LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 43

Search options

  1. Article ; Online: Lymphoid cell development from fetal hematopoietic progenitors and human pluripotent stem cells

    Sun, Shicheng / Wijanarko, Kevin / Liani, Oniko / Strumila, Kathleen / Ng, Elizabeth S. / Elefanty, Andrew G. / Stanley, Edouard G.

    Immunological Reviews. 2023 May, v. 315, no. 1, p. 154-170

    2023  , Page(s) 154–170

    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 ... ...

    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.
    Keywords adaptive immunity ; adults ; bone marrow ; embryogenesis ; fetal development ; hematopoiesis ; humans ; lymphocytes ; ontogeny ; sequence analysis ; stem cells
    Language English
    Dates of publication 2023-05
    Size p. 154-170
    Publishing place John Wiley & Sons, Ltd
    Document type Article ; Online
    Note REVIEW
    ZDB-ID 391796-4
    ISSN 1600-065X ; 0105-2896
    ISSN (online) 1600-065X
    ISSN 0105-2896
    DOI 10.1111/imr.13197
    Database NAL-Catalogue (AGRICOLA)

    More links

    Kategorien

  2. Article ; Online: VEGF, FGF2, and BMP4 regulate transitions of mesoderm to endothelium and blood cells in a human model of yolk sac hematopoiesis.

    Bruveris, Freya F / Ng, Elizabeth S / Stanley, Edouard G / Elefanty, Andrew G

    Experimental hematology

    2021  Volume 103, Page(s) 30–39.e2

    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 ... ...

    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
    MeSH term(s) Bone Morphogenetic Protein 4/metabolism ; Cell Line ; Endothelium/cytology ; Endothelium/metabolism ; Fibroblast Growth Factor 2/metabolism ; Hematopoiesis ; Humans ; Mesoderm/cytology ; Mesoderm/metabolism ; Vascular Endothelial Growth Factor A/metabolism ; Yolk Sac/cytology ; Yolk Sac/metabolism
    Chemical Substances BMP4 protein, human ; Bone Morphogenetic Protein 4 ; VEGFA protein, human ; Vascular Endothelial Growth Factor A ; Fibroblast Growth Factor 2 (103107-01-3)
    Language English
    Publishing date 2021-08-23
    Publishing country Netherlands
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 185107-x
    ISSN 1873-2399 ; 0531-5573 ; 0301-472X
    ISSN (online) 1873-2399
    ISSN 0531-5573 ; 0301-472X
    DOI 10.1016/j.exphem.2021.08.006
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Lymphoid cell development from fetal hematopoietic progenitors and human pluripotent stem cells.

    Sun, Shicheng / Wijanarko, Kevin / Liani, Oniko / Strumila, Kathleen / Ng, Elizabeth S / Elefanty, Andrew G / Stanley, Edouard G

    Immunological reviews

    2023  Volume 315, Issue 1, Page(s) 154–170

    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 ... ...

    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.
    MeSH term(s) Adult ; Humans ; Cell Differentiation ; Pluripotent Stem Cells ; Hematopoietic Stem Cells ; Killer Cells, Natural ; Hematopoiesis
    Language English
    Publishing date 2023-03-20
    Publishing country England
    Document type Journal Article ; Review ; Research Support, Non-U.S. Gov't
    ZDB-ID 391796-4
    ISSN 1600-065X ; 0105-2896
    ISSN (online) 1600-065X
    ISSN 0105-2896
    DOI 10.1111/imr.13197
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Generation of heterozygous (MCRIi030-A-1) and homozygous (MCRIi030-A-2) NR2F2/COUP-TFII knockout human iPSC lines.

    Ferreira, Lucas G A / Cabral-da-Silva, Mauricio C / Pachernegg, Svenja / van den Bergen, Jocelyn A / Robevska, Gorjana / Vlahos, Katerina / Howden, Sara E / Ng, Elizabeth S / Dias-da-Silva, Magnus R / Sinclair, Andrew H / Ayers, Katie L

    Stem cell research

    2024  Volume 76, Page(s) 103374

    Abstract: The NR2F2 gene encodes the transcription factor COUP-TFII, which is upregulated in embryonic mesoderm. Heterozygous variants in NR2F2 cause a spectrum of congenital anomalies including cardiac and gonadal phenotypes. We generated heterozygous (MCRIi030-A- ...

    Abstract The NR2F2 gene encodes the transcription factor COUP-TFII, which is upregulated in embryonic mesoderm. Heterozygous variants in NR2F2 cause a spectrum of congenital anomalies including cardiac and gonadal phenotypes. We generated heterozygous (MCRIi030-A-1) and homozygous (MCRIi030-A-2) NR2F2-knockout induced pluripotent stem cell (iPSC) lines from human fibroblasts using a one-step protocol for CRISPR/Cas9 gene-editing and episomal-based reprogramming. Both iPSC lines exhibited a normal karyotype, typical pluripotent cell morphology, pluripotency marker expression, and the capacity to differentiate into the three embryonic germ layers. These lines will allow us to explore the role of NR2F2 during development and disease.
    MeSH term(s) Humans ; Induced Pluripotent Stem Cells/metabolism ; Heart ; Heterozygote ; Homozygote ; Phenotype ; CRISPR-Cas Systems/genetics ; COUP Transcription Factor II/genetics ; COUP Transcription Factor II/metabolism
    Chemical Substances NR2F2 protein, human ; COUP Transcription Factor II
    Language English
    Publishing date 2024-03-02
    Publishing country England
    Document type Journal Article
    ZDB-ID 2393143-7
    ISSN 1876-7753 ; 1873-5061
    ISSN (online) 1876-7753
    ISSN 1873-5061
    DOI 10.1016/j.scr.2024.103374
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Expression of RUNX1-ETO Rapidly Alters the Chromatin Landscape and Growth of Early Human Myeloid Precursor Cells.

    Nafria, Monica / Keane, Peter / Ng, Elizabeth S / Stanley, Edouard G / Elefanty, Andrew G / Bonifer, Constanze

    Cell reports

    2020  Volume 31, Issue 8, Page(s) 107691

    Abstract: Acute myeloid leukemia (AML) is a hematopoietic malignancy caused by recurrent mutations in genes encoding transcriptional, chromatin, and/or signaling regulators. The t(8;21) translocation generates the aberrant transcription factor RUNX1-ETO (RUNX1- ... ...

    Abstract Acute myeloid leukemia (AML) is a hematopoietic malignancy caused by recurrent mutations in genes encoding transcriptional, chromatin, and/or signaling regulators. The t(8;21) translocation generates the aberrant transcription factor RUNX1-ETO (RUNX1-RUNX1T1), which by itself is insufficient to cause disease. t(8;21) AML patients show extensive chromatin reprogramming and have acquired additional mutations. Therefore, the genomic and developmental effects directly and solely attributable to RUNX1-ETO expression are unclear. To address this, we employ a human embryonic stem cell differentiation system capable of forming definitive myeloid progenitor cells to express RUNX1-ETO in an inducible fashion. Induction of RUNX1-ETO causes extensive chromatin reprogramming by interfering with RUNX1 binding, blocks differentiation, and arrests cellular growth, whereby growth arrest is reversible following RUNX1-ETO removal. Single-cell gene expression analyses show that RUNX1-ETO induction alters the differentiation of early myeloid progenitors, but not of other progenitor types, indicating that oncoprotein-mediated transcriptional reprogramming is highly target cell specific.
    MeSH term(s) Cell Differentiation ; Cell Proliferation ; Chromatin/metabolism ; Core Binding Factor Alpha 2 Subunit/metabolism ; Humans ; Myeloid Progenitor Cells/metabolism
    Chemical Substances Chromatin ; Core Binding Factor Alpha 2 Subunit ; RUNX1 protein, human
    Language English
    Publishing date 2020-05-26
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2649101-1
    ISSN 2211-1247 ; 2211-1247
    ISSN (online) 2211-1247
    ISSN 2211-1247
    DOI 10.1016/j.celrep.2020.107691
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Modeling human skeletal development using human pluripotent stem cells.

    Lamandé, Shireen R / Ng, Elizabeth S / Cameron, Trevor L / Kung, Louise H W / Sampurno, Lisa / Rowley, Lynn / Lilianty, Jinia / Patria, Yudha Nur / Stenta, Tayla / Hanssen, Eric / Bell, Katrina M / Saxena, Ritika / Stok, Kathryn S / Stanley, Edouard G / Elefanty, Andrew G / Bateman, John F

    Proceedings of the National Academy of Sciences of the United States of America

    2023  Volume 120, Issue 19, Page(s) e2211510120

    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 ... ...

    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.
    MeSH term(s) Humans ; Cartilage/metabolism ; Chondrocytes/metabolism ; Cell Differentiation ; Osteoblasts ; Induced Pluripotent Stem Cells/metabolism
    Language English
    Publishing date 2023-05-01
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 209104-5
    ISSN 1091-6490 ; 0027-8424
    ISSN (online) 1091-6490
    ISSN 0027-8424
    DOI 10.1073/pnas.2211510120
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Human pluripotent stem cell-derived macrophages host Mycobacterium abscessus infection.

    Sun, Shicheng / See, Michael / Nim, Hieu T / Strumila, Kathleen / Ng, Elizabeth S / Hidalgo, Alejandro / Ramialison, Mirana / Sutton, Philip / Elefanty, Andrew G / Sarkar, Sohinee / Stanley, Edouard G

    Stem cell reports

    2022  Volume 17, Issue 9, Page(s) 2156–2166

    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 ...

    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.
    MeSH term(s) Humans ; Macrophages/metabolism ; Mycobacterium ; Mycobacterium Infections, Nontuberculous/metabolism ; Mycobacterium Infections, Nontuberculous/microbiology ; Mycobacterium abscessus ; Pluripotent Stem Cells
    Language English
    Publishing date 2022-08-18
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2720528-9
    ISSN 2213-6711 ; 2213-6711
    ISSN (online) 2213-6711
    ISSN 2213-6711
    DOI 10.1016/j.stemcr.2022.07.013
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Mimicry of embryonic circulation enhances the hoxa hemogenic niche and human blood development.

    Li, Jingjing / Lao, Osmond / Bruveris, Freya F / Wang, Liyuan / Chaudry, Kajal / Yang, Ziqi / Farbehi, Nona / Ng, Elizabeth S / Stanley, Edouard G / Harvey, Richard P / Elefanty, Andrew G / Nordon, Robert E

    Cell reports

    2022  Volume 40, Issue 11, Page(s) 111339

    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 ... ...

    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
    MeSH term(s) Adult ; Antigens, CD34 ; Cell Differentiation ; Hematopoiesis ; Hematopoietic Stem Cells ; Humans ; Mesonephros ; Yolk Sac
    Chemical Substances Antigens, CD34
    Language English
    Publishing date 2022-09-09
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2649101-1
    ISSN 2211-1247 ; 2211-1247
    ISSN (online) 2211-1247
    ISSN 2211-1247
    DOI 10.1016/j.celrep.2022.111339
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Rapid training of healthcare staff for protected cardiopulmonary resuscitation in the COVID-19 pandemic.

    Foong, Theng W / Hui Ng, Elizabeth S / Wee Khoo, Cicely Y / Ashokka, Balakrishnan / Khoo, Deborah / Agrawal, Rohit

    British journal of anaesthesia

    2020  Volume 125, Issue 2, Page(s) e257–e259

    MeSH term(s) COVID-19 ; Cardiopulmonary Resuscitation/education ; Coronavirus Infections/therapy ; Health Personnel ; Humans ; Masks ; Pandemics ; Pneumonia, Viral/therapy ; Protective Clothing ; Simulation Training
    Keywords covid19
    Language English
    Publishing date 2020-05-06
    Publishing country England
    Document type Letter
    ZDB-ID 80074-0
    ISSN 1471-6771 ; 0007-0912
    ISSN (online) 1471-6771
    ISSN 0007-0912
    DOI 10.1016/j.bja.2020.04.081
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article: WNT9A Is a Conserved Regulator of Hematopoietic Stem and Progenitor Cell Development.

    Richter, Jenna / Stanley, Edouard G / Ng, Elizabeth S / Elefanty, Andrew G / Traver, David / Willert, Karl

    Genes

    2018  Volume 9, Issue 2

    Abstract: Hematopoietic stem cells (HSCs) differentiate into all cell types of the blood and can be used therapeutically to treat hematopoietic cancers and disorders. Despite decades of research, it is not yet possible to derive therapy-grade HSCs from pluripotent ...

    Abstract Hematopoietic stem cells (HSCs) differentiate into all cell types of the blood and can be used therapeutically to treat hematopoietic cancers and disorders. Despite decades of research, it is not yet possible to derive therapy-grade HSCs from pluripotent precursors. Analysis of HSC development in model organisms has identified some of the molecular cues that are necessary to instruct hematopoiesis in vivo, including Wnt9A, which is required during an early time window in zebrafish development. Although bona fide HSCs cannot be derived in vitro, it is possible to model human hematopoietic progenitor development by differentiating human pluripotent stem cells to hematopoietic cells. Herein, we modulate WNT9A expression during the in vitro differentiation of human embryonic stem cells to hematopoietic progenitor cells and demonstrate that WNT9A also regulates human hematopoietic progenitor cell development in vitro. Overexpression of WNT9A only impacts differentiation to CD34⁺/CD45⁺ cells during early time windows and does so in a dose-dependent manner. The cells that receive the Wnt signal-not the cells that secrete WNT9A-differentiate most efficiently to hematopoietic progenitors; this mimics the paracrine action of Wnt9a during in vivo hematopoiesis. Taken together, these data indicate that WNT9A is a conserved regulator of zebrafish and human hematopoietic development.
    Language English
    Publishing date 2018-01-29
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2527218-4
    ISSN 2073-4425
    ISSN 2073-4425
    DOI 10.3390/genes9020066
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top