Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 7668, 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-37996457

ABSTRACT

Uncovering the mechanisms regulating hematopoietic specification not only would overcome current limitations related to hematopoietic stem and progenitor cell (HSPC) transplantation, but also advance cellular immunotherapies. However, generating functional human induced pluripotent stem cell (hiPSC)-derived HSPCs and their derivatives has been elusive, necessitating a better understanding of the developmental mechanisms that trigger HSPC specification. Here, we reveal that early activation of the Nod1-Ripk2-NF-kB inflammatory pathway in endothelial cells (ECs) primes them to switch fate towards definitive hemogenic endothelium, a pre-requisite to specify HSPCs. Our genetic and chemical embryonic models show that HSPCs fail to specify in the absence of Nod1 and its downstream kinase Ripk2 due to a failure on hemogenic endothelial (HE) programming, and that small Rho GTPases coordinate the activation of this pathway. Manipulation of NOD1 in a human system of definitive hematopoietic differentiation indicates functional conservation. This work establishes the RAC1-NOD1-RIPK2-NF-kB axis as a critical intrinsic inductor that primes ECs prior to HE fate switch and HSPC specification. Manipulation of this pathway could help derive a competent HE amenable to specify functional patient specific HSPCs and their derivatives for the treatment of blood disorders.


Subject(s)
Hemangioblasts , Induced Pluripotent Stem Cells , Monomeric GTP-Binding Proteins , Humans , Cell Differentiation , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Monomeric GTP-Binding Proteins/metabolism , NF-kappa B/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
2.
Zebrafish ; 19(3): 104-108, 2022 06.
Article in English | MEDLINE | ID: mdl-35704898

ABSTRACT

Cleavage Under Targets and Release Using Nuclease (CUT&RUN) has emerged as a chromatin profiling strategy that excels traditional methods. Although CUT&RUN has been widely utilized in mammalian cells, its use in the zebrafish is at its early stages. In this study, we have developed a protocol to successfully perform CUT&RUN to map transcription factor (TF) binding sites in embryonic, adult tissues, and FACS-sorted zebrafish cells. We also provide a detailed workflow for the identification of predicted TF binding sites that can be utilized in any animal species. Altogether, our strategy will expand this invaluable tool to the zebrafish community, improving the epigenetic resolution that can be achieved in this model organism.


Subject(s)
Chromatin , Zebrafish , Animals , Binding Sites , Endonucleases/metabolism , Mammals/metabolism , Protein Binding , Transcription Factors/metabolism , Zebrafish/metabolism
3.
Blood Adv ; 5(3): 796-811, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33560393

ABSTRACT

Granulin is a pleiotropic protein involved in inflammation, wound healing, neurodegenerative disease, and tumorigenesis. These roles in human health have prompted research efforts to use granulin to treat rheumatoid arthritis and frontotemporal dementia and to enhance wound healing. But how granulin contributes to each of these diverse biological functions remains largely unknown. Here, we have uncovered a new role for granulin during myeloid cell differentiation. We have taken advantage of the tissue-specific segregation of the zebrafish granulin paralogues to assess the functional role of granulin in hematopoiesis without perturbing other tissues. By using our zebrafish model of granulin deficiency, we revealed that during normal and emergency myelopoiesis, myeloid progenitors are unable to terminally differentiate into neutrophils and macrophages in the absence of granulin a (grna), failing to express the myeloid-specific genes cebpa, rgs2, lyz, mpx, mpeg1, mfap4, and apoeb. Functionally, macrophages fail to recruit to the wound, resulting in abnormal healing. Our CUT&RUN experiments identify Pu.1, which together with Irf8, positively regulates grna expression. In vivo imaging and RNA sequencing experiments show that grna inhibits the expression of gata1, leading to the repression of the erythroid program. Importantly, we demonstrated functional conservation between the mammalian granulin and the zebrafish ortholog grna. Our findings uncover a previously unrecognized role for granulin during myeloid cell differentiation, which opens a new field of study that can potentially have an impact on different aspects of human health and expand the therapeutic options for treating myeloid disorders such as neutropenia or myeloid leukemia.


Subject(s)
Neurodegenerative Diseases , Zebrafish , Animals , Carrier Proteins , Extracellular Matrix Proteins , Glycoproteins , Granulins , Hematopoiesis , Humans , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Cell ; 159(5): 1070-1085, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25416946

ABSTRACT

Hematopoietic stem cells (HSCs) underlie the production of blood and immune cells for the lifetime of an organism. In vertebrate embryos, HSCs arise from the unique transdifferentiation of hemogenic endothelium comprising the floor of the dorsal aorta during a brief developmental window. To date, this process has not been replicated in vitro from pluripotent precursors, partly because the full complement of required signaling inputs remains to be determined. Here, we show that TNFR2 via TNF? activates the Notch and NF-?B signaling pathways to establish HSC fate, indicating a requirement for inflammatory signaling in HSC generation. We determine that primitive neutrophils are the major source of TNF?, assigning a role for transient innate immune cells in establishing the HSC program. These results demonstrate that proinflammatory signaling, in the absence of infection, is utilized by the developing embryo to generate the lineal precursors of the adult hematopoietic system.


Subject(s)
Hematopoietic Stem Cells/metabolism , Signal Transduction , Animals , Embryo, Nonmammalian/metabolism , Hematopoiesis , Hematopoietic Stem Cells/cytology , NF-kappa B/metabolism , Neutrophils/metabolism , Receptors, Notch/metabolism , Tumor Necrosis Factor-alpha/metabolism , Zebrafish/metabolism
5.
Blood ; 122(24): 3918-28, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24128862

ABSTRACT

Granulocyte colony-stimulating factor (Gcsf) drives the proliferation and differentiation of granulocytes, monocytes, and macrophages (mφs) from hematopoietic stem and progenitor cells (HSPCs). Analysis of the zebrafish genome indicates the presence of 2 Gcsf ligands, likely resulting from a duplication event in teleost evolution. Although Gcsfa and Gcsfb share low sequence conservation, they share significant similarity in their predicted ligand/receptor interaction sites and structure. Each ligand displays differential temporal expression patterns during embryogenesis and spatial expression patterns in adult animals. To determine the functions of each ligand, we performed loss- and gain-of-function experiments. Both ligands signal through the Gcsf receptor to expand primitive neutrophils and mφs, as well as definitive granulocytes. To further address their functions, we generated recombinant versions and tested them in clonal progenitor assays. These sensitive in vitro techniques indicated similar functional attributes in supporting HSPC growth and differentiation. Finally, in addition to supporting myeloid differentiation, zebrafish Gcsf is required for the specification and proliferation of hematopoietic stem cells, suggesting that Gcsf represents an ancestral cytokine responsible for the broad support of HSPCs. These findings may inform how hematopoietic cytokines evolved following the diversification of teleosts and mammals from a common ancestor.


Subject(s)
Granulocyte Colony-Stimulating Factor/genetics , Hematopoiesis/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic System/embryology , Hematopoietic System/metabolism , In Situ Hybridization , Ligands , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Microscopy, Confocal , Myelopoiesis/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Zebrafish/embryology , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL