Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nature ; 629(8010): 211-218, 2024 May.
Article in English | MEDLINE | ID: mdl-38600391

ABSTRACT

A major limitation of chimeric antigen receptor (CAR) T cell therapies is the poor persistence of these cells in vivo1. The expression of memory-associated genes in CAR T cells is linked to their long-term persistence in patients and clinical efficacy2-6, suggesting that memory programs may underpin durable CAR T cell function. Here we show that the transcription factor FOXO1 is responsible for promoting memory and restraining exhaustion in human CAR T cells. Pharmacological inhibition or gene editing of endogenous FOXO1 diminished the expression of memory-associated genes, promoted an exhaustion-like phenotype and impaired the antitumour activity of CAR T cells. Overexpression of FOXO1 induced a gene-expression program consistent with T cell memory and increased chromatin accessibility at FOXO1-binding motifs. CAR T cells that overexpressed FOXO1 retained their function, memory potential and metabolic fitness in settings of chronic stimulation, and exhibited enhanced persistence and tumour control in vivo. By contrast, overexpression of TCF1 (encoded by TCF7) did not enforce canonical memory programs or enhance the potency of CAR T cells. Notably, FOXO1 activity correlated with positive clinical outcomes of patients treated with CAR T cells or tumour-infiltrating lymphocytes, underscoring the clinical relevance of FOXO1 in cancer immunotherapy. Our results show that overexpressing FOXO1 can increase the antitumour activity of human CAR T cells, and highlight memory reprogramming as a broadly applicable approach for optimizing therapeutic T cell states.


Subject(s)
Forkhead Box Protein O1 , Immunologic Memory , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , T-Lymphocytes , Animals , Humans , Mice , Cell Line, Tumor , Chromatin/metabolism , Chromatin/genetics , Forkhead Box Protein O1/metabolism , Gene Editing , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/cytology
3.
Cell Rep ; 42(8): 112905, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37527035

ABSTRACT

CD8+ T cell exhaustion (TEX) impairs the ability of T cells to clear chronic infection or cancer. While TEX are hypofunctional, some TEX retain effector gene signatures, a feature associated with killer lectin-like receptor (KLR) expression. Although KLR+ TEX (TKLR) may improve control of chronic antigen, the signaling molecules regulating this population are poorly understood. Using single-cell RNA sequencing (scRNA-seq), flow cytometry, RNA velocity, and single-cell T cell receptor sequencing (scTCR-seq), we demonstrate that deleting the pseudokinase Trib1 shifts TEX toward CX3CR1+ intermediates with robust enrichment of TKLR via clonal T cell expansion. Adoptive transfer studies demonstrate this shift toward CD8+ TKLR in Trib1-deficient cells is CD8 intrinsic, while CD4-depletion studies demonstrate CD4+ T cells are required for improved viral control in Trib1 conditional knockout mice. Further, Trib1 loss augments anti-programmed death-ligand 1 (PD-L1) blockade to improve viral clearance. These data identify Trib1 as an important regulator of CD8+ TEX whose targeting enhances the TKLR effector state and improves checkpoint inhibitor therapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Animals , Mice , Neoplasms/metabolism , Receptors, Antigen, T-Cell/metabolism , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
4.
Hematol Oncol Clin North Am ; 37(6): 1041-1052, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37500380

ABSTRACT

Over the past decade, CAR T cell therapy has transformed the treatment of relapsed or refractory B-ALL in children and adults. CD19-directed CAR T cells can induce complete remissions in a large majority of patients with B-ALL, and up to half of these patients will go on to maintain durable remissions. However, significant challenges remain for patients who relapse or do not respond. This review will discuss the history of CAR T cell therapy for B-ALL, the treatment considerations for CAR T cell recipients, and current clinical trials and future directions for CAR T cell therapy in B-ALL.

5.
bioRxiv ; 2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36824931

ABSTRACT

T cell exhaustion (T EX ) impairs the ability of T cells to clear chronic infection or cancer. While exhausted T cells are hypofunctional, some exhausted T cells retain effector gene signatures, a feature that is associated with expression of KLRs (killer lectin-like receptors). Although KLR + T cells may improve control of chronic antigen, the signaling molecules regulating this population are poorly understood. Using scRNA-seq, flow cytometry, RNA velocity, and scTCR-seq, we demonstrate that deleting the pseudokinase Trib1 shifts T EX towards CX3CR1 + intermediates (T INT ) with robust enrichment of KLR + CD8 + T cells (T KLR ) via clonal T cell expansion. These changes are associated with globally increased KLR gene expression throughout the exhaustion program. Further, Trib1 loss augments anti-PD-L1 blockade to improve viral clearance by expanding the T KLR population. Together, these data identify Trib1 as an important regulator of T cell exhaustion whose targeting enhances the KLR + effector state and improves the response to checkpoint inhibitor therapy.

6.
J Exp Med ; 217(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32150623

ABSTRACT

In chronic infections, the immune response fails to control virus, leading to persistent antigen stimulation and the progressive development of T cell exhaustion. T cell effector differentiation is poorly understood in the context of exhaustion, but targeting effector programs may provide new strategies for reinvigorating T cell function. We identified Tribbles pseudokinase 1 (Trib1) as a central regulator of antiviral T cell immunity, where loss of Trib1 led to a sustained enrichment of effector-like KLRG1+ T cells, enhanced function, and improved viral control. Single-cell profiling revealed that Trib1 restrains a population of KLRG1+ effector CD8 T cells that is transcriptionally distinct from exhausted cells. Mechanistically, we identified an interaction between Trib1 and the T cell receptor (TCR) signaling activator, MALT1, which disrupted MALT1 signaling complexes. These data identify Trib1 as a negative regulator of TCR signaling and downstream function, and reveal a link between Trib1 and effector versus exhausted T cell differentiation that can be targeted to improve antiviral immunity.


Subject(s)
Cell Differentiation , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Chronic Disease , Humans , Immunity , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/deficiency , Lymphocyte Activation/immunology , Lymphocyte Subsets/immunology , Lymphocytic choriomeningitis virus/immunology , Mice, Inbred C57BL , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Phenotype , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcription, Genetic , Viral Load
7.
J Immunol ; 199(7): 2333-2342, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28842466

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are important regulators of the immune system, maintaining homeostasis in the presence of commensal bacteria, but activating immune defenses in response to microbial pathogens. ILC3s are a robust source of IL-22, a cytokine critical for stimulating the antimicrobial response. We sought to identify cytokines that can promote proliferation and induce or maintain IL-22 production by ILC3s and determine a molecular mechanism for this process. We identified IL-18 as a cytokine that cooperates with an ILC3 survival factor, IL-15, to induce proliferation of human ILC3s, as well as induce and maintain IL-22 production. To determine a mechanism of action, we examined the NF-κB pathway, which is activated by IL-18 signaling. We found that the NF-κB complex signaling component, p65, binds to the proximal region of the IL22 promoter and promotes transcriptional activity. Finally, we observed that CD11c+ dendritic cells expressing IL-18 are found in close proximity to ILC3s in human tonsils in situ. Therefore, we identify a new mechanism by which human ILC3s proliferate and produce IL-22, and identify NF-κB as a potential therapeutic target to be considered in pathologic states characterized by overproduction of IL-18 and/or IL-22.


Subject(s)
Cell Proliferation , Interleukin-18/metabolism , Interleukins/biosynthesis , Lymphocytes/physiology , NF-kappa B/metabolism , Signal Transduction , Dendritic Cells/physiology , Humans , Immunity, Innate , Interleukin-15/immunology , Interleukins/genetics , Interleukins/immunology , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Promoter Regions, Genetic , Signal Transduction/immunology , Transcription Factor RelA/metabolism , Interleukin-22
8.
Immunity ; 44(5): 1140-50, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27178467

ABSTRACT

The current model of murine innate lymphoid cell (ILC) development holds that mouse ILCs are derived downstream of the common lymphoid progenitor through lineage-restricted progenitors. However, corresponding lineage-restricted progenitors in humans have yet to be discovered. Here we identified a progenitor population in human secondary lymphoid tissues (SLTs) that expressed the transcription factor RORγt and was unique in its ability to generate all known ILC subsets, including natural killer (NK) cells, but not other leukocyte populations. In contrast to murine fate-mapping data, which indicate that only ILC3s express Rorγt, these human progenitor cells as well as human peripheral blood NK cells and all mature ILC populations expressed RORγt. Thus, all human ILCs can be generated through an RORγt(+) developmental pathway from a common progenitor in SLTs. These findings help establish the developmental signals and pathways involved in human ILC development.


Subject(s)
Killer Cells, Natural/physiology , Lymph Nodes/immunology , Lymphocyte Subsets/physiology , Lymphoid Progenitor Cells/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Palatine Tonsil/immunology , Adult , Animals , Antigens, CD34/metabolism , Cell Differentiation , Cell Line , Child , Gene Expression Regulation , Humans , Immunity, Innate , Leukocyte Common Antigens/metabolism , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
9.
J Immunol ; 195(5): 1995-2005, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26238487

ABSTRACT

Sorafenib is an oral multikinase inhibitor that was originally developed as a Raf kinase inhibitor. We hypothesized that sorafenib would also have inhibitory effects on cytokine signaling pathways in immune cells. PBMCs from normal donors were treated with varying concentrations of sorafenib and stimulated with IFN-α or IL-2. Phosphorylation of STAT1 and STAT5 was measured by flow cytometry and confirmed by immunoblot analysis. Changes in IFN-α- and IL-2-stimulated gene expression were measured by quantitative PCR, and changes in cytokine production were evaluated by ELISA. Cryopreserved PBMCs were obtained from cancer patients before and after receiving 400 mg sorafenib twice daily. Patient PBMCs were thawed, stimulated with IL-2 or IFN-α, and evaluated for phosphorylation of STAT1 and STAT5. Pretreatment of PBMCs with 10 µM sorafenib decreased STAT1 and STAT5 phosphorylation after treatment with IFN-α or IL-2. This inhibitory effect was observed in PBMCs from healthy donors over a range of concentrations of sorafenib (5-20 µM), IL-2 (2-24 nM), and IFN-α (10(1)-10(6) U/ml). This effect was observed in immune cell subsets, including T cells, B cells, NK cells, regulatory T cells, and myeloid-derived suppressor cells. Pretreatment with sorafenib also inhibited PBMC expression of IFN-α- and IL-2-regulated genes and inhibited NK cell production of IFN-γ, RANTES, MIP1-α, and MIG in response to IFN-α stimulation. PBMCs from patients receiving sorafenib therapy showed decreased responsiveness to IL-2 and IFN-α treatment. Sorafenib is a Raf kinase inhibitor that could have off-target effects on cytokine-induced signal transduction in immune effector cells.


Subject(s)
Janus Kinase 1/metabolism , Leukocytes, Mononuclear/drug effects , Protein Kinase Inhibitors/pharmacology , STAT1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression/drug effects , Humans , Immunoblotting , Interferon-alpha/pharmacology , Interleukin-2/pharmacology , K562 Cells , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice, Inbred BALB C , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sorafenib , Thyroid Neoplasms/blood , Thyroid Neoplasms/drug therapy , raf Kinases/antagonists & inhibitors , raf Kinases/metabolism
10.
Cell Rep ; 8(1): 150-62, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24953655

ABSTRACT

Accumulating evidence indicates that human natural killer (NK) cells develop in secondary lymphoid tissue (SLT) through a so-called "stage 3" developmental intermediate minimally characterized by a CD34(-)CD117(+)CD94(-) immunophenotype that lacks mature NK cell function. This stage 3 population is heterogeneous, potentially composed of functionally distinct innate lymphoid cell (ILC) types that include interleukin-1 receptor (IL-1R1)-positive, IL-22-producing ILC3s. Whether human ILC3s are developmentally related to NK cells is a subject of ongoing investigation. Here, we show that antagonism of the aryl hydrocarbon receptor (AHR) or silencing of AHR gene expression promotes the differentiation of tonsillar IL-22-producing IL-1R1(hi) human ILC3s to CD56(bright)CD94(+) interferon (IFN)-γ-producing cytolytic mature NK cells expressing eomesodermin (EOMES) and T-Box Protein 21 (TBX21 or TBET). Hence, we demonstrate the lineage plasticity of human ILCs by identifying AHR as a transcription factor that prevents IL-1R1(hi) ILC3s from differentiating into NK cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Killer Cells, Natural/immunology , Lymphocytes/immunology , Receptors, Aryl Hydrocarbon/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , CD56 Antigen/genetics , CD56 Antigen/metabolism , Cell Lineage , Cells, Cultured , Humans , Interleukins/genetics , Interleukins/metabolism , Killer Cells, Natural/cytology , Lymphocytes/cytology , Palatine Tonsil/cytology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Interleukin-22
11.
J Clin Invest ; 122(4): 1403-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22378041

ABSTRACT

The development of a broad repertoire of T cells, which is essential for effective immune function, occurs in the thymus. Although some data suggest that T cell development can occur extrathymically, many researchers remain skeptical that extrathymic T cell development has an important role in generating the T cell repertoire in healthy individuals. However, it may be important in the setting of poor thymic function or congenital deficit and in the context of autoimmunity, cancer, or regenerative medicine. Here, we report evidence that a stepwise program of T cell development occurs within the human tonsil. We identified 5 tonsillar T cell developmental intermediates: (a) CD34⁺CD38dimLin⁻ cells, which resemble multipotent progenitors in the bone marrow and thymus; (b) more mature CD34⁺CD38brightLin⁻ cells; (c) CD34⁺CD1a⁺CD11c⁻ cells, which resemble committed T cell lineage precursors in the thymus; (d) CD34⁻CD1a⁺CD3⁻CD11c⁻ cells, which resemble CD4⁺CD8⁺ double-positive T cells in the thymus; and (e) CD34⁻CD1a⁺CD3⁺CD11c⁻ cells. The phenotype of each subset closely resembled that of its thymic counterpart. The last 4 populations expressed RAG1 and PTCRA, genes required for TCR rearrangement, and all 5 subsets were capable of ex vivo T cell differentiation. TdT⁺ cells found within the tonsillar fibrous scaffold expressed CD34 and/or CD1a, indicating that this distinct anatomic region contributes to pre-T cell development, as does the subcapsular region of the thymus. Thus, we provide evidence of a role for the human tonsil in a comprehensive program of extrathymic T cell development.


Subject(s)
Killer Cells, Natural/cytology , Lymphopoiesis , Palatine Tonsil/immunology , T-Lymphocyte Subsets/cytology , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , Cell Lineage , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/analysis , Humans , Immunophenotyping , Killer Cells, Natural/chemistry , Membrane Glycoproteins/analysis , Organ Specificity , Palatine Tonsil/cytology , Palatine Tonsil/ultrastructure , Receptors, Antigen, T-Cell, alpha-beta/analysis , Stem Cell Niche , T-Lymphocyte Subsets/chemistry , Thymus Gland/cytology , Thymus Gland/immunology
12.
J Immunol ; 185(7): 3913-8, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20802153

ABSTRACT

NK cells are the dominant population of immune cells in the endometrium in the secretory phase of the menstrual cycle and in the decidua in early pregnancy. The possibility that this is a site of NK cell development is of particular interest because of the cyclical death and regeneration of the NK population during the menstrual cycle. To investigate this, we searched for NK developmental stages 1-4, based on expression of CD34, CD117, and CD94. In this study, we report that a heterogeneous population of stage 3 NK precursor (CD34(-)CD117(+)CD94(-)) and mature stage 4 NK (CD34(-)CD117(-/+)CD94(+)) cells, but not multipotent stages 1 and 2 (CD34(+)), are present in the uterine mucosa. Cells within the uterine stage 3 population are able to give rise to mature stage 4-like cells in vitro but also produce IL-22 and express RORC and LTA. We also found stage 3 cells with NK progenitor potential in peripheral blood. We propose that stage 3 cells are recruited from the blood to the uterus and mature in the uterine microenvironment to become distinctive uterine NK cells. IL-22 producers in this population might have a physiological role in this specialist mucosa dedicated to reproduction.


Subject(s)
Immunity, Mucosal/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mucous Membrane/immunology , Uterus/immunology , Cell Differentiation/immunology , Cell Separation , Female , Flow Cytometry , Humans , Immunohistochemistry , Interleukins/immunology , Interleukins/metabolism , Mucous Membrane/cytology , Reverse Transcriptase Polymerase Chain Reaction , Uterus/cytology , Interleukin-22
13.
Immunity ; 32(6): 803-14, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20620944

ABSTRACT

Among human natural killer (NK) cell intermediates in secondary lymphoid tissue (SLT), stage 3 CD34(-)CD117(+)CD161(+)CD94(-) immature NK (iNK) cells uniquely express aryl hydrocarbon receptor (AHR) and interleukin-22 (IL-22), supporting a role in mucosal immunity. The mechanisms controlling proliferation and differentiation of these cells are unknown. Here we demonstrate that the IL-1 receptor IL-1R1 was selectively expressed by a subpopulation of iNK cells that localized proximal to IL-1beta-producing conventional dendritic cells (cDCs) within SLT. IL-1R1(hi) iNK cells required continuous exposure to IL-1beta to retain AHR and IL-22 expression, and they proliferate in direct response to cDC-derived IL-15 and IL-1beta. In the absence of IL-1beta, a substantially greater fraction of IL-1R1(hi) iNK cells differentiated to stage 4 NK cells and acquired the ability to kill and secrete IFN-gamma. Thus, cDC-derived IL-1beta preserves and expands IL-1R1(hi)IL-22(+)AHR(+) iNK cells, potentially influencing human mucosal innate immunity during infection.


Subject(s)
Cell Differentiation/immunology , Interleukin-1beta/immunology , Interleukins/immunology , Killer Cells, Natural/cytology , Cell Proliferation , Cell Separation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Humans , Immunity, Mucosal/immunology , Immunohistochemistry , Interleukin-1beta/metabolism , Interleukins/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Interleukin-22
14.
Blood ; 115(2): 274-81, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19897577

ABSTRACT

Human CD56(bright) natural killer (NK) cells possess little or no killer immunoglobulin-like receptors (KIRs), high interferon-gamma (IFN-gamma) production, but little cytotoxicity. CD56(dim) NK cells have high KIR expression, produce little IFN-gamma, yet display high cytotoxicity. We hypothesized that, if human NK maturation progresses from a CD56(bright) to a CD56(dim) phenotype, an intermediary NK cell must exist, which demonstrates more functional overlap than these 2 subsets, and we used CD94 expression to test our hypothesis. CD94(high)CD56(dim) NK cells express CD62L, CD2, and KIR at levels between CD56(bright) and CD94(low)CD56(dim) NK cells. CD94(high)CD56(dim) NK cells produce less monokine-induced IFN-gamma than CD56(bright) NK cells but much more than CD94(low)CD56(dim) NK cells because of differential interleukin-12-mediated STAT4 phosphorylation. CD94(high)CD56(dim) NK cells possess a higher level of granzyme B and perforin expression and CD94-mediated redirected killing than CD56(bright) NK cells but lower than CD94(low)CD56(dim) NK cells. Collectively, our data suggest that the density of CD94 surface expression on CD56(dim) NK cells identifies a functional and likely developmental intermediary between CD56(bright) and CD94(low)CD56(dim) NK cells. This supports the notion that, in vivo, human CD56(bright) NK cells progress through a continuum of differentiation that ends with a CD94(low)CD56(dim) phenotype.


Subject(s)
CD56 Antigen/immunology , Cell Differentiation/immunology , Gene Expression Regulation/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , NK Cell Lectin-Like Receptor Subfamily D/immunology , Cells, Cultured , Humans , Interferon-gamma/immunology , Interleukin-12/immunology , Killer Cells, Natural/cytology , L-Selectin/immunology , Lymphocyte Subsets/cytology , Phosphorylation/immunology , STAT4 Transcription Factor/immunology
15.
Blood ; 113(17): 4008-10, 2009 Apr 23.
Article in English | MEDLINE | ID: mdl-19244159

ABSTRACT

Considerable functional heterogeneity within human natural killer (NK) cells has been revealed through the characterization of distinct NK-cell subsets. Accordingly, a small subset of CD56(+)NKp44(+)NK cells, termed NK-22 cells, was recently described within secondary lymphoid tissue (SLT) as IL-22(-) when resting, with a minor fraction of this population becoming IL-22(+) when activated. Here we discover that the vast majority of stage 3 immature NK (iNK) cells in SLT constitutively and selectively express IL-22, a T(H)17 cytokine important for mucosal immunity, whereas earlier and later stages of NK developmental intermediates do not express IL-22. These iNK cells have a surface phenotype of CD34(-)CD117(+)CD161(+)CD94(-), largely lack expression of NKp44 and CD56, and do not produce IFN-gamma or possess cytolytic activity. In summary, stage 3 iNK cells are highly enriched for IL-22 and IL-26 messenger RNA, and IL-22 protein production, but do not express IL-17A or IL-17F.


Subject(s)
Interleukin-17/immunology , Interleukins/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphoid Tissue/immunology , Lymphopoiesis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Humans , Interleukins/genetics , Interleukins/metabolism , Killer Cells, Natural/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Phenotype , RNA, Messenger/genetics , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL
...