Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Nat Immunol ; 24(3): 474-486, 2023 03.
Article in English | MEDLINE | ID: mdl-36703005

ABSTRACT

The cross-talk between thymocytes and thymic stromal cells is fundamental for T cell development. In humans, intrathymic development of dendritic cells (DCs) is evident but its physiological significance is unknown. Here we showed that DC-biased precursors depended on the expression of the transcription factor IRF8 to express the membrane-bound precursor form of the cytokine TNF (tmTNF) to promote differentiation of thymus seeding hematopoietic progenitors into T-lineage specified precursors through activation of the TNF receptor (TNFR)-2 instead of TNFR1. In vitro recapitulation of TNFR2 signaling by providing low-density tmTNF or a selective TNFR2 agonist enhanced the generation of human T cell precursors. Our study shows that, in addition to mediating thymocyte selection and maturation, DCs function as hematopoietic stromal support for the early stages of human T cell development and provide proof of concept that selective targeting of TNFR2 can enhance the in vitro generation of T cell precursors for clinical application.


Subject(s)
Dendritic Cells , Receptors, Tumor Necrosis Factor, Type II , Humans , Cell Differentiation , Cell Lineage , Interferon Regulatory Factors/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Thymus Gland/metabolism , Tumor Necrosis Factors/metabolism
2.
Nat Immunol ; 21(10): 1280-1292, 2020 10.
Article in English | MEDLINE | ID: mdl-32719521

ABSTRACT

The development of TCRαß and TCRγδ T cells comprises a step-wise process in which regulatory events control differentiation and lineage outcome. To clarify these mechanisms, we employed RNA-sequencing, ATAC-sequencing and ChIPmentation on well-defined thymocyte subsets that represent the continuum of human T cell development. The chromatin accessibility dynamics show clear stage specificity and reveal that human T cell-lineage commitment is marked by GATA3- and BCL11B-dependent closing of PU.1 sites. A temporary increase in H3K27me3 without open chromatin modifications is unique for ß-selection, whereas emerging γδ T cells, which originate from common precursors of ß-selected cells, show large chromatin accessibility changes due to strong T cell receptor (TCR) signaling. Furthermore, we unravel distinct chromatin landscapes between CD4+ and CD8+ αß-lineage cells that support their effector functions and reveal gene-specific mechanisms that define mature T cells. This resource provides a framework for studying gene regulatory mechanisms that drive normal and malignant human T cell development.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/physiology , Thymocytes/physiology , Cell Differentiation , Cell Lineage , Cells, Cultured , Chromatin/metabolism , Clonal Selection, Antigen-Mediated , Epigenesis, Genetic , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Lymphocyte Activation , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Analysis, RNA , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
Immunity ; 52(6): 1088-1104.e6, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32304633

ABSTRACT

During postnatal life, thymopoiesis depends on the continuous colonization of the thymus by bone-marrow-derived hematopoietic progenitors that migrate through the bloodstream. The current understanding of the nature of thymic immigrants is largely based on data from pre-clinical models. Here, we employed single-cell RNA sequencing (scRNA-seq) to examine the immature postnatal thymocyte population in humans. Integration of bone marrow and peripheral blood precursor datasets identified two putative thymus seeding progenitors that varied in expression of CD7; CD10; and the homing receptors CCR7, CCR9, and ITGB7. Whereas both precursors supported T cell development, only one contributed to intrathymic dendritic cell (DC) differentiation, predominantly of plasmacytoid dendritic cells. Trajectory inference delineated the transcriptional dynamics underlying early human T lineage development, enabling prediction of transcription factor (TF) modules that drive stage-specific steps of human T cell development. This comprehensive dataset defines the expression signature of immature human thymocytes and provides a resource for the further study of human thymopoiesis.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , RNA, Small Cytoplasmic/genetics , Thymocytes/cytology , Thymocytes/metabolism , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunophenotyping , Single-Cell Analysis , Thymocytes/immunology , Transcriptome
4.
Blood ; 136(3): 288-298, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32350509

ABSTRACT

Natural killer (NK) cells are important in the immune defense against tumor cells and pathogens, and they regulate other immune cells by cytokine secretion. Although murine NK cell biology has been extensively studied, knowledge about transcriptional circuitries controlling human NK cell development and maturation is limited. By generating ETS1-deficient human embryonic stem cells and by expressing the dominant-negative ETS1 p27 isoform in cord blood hematopoietic progenitor cells, we show that the transcription factor ETS1 is critically required for human NK cell differentiation. Genome-wide transcriptome analysis determined by RNA-sequencing combined with chromatin immunoprecipitation-sequencing analysis reveals that human ETS1 directly induces expression of key transcription factors that control NK cell differentiation (ie, E4BP4, TXNIP, TBET, GATA3, HOBIT, BLIMP1). In addition, ETS1 regulates expression of genes involved in apoptosis and NK cell activation. Our study provides important molecular insights into the role of ETS1 as an important regulator of human NK cell development and terminal differentiation.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation/immunology , Human Embryonic Stem Cells/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Proto-Oncogene Protein c-ets-1/immunology , Apoptosis/genetics , Apoptosis/immunology , Cell Differentiation/genetics , Cell Line , Gene Expression Profiling , Genome-Wide Association Study , Human Embryonic Stem Cells/cytology , Humans , Killer Cells, Natural/cytology , Protein Isoforms/genetics , Protein Isoforms/immunology , Proto-Oncogene Protein c-ets-1/genetics
5.
Cytotherapy ; 24(2): 213-222, 2022 02.
Article in English | MEDLINE | ID: mdl-34696961

ABSTRACT

Messenger RNA (mRNA) has become a promising tool in therapeutic cancer vaccine strategies. Owing to its flexible design and rapid production, mRNA is an attractive antigen delivery format for cancer vaccines targeting mutated peptides expressed in a tumor-the so-called neoantigens. These neoantigens are rarely shared between patients, and inclusion of these antigens in a vaccine requires the production of individual batches of patient-tailored mRNA. The authors have developed MIDRIXNEO, a personalized mRNA-loaded dendritic cell vaccine targeting tumor neoantigens, which is currently being evaluated in a phase 1 clinical study in lung cancer patients. To facilitate this study, the authors set up a Good Manufacturing Practice (GMP)-compliant production process for the manufacture of small batches of personalized neoantigen-encoding mRNA. In this article, the authors describe the complete mRNA production process and the extensive quality assessment to which the mRNA is subjected. Validation runs have shown that the process delivers mRNA of reproducible, high quality. This process is now successfully applied for the production of neoantigen-encoding mRNA for the clinical evaluation of MIDRIXNEO. To the authors' knowledge, this is the first time that a GMP-based production process of patient-tailored neoantigen mRNA has been described.


Subject(s)
Cancer Vaccines , Lung Neoplasms , Neoplasms , Antigens, Neoplasm/genetics , Humans , Immunotherapy , Neoplasms/genetics , Neoplasms/therapy , Peptides , RNA, Messenger/genetics
6.
EMBO Rep ; 21(5): e49006, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32255245

ABSTRACT

γδ and αß T cells have unique roles in immunity and both originate in the thymus from T-lineage committed precursors through distinct but unclear mechanisms. Here, we show that Notch1 activation is more stringently required for human γδ development compared to αß-lineage differentiation and performed paired mRNA and miRNA profiling across 11 discrete developmental stages of human T cell development in an effort to identify the potential Notch1 downstream mechanism. Our data suggest that the miR-17-92 cluster is a Notch1 target in immature thymocytes and that miR-17 can restrict BCL11B expression in these Notch-dependent T cell precursors. We show that enforced miR-17 expression promotes human γδ T cell development and, consistently, that BCL11B is absolutely required for αß but less for γδ T cell development. This study suggests that human γδ T cell development is mediated by a stage-specific Notch-driven negative feedback loop through which miR-17 temporally restricts BCL11B expression and provides functional insights into the developmental role of the disease-associated genes BCL11B and the miR-17-92 cluster in a human context.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta , Receptors, Antigen, T-Cell, gamma-delta , Cell Differentiation , Cell Lineage/genetics , Humans , Receptor, Notch1/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Repressor Proteins , Signal Transduction , Thymus Gland , Tumor Suppressor Proteins
7.
Int J Mol Sci ; 23(19)2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36232644

ABSTRACT

The ability of natural killer (NK) cells to kill tumor cells without prior sensitization makes them a rising player in immunotherapy. Increased understanding of the development and functioning of NK cells will improve their clinical utilization. As opposed to murine NK cell development, human NK cell development is still less understood. Here, we studied the role of thioredoxin-interacting protein (TXNIP) in human NK cell differentiation by stable TXNIP knockdown or overexpression in cord blood hematopoietic stem cells, followed by in vitro NK cell differentiation. TXNIP overexpression only had marginal effects, indicating that endogenous TXNIP levels are sufficient in this process. TXNIP knockdown, however, reduced proliferation of early differentiation stages and greatly decreased NK cell numbers. Transcriptome analysis and experimental confirmation showed that reduced protein synthesis upon TXNIP knockdown likely caused this low proliferation. Contrary to its profound effects on the early differentiation stages, TXNIP knockdown led to limited alterations in NK cell phenotype, and it had no effect on NK cell cytotoxicity or cytokine production. Thus, TXNIP promotes human NK cell differentiation by affecting protein synthesis and proliferation of early NK cell differentiation stages, but it is redundant for functional NK cell maturation.


Subject(s)
Killer Cells, Natural , Thioredoxins , Animals , Carrier Proteins/genetics , Cell Differentiation/genetics , Cytokines/metabolism , Gene Expression , Humans , Killer Cells, Natural/metabolism , Mice , Thioredoxins/genetics , Thioredoxins/metabolism
8.
Haematologica ; 106(1): 130-141, 2021 01 01.
Article in English | MEDLINE | ID: mdl-31919081

ABSTRACT

In both mouse and human, Notch1 activation is the main initial driver to induce T-cell development in hematopoietic progenitor cells. The initiation of this developmental process coincides with Notch1-dependent repression of differentiation towards other hematopoietic lineages. Although well described in mice, the role of the individual Notch1 target genes during these hematopoietic developmental choices is still unclear in human, particularly for HES4 since no orthologous gene is present in the mouse. Here, we investigated the functional capacity of the Notch1 target genes HES1 and HES4 to modulate human Notch1-dependent hematopoietic lineage decisions and their requirement during early T-cell development. We show that both genes are upregulated in a Notch-dependent manner during early T-cell development and that HES1 acts as a repressor of differentiation by maintaining a quiescent stem cell signature in CD34+ hematopoietic progenitor cells. While HES4 can also inhibit natural killer and myeloid cell development like HES1, it acts differently on the T- versus B-cell lineage choice. Surprisingly, HES4 is not capable of repressing B-cell development, the most sensitive hematopoietic lineage with respect to Notch-mediated repression. In contrast to HES1, HES4 promotes initiation of early T-cell development, but ectopic expression of HES4, or HES1 and HES4 combined, is not sufficient to induce T-lineage differentiation. Importantly, knockdown of HES1 or HES4 significantly reduces human T-cell development. Overall, we show that the Notch1 target genes HES1 and HES4 have non-redundant roles during early human T-cell development which may relate to differences in mediating Notch-dependent human hematopoietic lineage decisions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Hematopoietic Stem Cells , T-Lymphocytes , Transcription Factor HES-1 , Animals , Cell Differentiation , Hematopoiesis , Homeodomain Proteins/genetics , Humans , Mice , Receptor, Notch1/genetics , Transcription Factor HES-1/genetics
9.
J Immunol ; 203(12): 3339-3348, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31676671

ABSTRACT

Mice deficient in IFN-γ (IFN-γ knockout [KO] mice) develop a systemic inflammatory syndrome in response to CFA, in contrast to CFA-challenged wild-type (WT) mice who only develop a mild inflammation. Symptoms in CFA-challenged IFN-γ KO resemble systemic juvenile idiopathic arthritis (sJIA), a childhood immune disorder of unknown cause. Dysregulation of innate immune cells is considered to be important in the disease pathogenesis. In this study, we used this murine model to investigate the role of NK cells in the pathogenesis of sJIA. NK cells of CFA-challenged IFN-γ KO mice displayed an aberrant balance of activating and inhibitory NK cell receptors, lower expression of cytotoxic proteins, and a defective NK cell cytotoxicity. Depletion of NK cells (via anti-IL-2Rß and anti-Asialo-GM1 Abs) or blockade of the NK cell activating receptor NKG2D in CFA-challenged WT mice resulted in increased severity of systemic inflammation and appearance of sJIA-like symptoms. NK cells of CFA-challenged IFN-γ KO mice and from anti-NKG2D-treated mice showed defective degranulation capacities toward autologous activated immune cells, predominantly monocytes. This is in line with the increased numbers of activated inflammatory monocytes in these mice which was particularly reflected in the expression of CCR2, a chemokine receptor, and in the expression of Rae-1, a ligand for NKG2D. In conclusion, NK cells are defective in a mouse model of sJIA and impede disease development in CFA-challenged WT mice. Our findings point toward a regulatory role for NK cells in CFA-induced systemic inflammation via a NKG2D-dependent control of activated immune cells.


Subject(s)
Arthritis, Juvenile/immunology , Arthritis, Juvenile/metabolism , Disease Susceptibility , Immunomodulation , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Arthritis, Juvenile/pathology , Biomarkers , Cytotoxicity, Immunologic , Disease Models, Animal , Immunophenotyping , Interferon-gamma/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Models, Biological , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , Osteoclasts/immunology , Osteoclasts/metabolism
10.
Int J Mol Sci ; 21(22)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233766

ABSTRACT

Human thymic CD8αα+ CD10+ PD-1+ αß T cells selected through early agonist selection have been proposed as the putative thymic precursors of the human CD8αα+ intestinal intraepithelial lymphocytes (IELs). However, the progeny of these thymic precursor cells in human blood or tissues has not yet been characterized. Here, we studied the phenotypical and transcriptional differentiation of the thymic IEL precursor (IELp) lineage upon in vitro exposure to cytokines prominent in the peripheral tissues such as interleukin-15 (IL-15) and the inflammatory cytokines interleukin-12 (IL-12) and interleukin-18 (IL-18). We showed that only the CD1a- fraction of the CD10+ PD-1+ IELp population was able to proliferate with IL-15, suggesting that this subset had acquired functionality. These cells downregulated PD-1 expression and completely lost CD10 expression, whereas other surface markers such as CD95 and CXCR3 remained highly expressed. RNA-seq analysis of the IL-15-cultured cells clearly showed induction of innate-like and effector genes. Induction of the cytotoxic machinery by the CD10+ PD-1+ population was acquired in the presence of IL-15 and was further augmented by inflammatory cytokines. Our data suggest that only the CD1a- CD10+ PD-1+ population exits the thymus and survives in the periphery. Furthermore, PD-1 and CD10 expression is not an intrinsic property of this lineage, but rather characterizes a transient stage in differentiation. CD95 and CXCR3 expression combined with the absence of CD28, CCR7, and CD6 expression might be more powerful markers to define this lineage in the periphery.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Differentiation , Interleukin-15/pharmacology , Receptors, Cell Surface/metabolism , Thymocytes , Adult , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Child , Humans , Thymocytes/cytology , Thymocytes/metabolism
11.
Int J Mol Sci ; 21(3)2020 Jan 30.
Article in English | MEDLINE | ID: mdl-32019116

ABSTRACT

Recent approval of chimeric antigen receptor (CAR) T cell therapy by the European Medicines Agency (EMA)/Federal and Drug Administration (FDA) and the remarkable results of CAR T clinical trials illustrate the curative potential of this therapy. While CARs against a multitude of different antigens are being developed and tested (pre)clinically, there is still a need for optimization. The use of single-chain variable fragments (scFvs) as targeting moieties hampers the quick generation of functional CARs and could potentially limit the efficacy. Instead, nanobodies may largely circumvent these difficulties. We used an available nanobody library generated after immunization of llamas against Cluster of Differentiation (CD) 20 through DNA vaccination or against the ectodomain of CD33 using soluble protein. The nanobody specific sequences were amplified by PCR and cloned by Gibson Assembly into a retroviral vector containing two different second-generation CAR constructs. After transduction in T cells, we observed high cell membrane nanoCAR expression in all cases. Following stimulation of nanoCAR-expressing T cells with antigen-positive cell lines, robust T cell activation, cytokine production and tumor cell lysis both in vitro and in vivo was observed. The use of nanobody technology in combination with PCR and Gibson Assembly allows for the rapid and effective generation of compact CARs.


Subject(s)
Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/immunology , Single-Chain Antibodies/immunology , Single-Domain Antibodies/immunology , Cell Line , Genetic Vectors , Humans , Lymphocyte Activation , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/genetics , Single-Chain Antibodies/genetics , T-Lymphocytes/immunology
12.
Eur Respir J ; 52(6)2018 12.
Article in English | MEDLINE | ID: mdl-30385529

ABSTRACT

Although type 1, 2 and 3 innate lymphoid cells (ILC1s, ILC2s and ILC3s, respectively) are emerging as important cell populations regulating tissue homeostasis, remodelling and inflammation, a vast majority of our knowledge stems from in vitro and murine experiments, and requires thorough confirmation in human diseases.Relative levels of ILCs were evaluated by means of flow cytometry in freshly resected human upper airways mucosa of patients with chronic rhinosinusitis without nasal polyps (CRSsNP) and with nasal polyps (CRSwNP), taking into account the patient's clinical parameters and disease comorbidities.We report that the CD117 and interleukin-receptor type I (IL-1RI) expression status of human ILC2s depends on the local tissue environment. Only CD117+ IL-1RI+ ILC2s, exclusively present in CRSwNP, possess an interrelationship with type 2 T-helper cell cytokine and eosinophil levels in human upper airway mucosa. In CRSsNP, mainly CD117-IL-1RI- ILC2s are increased, yielding lower eosinophilia in this disease despite the high levels of ILC2s.These data unveil that the CD117- and CD117+ fractions within the native human ILC2 population are not a random phenomenon, in contrast to what could be concluded from in vitro data, and that the IL-1RI expression is not ubiquitous in ILC2s in vivo in humans, which cannot be assessed via in vitro and murine experiments.


Subject(s)
Lymphocytes/cytology , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Interleukin-1 Type I/metabolism , Rhinitis/immunology , Sinusitis/immunology , Adolescent , Adult , Aged , Chronic Disease , Eosinophils/cytology , Eosinophils/metabolism , Female , Flow Cytometry , Humans , Lymphocytes/metabolism , Male , Middle Aged , Nasal Polyps/complications , Rhinitis/complications , Sinusitis/complications , Young Adult
13.
Immunity ; 30(3): 337-47, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19249231

ABSTRACT

Natural killer (NK) cells show enhanced functional competence when they express inhibitory receptors specific for inherited major histocompatibility complex class I (MHC-I) molecules. Current models imply that NK cell education requires an interaction of inhibitory receptors with MHC-I expressed on other cells. However, the inhibitory Ly49A receptor can also bind MHC-I ligand on the NK cell itself (in cis). Here we describe a Ly49A variant, which can engage MHC-I expressed on other cells but not in cis. Even though this variant inhibited NK cell effector function, it failed to educate NK cells. The association with MHC-I in cis sequestered wild-type Ly49A, and this was found to relieve NK cells from a suppressive effect of unengaged Ly49A. These data explain how inhibitory MHC-I receptors can facilitate NK cell activation. They dissociate classical inhibitory from educating functions of Ly49A and suggest that cis interaction of Ly49A is necessary for NK cell education.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Flow Cytometry , Genetic Variation , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily A/genetics
14.
Cancer Immunol Immunother ; 65(11): 1365-1375, 2016 11.
Article in English | MEDLINE | ID: mdl-27585789

ABSTRACT

Ly49E is a member of the Ly49 family of NK receptors and is distinct from other members of this family on the basis of its structural properties, expression pattern and ligand recognition. Importantly, Ly49E receptor expression is high on small intestinal and colonic intraepithelial lymphocytes (IELs). Intestinal IELs are regulators of the mucosal immune system and contribute to front-line defense at the mucosal barrier, including anti-tumor immune response. Whereas most Ly49 receptors have MHC class-I ligands, we showed that Ly49E is instead triggered by urokinase plasminogen activator (uPA). uPA has been extensively implicated in tumor development, where increased uPA expression correlates with poor prognosis. As such, we investigated the role of Ly49E receptor expression on intestinal IELs in the anti-tumor immune response. For this purpose, we compared Ly49E wild-type mice to Ly49E knockout mice in two established tumor models: ApcMin/+-mediated and azoxymethane-induced intestinal cancer. Our results indicate that Ly49E expression on IELs does not influence the development or progression of intestinal cancer.


Subject(s)
Carcinoma in Situ/immunology , Epithelium/immunology , Intestinal Neoplasms/immunology , Lymphocytes/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Azoxymethane , Carcinogenesis , Carcinoma in Situ/chemically induced , Carcinoma in Situ/genetics , Disease Models, Animal , Epithelium/pathology , Gene Expression Regulation, Neoplastic , Immunity, Cellular , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , Tumor Burden , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
15.
J Immunol ; 193(12): 5997-6004, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25381438

ABSTRACT

Although the role for the individual Notch receptors in early hematopoiesis have been thoroughly investigated in mouse, studies in human have been mostly limited to the use of pan-Notch inhibitors. However, such studies in human are important to predict potential side effects of specific Notch receptor blocking reagents because these are currently being considered as therapeutic tools to treat various Notch-dependent diseases. In this study, we studied the individual roles of Notch1 and Notch3 in early human hematopoietic lineage decisions, particularly during T-lineage specification. Although this process in mice is solely dependent on Notch1 activation, we recently reported Notch3 expression in human uncommitted thymocytes, raising the possibility that Notch3 mediates human T-lineage specification. Although expression of a constitutive activated form of Notch3 (ICN3) results in the induction of T-lineage specification in human CD34(+) hematopoietic progenitor cells, similar to ICN1 overexpression, loss-of-function studies using blocking Abs reveal that only Notch1, but not Notch3, is critical in this process. Blocking of Notch1 activation in OP9-DLL4 cocultures resulted in a complete block in T-lineage specification and induced monocytic and plasmacytoid dendritic cell differentiation instead. In fetal thymus organ cultures, impeded Notch1 activation resulted in B and dendritic cell development. In contrast, Notch3 blocking Abs only marginally affected T-lineage specification and hematopoietic differentiation with a slight increase in monocyte development. No induction of B or dendritic cell development was observed. Thus, our results unambiguously reveal a nonredundant role for Notch1 in human T-lineage specification, despite the expression of other Notch receptors.


Subject(s)
Cell Differentiation , Cell Lineage , Receptors, Notch/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Gene Expression , Humans , Immunophenotyping , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Mice , Phenotype , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch3 , Receptors, Notch/genetics , Thymocytes/cytology , Thymocytes/metabolism
16.
Toxicol Appl Pharmacol ; 288(2): 161-78, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26187750

ABSTRACT

Targeting excessive production of reactive oxygen species (ROS) could be an effective therapeutic strategy to prevent oxidative stress-associated gastrointestinal inflammation. NADPH oxidase (NOX) and mitochondrial complexes (I and II) are the major sources of ROS production contributing to TNF-α/cycloheximide (CHX)-induced apoptosis in the mouse intestinal epithelial cell line, MODE-K. In the current study, the influence of a polyphenolic compound (resveratrol) and a water-soluble carbon monoxide (CO)-releasing molecule (CORM-A1) on the different sources of TNF-α/CHX-induced ROS production in MODE-K cells was assessed. This was compared with H2O2-, rotenone- or antimycin-A-induced ROS-generating systems. Intracellular total ROS, mitochondrial-derived ROS and mitochondrial superoxide anion (O2(-)) production levels were assessed. Additionally, the influence on TNF-α/CHX-induced changes in mitochondrial membrane potential (Ψm) and mitochondrial function was studied. In basal conditions, CORM-A1 did not affect intracellular total or mitochondrial ROS levels, while resveratrol increased intracellular total ROS but reduced mitochondrial ROS production. TNF-α/CHX- and H2O2-mediated increase in intracellular total ROS production was reduced by both resveratrol and CORM-A1, whereas only resveratrol attenuated the increase in mitochondrial ROS triggered by TNF-α/CHX. CORM-A1 decreased antimycin-A-induced mitochondrial O2(-) production without any influence on TNF-α/CHX- and rotenone-induced mitochondrial O2(-) levels, while resveratrol abolished all three effects. Finally, resveratrol greatly reduced and abolished TNF-α/CHX-induced mitochondrial depolarization and mitochondrial dysfunction, while CORM-A1 only mildly affected these parameters. These data indicate that the cytoprotective effect of resveratrol is predominantly due to mitigation of mitochondrial ROS, while CORM-A1 acts solely on NOX-derived ROS to protect MODE-K cells from TNF-α/CHX-induced cell death. This might explain the more pronounced cytoprotective effect of resveratrol.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Boranes/pharmacology , Carbonates/pharmacology , Cycloheximide/toxicity , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Oxidative Stress/drug effects , Stilbenes/pharmacology , Tumor Necrosis Factor-alpha/toxicity , Animals , Cell Line , Cytoprotection , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NADPH Oxidases/metabolism , Oxygen Consumption/drug effects , Resveratrol , Superoxides/metabolism
17.
Haematologica ; 100(2): 157-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25381126

ABSTRACT

Although hematopoietic precursor activity can be generated in vitro from human embryonic stem cells, there is no solid evidence for the appearance of multipotent, self-renewing and transplantable hematopoietic stem cells. This could be due to short half-life of hematopoietic stem cells in culture or, alternatively, human embryonic stem cell-initiated hematopoiesis may be hematopoietic stem cell-independent, similar to yolk sac hematopoiesis, generating multipotent progenitors with limited expansion capacity. Since a MYB was reported to be an excellent marker for hematopoietic stem cell-dependent hematopoiesis, we generated a MYB-eGFP reporter human embryonic stem cell line to study formation of hematopoietic progenitor cells in vitro. We found CD34(+) hemogenic endothelial cells rounding up and developing into CD43(+) hematopoietic cells without expression of MYB-eGFP. MYB-eGFP(+) cells appeared relatively late in embryoid body cultures as CD34(+)CD43(+)CD45(-/lo) cells. These MYB-eGFP(+) cells were CD33 positive, proliferated in IL-3 containing media and hematopoietic differentiation was restricted to the granulocytic lineage. In agreement with data obtained on murine Myb(-/-) embryonic stem cells, bright eGFP expression was observed in a subpopulation of cells, during directed myeloid differentiation, which again belonged to the granulocytic lineage. In contrast, CD14(+) macrophage cells were consistently eGFP(-) and were derived from eGFP-precursors only. In summary, no evidence was obtained for in vitro generation of MYB(+) hematopoietic stem cells during embryoid body cultures. The observed MYB expression appeared late in culture and was confined to the granulocytic lineage.


Subject(s)
Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins c-myb/metabolism , Yolk Sac/cytology , Cells, Cultured , Embryoid Bodies , Embryonic Stem Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Flow Cytometry , Granulocytes/cytology , Granulocytes/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Humans , In Vitro Techniques , Macrophages/cytology , Macrophages/metabolism , Proto-Oncogene Proteins c-myb/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transgenes/physiology , Yolk Sac/metabolism
18.
J Immunol ; 191(10): 5044-51, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24098052

ABSTRACT

Although NK cells use invariant receptors to identify diseased cells, they nevertheless adapt to their environment, including the presence of certain MHC class I (MHC-I) molecules. This NK cell education, which is mediated by inhibitory receptors specific for MHC-I molecules, changes the responsiveness of activating NK cell receptors (licensing) and modifies the repertoire of MHC-I receptors used by NK cells. The fact that certain MHC-I receptors have the unusual capacity to recognize MHC-I molecules expressed by other cells (trans) and by the NK cell itself (cis) has raised the question regarding possible contributions of the two types of interactions to NK cell education. Although the analysis of an MHC-I receptor variant suggested a role for cis interaction for NK cell licensing, adoptive NK cell transfer experiments supported a key role for trans recognition. To reconcile some of these findings, we have analyzed the impact of cell type-specific deletion of an MHC-I molecule and of a novel MHC-I receptor variant on the education of murine NK cells when these mature under steady-state conditions in vivo. We find that MHC-I expression by NK cells (cis) and by T cells (trans), and MHC-I recognition in cis and in trans, are both needed for NK cell licensing. Unexpectedly, modifications of the MHC-I receptor repertoire are chiefly dependent on cis binding, which provides additional support for an essential role for this unconventional type of interaction for NK cell education. These data suggest that two separate functions of MHC-I receptors are needed to adapt NK cells to self-MHC-I.


Subject(s)
H-2 Antigens/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Adoptive Transfer , Animals , Cell Line , H-2 Antigens/genetics , Histocompatibility Antigens Class I/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/immunology
19.
J Immunol ; 190(5): 1982-90, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23338239

ABSTRACT

The Ly49 NK receptor family in mice is composed of several members that recognize MHC class I (MHC-I) or MHC-I-related molecules. We and others have shown before that Ly49E is a unique member, with a different expression pattern on NK cells and being triggered by the non-MHC-I-related protein urokinase plasminogen activator. Among the entire Ly49 receptor family, Ly49E is the only Ly49 member expressed by epidermal-localized γδ T cells and their fetal thymic TCRγδ precursors, and it is the most abundantly expressed member on intestinal intraepithelial γδ T cell lymphocytes. In this study, we provide mechanistic insights into the regulation of Ly49e expression in γδ T cells. First, we demonstrate that TCR-mediated activation of intraepithelial γδ T cells significantly increases Ly49E expression. This results from de novo Ly49E expression and is highly selective, because no other Ly49 family members are induced. TCR-mediated Ly49E induction is a conserved feature of skin- and gut-residing intraepithelial-localized γδ T cell subsets, whereas it is not observed in spleen γδ T cells. By investigating Ly49e promoter activities and lymphotoxin (LT) αß dependency in resting versus TCR-activated intraepithelial γδ T cells, we reveal two separate regulatory pathways for Ly49E expression, as follows: a LTαß-dependent pathway leading to basal Ly49E expression in resting cells that is induced by Pro2-mediated Ly49e transcription, and a LTαß-independent pathway leading to elevated, Pro3-driven Ly49E expression in TCR-stimulated cells.


Subject(s)
Epithelium/drug effects , Killer Cells, Natural/drug effects , NK Cell Lectin-Like Receptor Subfamily A/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/drug effects , Transcription, Genetic/drug effects , Animals , Epidermal Cells , Epidermis/drug effects , Epidermis/immunology , Epithelium/immunology , Gene Expression Regulation , Intestines/cytology , Intestines/drug effects , Intestines/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphotoxin alpha1, beta2 Heterotrimer/pharmacology , Mice , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/immunology , Promoter Regions, Genetic , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Urokinase-Type Plasminogen Activator/pharmacology
20.
Biomacromolecules ; 15(6): 2301-9, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24805802

ABSTRACT

During the past decade, extensive research has undeniably improved the formulation and delivery of oral vaccines. Nevertheless, several factors, such as the harsh gastrointestinal environment together with tolerance induction to exogenous antigens, have thus far impeded the optimal effectiveness and clinical application of oral delivery systems. The current study encompasses an initial evaluation of the stability, biocompatibility, and cellular uptake of two promising candidate systems for oral antigen delivery, that is, calcium carbonate- (CP) and mannitol-templated (MP) porous microspheres. Both spray-dried formulations were efficiently internalized by human intestinal epithelial cells (Caco-2 and HT-29) and degraded into phagolysosomal intracellular compartments. In addition, cellular particle uptake and processing significantly up-regulated the expression of (HLA) class-II and costimulatory molecules on intestinal epithelial cells. Even though the high surface-area-to-volume ratio of the microspheres was expected to favor protease access, antigen release was remarkably limited in simulated intestinal fluid and was even absent under gastric conditions. Finally, neither CP nor MP exerted cytotoxicity upon prolonged in vitro incubation with high antigen concentration. Altogether, these data support the potential of CP and MP for oral antigen delivery and motivate the further development of these promising carrier systems in in vivo studies.


Subject(s)
Antigens/metabolism , Biocompatible Materials/metabolism , Drug Delivery Systems/methods , Microspheres , Administration, Oral , Antigens/administration & dosage , Biocompatible Materials/administration & dosage , Caco-2 Cells , Cell Survival/drug effects , Cell Survival/physiology , Drug Stability , HT29 Cells , Humans , Ovalbumin/administration & dosage , Ovalbumin/metabolism , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL