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










Database
Type of study
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(20): e2210058120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155858

ABSTRACT

Notch signaling is essential for the emergence of definitive hematopoietic stem cells (HSCs) in the embryo and their development in the fetal liver niche. However, how Notch signaling is activated and which fetal liver cell type provides the ligand for receptor activation in HSCs is unknown. Here we provide evidence that endothelial Jagged1 (Jag1) has a critical early role in fetal liver vascular development but is not required for hematopoietic function during fetal HSC expansion. We demonstrate that Jag1 is expressed in many hematopoietic cells in the fetal liver, including HSCs, and that its expression is lost in adult bone marrow HSCs. Deletion of hematopoietic Jag1 does not affect fetal liver development; however, Jag1-deficient fetal liver HSCs exhibit a significant transplantation defect. Bulk and single-cell transcriptomic analysis of HSCs during peak expansion in the fetal liver indicates that loss of hematopoietic Jag1 leads to the downregulation of critical hematopoietic factors such as GATA2, Mllt3, and HoxA7, but does not perturb Notch receptor expression. Ex vivo activation of Notch signaling in Jag1-deficient fetal HSCs partially rescues the functional defect in a transplant setting. These findings indicate a new fetal-specific niche that is based on juxtracrine hematopoietic Notch signaling and reveal Jag1 as a fetal-specific niche factor essential for HSC function.


Subject(s)
Fetus , Hematopoietic Stem Cells , Adult , Humans , Endothelium/metabolism , Fetus/metabolism , Hematopoietic Stem Cells/metabolism , Liver/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism
2.
JCI Insight ; 7(9)2022 05 09.
Article in English | MEDLINE | ID: mdl-35349492

ABSTRACT

Long-term impairment in T cell-mediated adaptive immunity is a major clinical obstacle following treatment of blood disorders with hematopoietic stem cell transplantation. Although T cell development in the thymus has been extensively characterized, there are significant gaps in our understanding of prethymic processes that influence early T cell potential. We have uncovered a Notch/IL-21 signaling axis in bone marrow common lymphoid progenitor (CLP) cells. IL-21 receptor expression was driven by Notch activation in CLPs, and in vivo treatment with IL-21 induced Notch-dependent CLP proliferation. Taking advantage of this potentially novel signaling axis, we generated T cell progenitors ex vivo, which improved repopulation of the thymus and peripheral lymphoid organs of mice in an allogeneic transplant model. Importantly, Notch and IL-21 activation were equally effective in the priming and expansion of human cord blood cells toward the T cell fate, confirming the translational potential of the combined treatment.


Subject(s)
Hematopoietic Stem Cells , T-Lymphocytes , Animals , Bone Marrow/metabolism , Hematopoietic Stem Cells/metabolism , Interleukins , Mice , Signal Transduction
3.
Front Immunol ; 12: 723055, 2021.
Article in English | MEDLINE | ID: mdl-34394130

ABSTRACT

Lifelong mammalian hematopoiesis requires continuous generation of mature blood cells that originate from Hematopoietic Stem and Progenitor Cells (HSPCs) situated in the post-natal Bone Marrow (BM). The BM microenvironment is inherently complex and extensive studies have been devoted to identifying the niche that maintains HSPC homeostasis and supports hematopoietic potential. The Notch signaling pathway is required for the emergence of the definitive Hematopoietic Stem Cell (HSC) during embryonic development, but its role in BM HSC homeostasis is convoluted. Recent work has begun to explore novel roles for the Notch signaling pathway in downstream progenitor populations. In this review, we will focus an important role for Notch signaling in the establishment of a T cell primed sub-population of Common Lymphoid Progenitors (CLPs). Given that its activation mechanism relies primarily on cell-to-cell contact, Notch signaling is an ideal means to investigate and define a novel BM lymphopoietic niche. We will discuss how new genetic model systems indicate a pre-thymic, BM-specific role for Notch activation in early T cell development and what this means to the paradigm of lymphoid lineage commitment. Lastly, we will examine how leukemic T-cell acute lymphoblastic leukemia (T-ALL) blasts take advantage of Notch and downstream lymphoid signals in the pathological BM niche.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cells/metabolism , Lymphopoiesis/physiology , Osteoblasts/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Bone Marrow/pathology , Cell Differentiation/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/pathology , Homeostasis , Humans , Osteoblasts/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction/physiology , Tumor Microenvironment
4.
Cell Rep ; 26(11): 2942-2954.e5, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30865885

ABSTRACT

Lung alveolar type I cells (AT1) and alveolar type II cells (AT2) regulate the structural integrity and function of alveoli. AT1, covering ∼95% of the surface area, are responsible for gas exchange, whereas AT2 serve multiple functions, including alveolar repair through proliferation and differentiation into AT1. However, the signaling mechanisms for alveolar repair remain unclear. Here, we demonstrate, in Pseudomonas aeruginosa-induced acute lung injury in mice, that non-canonical Notch ligand Dlk1 (delta-like 1 homolog) is essential for AT2-to-AT1 differentiation. Notch signaling was activated in AT2 at the onset of repair but later suppressed by Dlk1. Deletion of Dlk1 in AT2 induced persistent Notch activation, resulting in stalled transition to AT1 and accumulation of an intermediate cell population that expressed low levels of both AT1 and AT2 markers. Thus, Dlk1 expression leads to precisely timed inhibition of Notch signaling and activates AT2-to-AT1 differentiation, leading to alveolar repair.


Subject(s)
Alveolar Epithelial Cells/metabolism , Calcium-Binding Proteins/metabolism , Pneumonia, Bacterial/metabolism , Pseudomonas Infections/metabolism , Receptors, Notch/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/microbiology , Animals , Calcium-Binding Proteins/genetics , Cell Differentiation , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Regeneration , Signal Transduction
5.
Haematologica ; 104(11): 2164-2177, 2019 11.
Article in English | MEDLINE | ID: mdl-30923091

ABSTRACT

Loss-of-function studies have determined that Notch signaling is essential for hematopoietic and endothelial development. By deleting a single allele of the Notch1 transcriptional activation domain we generated viable, post-natal mice exhibiting hypomorphic Notch signaling. These heterozygous mice, which lack only one copy of the transcriptional activation domain, appear normal and have no endothelial or hematopoietic phenotype, apart from an inherent, cell-autonomous defect in T-cell lineage development. Following chemotherapy, these hypomorphs exhibited severe pancytopenia, weight loss and morbidity. This phenotype was confirmed in an endothelial-specific, loss-of-function Notch1 model system. Ang1, secreted by hematopoietic progenitors after damage, activated endothelial Tie2 signaling, which in turn enhanced expression of Notch ligands and potentiated Notch1 receptor activation. In our heterozygous, hypomorphic model system, the mutant protein that lacks the Notch1 transcriptional activation domain accumulated in endothelial cells and interfered with optimal activity of the wildtype Notch1 transcriptional complex. Failure of the hypomorphic mutant to efficiently drive transcription of key gene targets such as Hes1 and Myc prolonged apoptosis and limited regeneration of the bone marrow niche. Thus, basal Notch1 signaling is sufficient for niche development, but robust Notch activity is required for regeneration of the bone marrow endothelial niche and hematopoietic recovery.


Subject(s)
Cellular Microenvironment , Endothelial Cells/physiology , Receptor, Notch1/metabolism , Receptor, TIE-2/metabolism , Regeneration , Signal Transduction , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Cellular Microenvironment/drug effects , Endothelial Cells/drug effects , Fluorouracil/pharmacology , Gamma Rays/adverse effects , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , Pancytopenia/etiology , Pancytopenia/metabolism , Pancytopenia/pathology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...