Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Leukoc Biol ; 116(1): 118-131, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38417030

ABSTRACT

Neutrophils are rapidly recruited to sites of infection and are critical for pathogen clearance. Therapeutic use of primary neutrophils has been limited, as they have a short lifespan and are not amenable to genetic manipulation. Human induced pluripotent stem cells (iPSCs) can provide a robust source of neutrophils for infusion and are genetically tractable. However, current work has indicated that dampened intracellular signaling limits iPSC-derived neutrophil (iNeutrophil) cellular activation and antimicrobial response. Here, we show that protein tyrosine phosphatase 1B (PTP1B) inhibits intracellular signaling and dampens iNeutrophil effector function. Deletion of the PTP1B phosphatase increased PI3K and ERK signaling and was associated with increased F-actin polymerization, cell migration, and phagocytosis. In contrast, other effector functions like NETosis and reactive oxygen species production were reduced. PTP1B-deficient neutrophils were more responsive to Aspergillus fumigatus and displayed rapid recruitment and control of hyphal growth. Accordingly, depletion of PTP1B increased production of inflammatory factors including the neutrophil chemokine interleukin-8. Taken together, these findings suggest that PTP1B limits iNeutrophil motility and antimicrobial function.


Subject(s)
Cell Movement , Induced Pluripotent Stem Cells , Neutrophils , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Neutrophils/metabolism , Neutrophils/immunology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Aspergillus fumigatus , Phagocytosis , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Extracellular Traps/metabolism , Extracellular Traps/immunology , Actins/metabolism
2.
iScience ; 26(5): 106621, 2023 May 19.
Article in English | MEDLINE | ID: mdl-37250328

ABSTRACT

Hemogenic endothelium (HE) is the main source of blood cells in the embryo. To improve blood manufacturing from human pluripotent stem cells (hPSCs), it is essential to define the molecular determinants that enhance HE specification and promote development of the desired blood lineage from HE. Here, using SOX18-inducible hPSCs, we revealed that SOX18 forced expression at the mesodermal stage, in contrast to its homolog SOX17, has minimal effects on arterial specification of HE, expression of HOXA genes and lymphoid differentiation. However, forced expression of SOX18 in HE during endothelial-to-hematopoietic transition (EHT) greatly increases NK versus T cell lineage commitment of hematopoietic progenitors (HPs) arising from HE predominantly expanding CD34+CD43+CD235a/CD41a-CD45- multipotent HPs and altering the expression of genes related to T cell and Toll-like receptor signaling. These studies improve our understanding of lymphoid cell specification during EHT and provide a new tool for enhancing NK cell production from hPSCs for immunotherapies.

3.
Methods Mol Biol ; 2429: 103-124, 2022.
Article in English | MEDLINE | ID: mdl-35507158

ABSTRACT

Endothelial-to-hematopoietic transition (EHT) is a unique morphogenic event in which flat, adherent hemogenic endothelial (HE) cells acquire round, non-adherent blood cell morphology. Investigating the mechanisms of EHT is critical for understanding the development of hematopoietic stem cells (HSCs) and the entirety of the adult immune system, and advancing technologies for manufacturing blood cells from human pluripotent stem cells (hPSCs). Here we describe a protocol to (a) generate and isolate subsets of HE from hPSCs, (b) assess EHT and hematopoietic potential of HE subsets in bulk cultures and at the single-cell level, and (c) evaluate the role of NOTCH signaling during HE specification and EHT. The generation of HE from hPSCs and EHT bulk cultures are performed in xenogen- and feeder-free system, providing the unique advantage of being able to investigate the role of individual signaling factors during EHT and the definitive lympho-myeloid cell specification from hPSCs.


Subject(s)
Hemangioblasts , Pluripotent Stem Cells , Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells , Humans
4.
Cell Rep ; 34(7): 108758, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33596423

ABSTRACT

SOX17 has been implicated in arterial specification and the maintenance of hematopoietic stem cells (HSCs) in the murine embryo. However, knowledge about molecular pathways and stage-specific effects of SOX17 in humans remains limited. Here, using SOX17-knockout and SOX17-inducible human pluripotent stem cells (hPSCs), paired with molecular profiling studies, we reveal that SOX17 is a master regulator of HOXA and arterial programs in hemogenic endothelium (HE) and is required for the specification of HE with robust lympho-myeloid potential and DLL4+CXCR4+ phenotype resembling arterial HE at the sites of HSC emergence. Along with the activation of NOTCH signaling, SOX17 directly activates CDX2 expression, leading to the upregulation of the HOXA cluster genes. Since deficiencies in HOXA and NOTCH signaling contribute to the impaired in vivo engraftment of hPSC-derived hematopoietic cells, the identification of SOX17 as a key regulator linking arterial and HOXA programs in HE may help to program HSC fate from hPSCs.


Subject(s)
Hematopoiesis/genetics , Homeodomain Proteins/metabolism , SOXF Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism
5.
Front Oncol ; 10: 585151, 2020.
Article in English | MEDLINE | ID: mdl-33363015

ABSTRACT

Numerous recurrent genetic mutations are known to occur in acute myeloid leukemia (AML). Among these common mutations, Fms-like tyrosine kinase 3 remains as one of the most frequently mutated genes in AML. We observed apparent marrow expansion of megakaryocytes in three out of six patients with Flt3-mutated AML following treatment with a recently FDA-approved Flt3 inhibitor, gilteritinib which possesses activity against internal tandem duplication and tyrosine kinase domain Flt3 mutations and also inhibits tyrosine kinase AXL. To assess whether biopsy findings can be attributed to promotion of megakaryocytic (Mk) differentiation with gilteritinib, we devised a cellular assay by overexpressing double mutated Flt3-ITDY591F/Y919F in chronic myeloid leukemia cell line K562 to study Mk differentiation in the presence of Flt3 and AXL inhibitors with non-mutually exclusive mechanisms. These experiments demonstrated the lack of direct effect Flt3 inhibitors gilteritinib and quizartinib on megakaryocytic differentiation at either transcriptional or phenotypic levels, and highlighted antileukemic effects of AXL receptor tyrosine kinase inhibitor and its potential role in megakaryocytic development.

6.
Cell Death Dis ; 9(11): 1136, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30429452

ABSTRACT

The regulation of osteogenesis is important for bone formation and fracture healing. Despite advances in understanding the molecular mechanisms of osteogenesis, crucial modulators in this process are not well-characterized. Here we demonstrate that suppression of signal transducer and activator of transcription 5A (STAT5A) activates distal-less homeobox 5 (DLX5) in human bone marrow-derived stromal cells (hBMSCs) and enhances osteogenesis in vitro and in vivo. We show that STAT5A negatively regulates expression of Dlx5 in vitro and that STAT5A deletion results in increased trabecular and cortical bone mass and bone mineral density in mice. Additionally, STAT5A deletion prevents age-related bone loss. In a murine fracture model, STAT5A deletion was found to significantly enhance bone remodeling by stimulating the formation of a fracture callus. Our findings indicate that STAT5A inhibition enhances bone formation by promoting osteogenesis of BMSCs.


Subject(s)
Fractures, Bone/genetics , Homeodomain Proteins/genetics , Osteoblasts/metabolism , Osteogenesis/genetics , Osteoporosis/genetics , STAT5 Transcription Factor/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Bone Density/genetics , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Femur/injuries , Femur/metabolism , Fracture Healing/genetics , Fractures, Bone/metabolism , Fractures, Bone/pathology , Fractures, Bone/therapy , Gene Expression Regulation , Homeodomain Proteins/metabolism , Humans , Integrin-Binding Sialoprotein/genetics , Integrin-Binding Sialoprotein/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoporosis/metabolism , Osteoporosis/pathology , Osteoporosis/prevention & control , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism
7.
Curr Protoc Stem Cell Biol ; 47(1): e63, 2018 11.
Article in English | MEDLINE | ID: mdl-30281932

ABSTRACT

Human pluripotent stem cells (hPSCs) emerged as an important tool to investigate human development and disease. These studies often require genetically engineering hPSCs to stably express a transgene, which remains functional in various hPSC progeny. PiggyBac transposon is a highly effective and technically simple vector system with large cargo space available for permanent gene delivery. This unit describes the use of PiggyBac transposons to genetically engineer hPSCs to introduce conditionally expressed transgene or reporter to effectively monitor gene expression during differentiation. Both methods enable robust generation of stable hPSC lines within 1 month. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Cell Culture Techniques/methods , DNA Transposable Elements , Genetic Engineering/methods , Pluripotent Stem Cells/cytology , Cells, Cultured , Gene Expression , Genetic Vectors , Humans , Transgenes
8.
Stem Cell Reports ; 11(1): 197-211, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29861167

ABSTRACT

The transcriptional factor GATA2 is required for blood and hematopoietic stem cell formation during the hemogenic endothelium (HE) stage of development in the embryo. However, it is unclear if GATA2 controls HE lineage specification or if it solely regulates endothelial-to-hematopoietic transition (EHT). To address this problem, we innovated a unique system, which involved generating GATA2 knockout human embryonic stem cell (hESC) lines with conditional GATA2 expression (iG2-/- hESCs). We demonstrated that GATA2 activity is not required for VE-cadherin+CD43-CD73+ non-HE or VE-cadherin+CD43-CD73- HE generation and subsequent HE diversification into DLL4+ arterial and DLL4- non-arterial lineages. However, GATA2 is primarily needed for HE to undergo EHT. Forced expression of GATA2 in non-HE failed to induce blood formation. The lack of GATA2 requirement for generation of HE and non-HE indicates the critical role of GATA2-independent pathways in specification of these two distinct endothelial lineages.


Subject(s)
Cell Differentiation/genetics , GATA2 Transcription Factor/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , GATA2 Transcription Factor/metabolism , Gene Editing , Gene Expression Profiling , Gene Knockout Techniques , Gene Targeting , Hemangioblasts/cytology , Hemangioblasts/metabolism , Humans , Leukocytes/cytology , Leukocytes/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism
9.
Nat Commun ; 9(1): 1828, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29739946

ABSTRACT

NOTCH signaling is required for the arterial specification and formation of hematopoietic stem cells (HSCs) and lympho-myeloid progenitors in the embryonic aorta-gonad-mesonephros region and extraembryonic vasculature from a distinct lineage of vascular endothelial cells with hemogenic potential. However, the role of NOTCH signaling in hemogenic endothelium (HE) specification from human pluripotent stem cell (hPSC) has not been studied. Here, using a chemically defined hPSC differentiation system combined with the use of DLL1-Fc and DAPT to manipulate NOTCH, we discover that NOTCH activation in hPSC-derived immature HE progenitors leads to formation of CD144+CD43-CD73-DLL4+Runx1 + 23-GFP+ arterial-type HE, which requires NOTCH signaling to undergo endothelial-to-hematopoietic transition and produce definitive lympho-myeloid and erythroid cells. These findings demonstrate that NOTCH-mediated arterialization of HE is an essential prerequisite for establishing definitive lympho-myeloid program and suggest that exploring molecular pathways that lead to arterial specification may aid in vitro approaches to enhance definitive hematopoiesis from hPSCs.


Subject(s)
Arteries/cytology , Endothelium, Vascular/cytology , Hemangioblasts/cytology , Hematopoiesis , Neovascularization, Physiologic , Pluripotent Stem Cells/cytology , Receptors, Notch/metabolism , Signal Transduction , Animals , Antigens, CD/immunology , Arteries/metabolism , Calcium-Binding Proteins , Cell Differentiation , Cell Line , Cell Lineage , Cell Tracking/instrumentation , Coculture Techniques , Embryo, Mammalian/cytology , Endothelium, Vascular/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/immunology , Hemangioblasts/immunology , Hematopoietic Stem Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Membrane Proteins/metabolism , Mice , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/immunology , Pluripotent Stem Cells/immunology
10.
Cell Rep ; 23(8): 2467-2481, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791856

ABSTRACT

Understanding the pathways guiding the development of definitive hematopoiesis with lymphoid potential is essential for advancing human pluripotent stem cell (hPSC) technologies for the treatment of blood diseases and immunotherapies. In the embryo, lymphoid progenitors and hematopoietic stem cells (HSCs) arise from hemogenic endothelium (HE) lining arteries but not veins. Here, we show that activation of the arterial program through ETS1 overexpression or by modulating MAPK/ERK signaling pathways at the mesodermal stage of development dramatically enhanced the formation of arterial-type HE expressing DLL4 and CXCR4. Blood cells generated from arterial HE were more than 100-fold enriched in T cell precursor frequency and possessed the capacity to produce B lymphocytes and red blood cells expressing high levels of BCL11a and ß-globin. Together, these findings provide an innovative strategy to aid in the generation of definitive lymphomyeloid progenitors and lymphoid cells from hPSCs for immunotherapy through enhancing arterial programming of HE.


Subject(s)
Arteries/metabolism , Hemangioblasts/metabolism , Lymphocytes/metabolism , Body Patterning , Hematopoiesis , Human Embryonic Stem Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Membrane Proteins/metabolism , Mesoderm/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Receptors, CXCR4/metabolism , Receptors, Notch/metabolism , SOXF Transcription Factors/metabolism , Transcription, Genetic , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL