17+) and hematopoietic (RUNX1C+) progenitors facilitated the identification of basic fibroblast growth and vascular endothelial growth factor as critical signals for the progression of mesoderm into endothelium. Bone morphogenetic protein 4 was needed for the subsequent generation of blood from hemogenic endothelium, and this was antagonized by Activin A or high concentrations of the WNT agonist CHIR-99021. Manipulations of the Hedgehog pathway or inhibition of Notch signaling reduced blast colony frequency but did not perturb cell differentiation. These data help to define distinct roles for prerequisite growth factors that commit mesoderm to hemogenic endothelium and subsequently allocate cells to blood lineages.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Fibroblast Growth Factor 2/metabolism , Hematopoiesis , Vascular Endothelial Growth Factor A/metabolism , Yolk Sac/cytology , Cell Line , Endothelium/cytology , Endothelium/metabolism , Humans , Mesoderm/cytology , Mesoderm/metabolism , Yolk Sac/metabolism
16.
Front Endocrinol (Lausanne) ; 12: 635662, 2021.
Article in English | MEDLINE | ID: mdl-33868170

ABSTRACT

Induced pluripotent stem cell (iPSC) technology is increasingly being used to create in vitro models of monogenic human disorders. This is possible because, by and large, the phenotypic consequences of such genetic variants are often confined to a specific and known cell type, and the genetic variants themselves can be clearly identified and controlled for using a standardized genetic background. In contrast, complex conditions such as autoimmune Type 1 diabetes (T1D) have a polygenic inheritance and are subject to diverse environmental influences. Moreover, the potential cell types thought to contribute to disease progression are many and varied. Furthermore, as HLA matching is critical for cell-cell interactions in disease pathogenesis, any model that seeks to test the involvement of particular cell types must take this restriction into account. As such, creation of an in vitro model of T1D will require a system that is cognizant of genetic background and enables the interaction of cells representing multiple lineages to be examined in the context of the relevant environmental disease triggers. In addition, as many of the lineages critical to the development of T1D cannot be easily generated from iPSCs, such models will likely require combinations of cell types derived from in vitro and in vivo sources. In this review we imagine what an ideal in vitro model of T1D might look like and discuss how the required elements could be feasibly assembled using existing technologies. We also examine recent advances towards this goal and discuss potential uses of this technology in contributing to our understanding of the mechanisms underlying this autoimmune condition.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Induced Pluripotent Stem Cells/cytology , Models, Biological , Animals , Antigen-Presenting Cells/cytology , Apoptosis , Autoantibodies , Autoimmune Diseases/metabolism , Cell Differentiation , Dendritic Cells/cytology , Disease Progression , Genetic Predisposition to Disease , Genetic Variation , Humans , In Vitro Techniques , Killer Cells, Natural/cytology , Macrophages/metabolism , Mice , Mice, Inbred NOD , T-Lymphocytes/cytology
17.
Sci Rep ; 11(1): 8315, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859325

ABSTRACT

The identification of cell surface markers specific to pancreatic beta cells is important for both the study of islet biology and for investigating the pathophysiology of diseases in which this cell type is lost or damaged. Following analysis of publicly available RNAseq data, we identified specific integrin subunits, integrin αv and integrin ß5, that were expressed in beta cells. This finding was further elaborated using immunofluorescence analysis of histological sections derived from donor human pancreas. Despite the broad expression of specific integrin subunits, we found that expression of integrin αvß5 heterodimers was restricted to beta cells and that this complex persisted in islet remnants of some type 1 diabetic individuals from which insulin expression had been lost. This study identifies αvß5 heterodimers as a novel cell surface marker of human pancreatic beta cells, a finding that will aid in the identification and characterisation of this important cell type.


Subject(s)
Diabetes Mellitus, Type 1/diagnosis , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Receptors, Vitronectin/metabolism , Adult , Biomarkers/metabolism , Female , Gene Expression , Humans , Insulin/genetics , Insulin/metabolism , Male , Middle Aged , Receptors, Vitronectin/genetics
18.
STAR Protoc ; 1(3): 100130, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377024

ABSTRACT

This protocol offers a detailed procedure for the in vitro differentiation of human pluripotent stem cells (hPSCs) to multipotent hematopoietic progenitors that arise from SOX17+ hemogenic endothelium, mimicking intra-embryonic, HOXA-positive, aorta-gonad mesonephros (AGM) hematopoiesis. The generated endothelium displays transcriptional similarities to cells sorted from human 5-week AGM, and CD45+CD34+RUNX1C+ progenitors share an accessible chromatin profile with adult hematopoietic stem cells and multipotent progenitors. Therefore, this protocol is suitable for the mechanistic study of human multipotent progenitor development and for modeling childhood leukemias. For complete details on the use and execution of this protocol, please refer to Nafria et al. (2020).


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/drug effects , Hematopoietic Stem Cells/cytology , Cell Differentiation/physiology , Gonads/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Humans , Mesonephros/cytology , Models, Biological , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology
19.
Development ; 147(20)2020 10 29.
Article in English | MEDLINE | ID: mdl-33028609

ABSTRACT

The genetic regulatory network controlling early fate choices during human blood cell development are not well understood. We used human pluripotent stem cell reporter lines to track the development of endothelial and haematopoietic populations in an in vitro model of human yolk-sac development. We identified SOX17-CD34+CD43- endothelial cells at day 2 of blast colony development, as a haemangioblast-like branch point from which SOX17-CD34+CD43+ blood cells and SOX17+CD34+CD43- endothelium subsequently arose. Most human blood cell development was dependent on RUNX1. Deletion of RUNX1 only permitted a single wave of yolk sac-like primitive erythropoiesis, but no yolk sac myelopoiesis or aorta-gonad-mesonephros (AGM)-like haematopoiesis. Blocking GFI1 and/or GFI1B activity with a small molecule inhibitor abrogated all blood cell development, even in cell lines with an intact RUNX1 gene. Together, our data define the hierarchical requirements for RUNX1, GFI1 and/or GFI1B during early human haematopoiesis arising from a yolk sac-like SOX17-negative haemogenic endothelial intermediate.


Subject(s)
Blood Cells/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/metabolism , Endothelium/metabolism , Hematopoiesis , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , SOXF Transcription Factors/metabolism , Transcription Factors/metabolism , Yolk Sac/metabolism , Blood Cells/cytology , Cell Differentiation , Cell Lineage , Erythroid Cells/cytology , Erythroid Cells/metabolism , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Humans , Models, Biological , Transcription, Genetic
20.
Stem Cell Res ; 48: 101942, 2020 10.
Article in English | MEDLINE | ID: mdl-32771907

ABSTRACT

To produce in vitro models of human chondrodysplasias caused by dominant missense mutations in TRPV4, we used CRISPR/Cas9 gene editing to introduce two heterozygous patient mutations (p.F273L and p.P799L) into an established control human iPSC line. This control line expressed a fluorescent reporter (tdTomato) at the SOX9 locus to allow real-time monitoring of cartilage differentiation by SOX9 expression. Both TRPV4 mutant iPSC lines had normal karyotypes, expressed pluripotency markers, and could differentiate into cells representative of the three embryonic germ layers. These iPSC lines, with the parental isogenic control, will be used to study TRPV4 chondrodysplasia mechanisms and explore therapeutic approaches.


Subject(s)
Gene Editing , Induced Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , SOX9 Transcription Factor , TRPV Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...