Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cytometry A ; 85(10): 901-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25088497

ABSTRACT

Natural regulatory T cells (nTreg) can suppress different immune-cell responses and maintain the balance between tolerance and immunity in the individual. These cells are defined by CD4+ , CD25hi, and FOXP3+ expression, although a variety of other nTreg-associated markers have been reported to be expressed at different levels (e.g., HLA-DR, CTLA-4, GARP, Helios, CD39, etc.), presumably reflecting different functional stages of the heterogeneous nTreg population. Several markers show low/negative expression (i.e., CD127, CD49d, and CD26), but none of these markers are specific to nTreg. CD25hi expression has been a useful surface marker to identify/isolate nTreg; however, CD25 is also expressed on "adaptive" or "induced" Treg, as well as in activated conventional T cells. In addition, the fact that FOXP3 is also found in CD25 low/negative CD4+ T cells, and in a subset of CD8+ T cells, further complicates the definition of a specific nTreg marker. Although CD4+, CD25hi, and CD127low/negative markers characterize the majority of nTreg, it is still imperative to find additional surface-marker combinations that improve the identification/isolation of nTreg and their subsets. Herein, we present evidence that CD4+ CD25hi CD6(lo/-) nTreg have high expression of FOXP3and exhibit in vitro suppressive activity on CD8+ T-cell proliferation. Dot-plot analyses of CD4+ cells, with CD6, CD127, CD49d, or CD26 reveal that a higher enrichment yield of CD25hi FOXP3+ cells can be achieved in the combined CD6(lo/-) CD127(lo/-) population. We conclude that FOXP3+ nTreg cells are characterized by CD6(lo/-) expression, providing a new tool for the identification of nTreg cells without recourse to intracellular staining, and for the purification of these cells by negative selection.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Flow Cytometry/methods , Forkhead Transcription Factors/biosynthesis , T-Lymphocytes, Regulatory/metabolism , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/metabolism
2.
PLoS One ; 5(11): e14062, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-21124918

ABSTRACT

BACKGROUND: The cancer stem cell theory hypothesizes that cancers are perpetuated by cancer stem cells (CSC) or tumor initiating cells (TIC) possessing self-renewal and other stem cell-like properties while differentiated non-stem/initiating cells have a finite life span. To investigate whether the hypothesis is applicable to lung cancer, identification of lung CSC and demonstration of these capacities is essential. METHODOLOGY/PRINCIPAL FINDING: The expression profiles of five stem cell markers (CD34, CD44, CD133, BMI1 and OCT4) were screened by flow cytometry in 10 lung cancer cell lines. CD44 was further investigated by testing for in vitro and in vivo tumorigenecity. Formation of spheroid bodies and in vivo tumor initiation ability were demonstrated in CD44(+) cells of 4 cell lines. Serial in vivo tumor transplantability in nude mice was demonstrated using H1299 cell line. The primary xenografts initiated from CD44(+) cells consisted of mixed CD44(+) and CD44(-) cells in similar ratio as the parental H1299 cell line, supporting in vivo differentiation. Semi-quantitative Real-Time PCR (RT-PCR) showed that both freshly sorted CD44(+) and CD44(+) cells derived from CD44(+)-initiated tumors expressed the pluripotency genes OCT4/POU5F1, NANOG, SOX2. These stemness markers were not expressed by CD44(-) cells. Furthermore, freshly sorted CD44(+) cells were more resistant to cisplatin treatment with lower apoptosis levels than CD44(-) cells. Immunohistochemical analysis of 141 resected non-small cell lung cancers showed tumor cell expression of CD44 in 50.4% of tumors while no CD34, and CD133 expression was observed in tumor cells. CD44 expression was associated with squamous cell carcinoma but unexpectedly, a longer survival was observed in CD44-expressing adenocarcinomas. CONCLUSION/SIGNIFICANCE: Overall, our results demonstrated that stem cell-like properties are enriched in CD44-expressing subpopulations of some lung cancer cell lines. Further investigation is required to clarify the role of CD44 in tumor cell renewal and cancer propagation in the in vivo environment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Hyaluronan Receptors/genetics , Lung Neoplasms/genetics , Neoplastic Stem Cells/metabolism , AC133 Antigen , Aged , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , Flow Cytometry , Gene Expression Profiling , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Hyaluronan Receptors/metabolism , Immunoblotting , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Peptides/genetics , Peptides/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
3.
J Immunol ; 181(3): 1717-27, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18641308

ABSTRACT

Contact sensitivity (CS) is related to delayed-type hypersensitivity and is a well-characterized prototype of T cell-mediated inflammation. However, the inflammatory response associated with CS is additionally dependent on Ag-specific IgM produced by a subpopulation of B cells in response to sensitization. Upon re-exposure to hapten, this IgM mediates rapid vascular activation and subsequent recruitment of proinflammatory T cells to the local site. Interference with this pathway prevents the full development of the classic delayed inflammatory response and is therefore termed the "CS initiation" pathway. In this study, we show that CS initiation is defective in mice deficient in activation-induced deaminase, an enzyme central to the process of somatic hypermutation. Using adoptive transfer experiments, we demonstrate that the defect is specific to a B-1-like population of B cells and that transfer of WT cells reconstitutes CS initiation mechanisms in deficient recipients. We went on to identify a novel subpopulation of Ag-binding B cells in the spleens of sensitized mice that possess initiation activity (CD19(+)CD5(+)Thy-1(int)IgM(high)IgD(high)) that we name "initiator B cells." Analysis of BCR H chain genes isolated from these cells revealed evidence of activation-induced deaminase-mediated somatic hypermutation. The sensitivity of CS initiation to very low amounts of sensitizing hapten suggests that the responsible B cells have increased IgM receptor gene mutations enabling selection to generate Abs with sufficient affinity to mediate the response.


Subject(s)
B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Cytidine Deaminase/immunology , Cytidine Deaminase/metabolism , Dermatitis, Contact/immunology , Lymphocyte Activation/immunology , Animals , Antigens/immunology , B-Lymphocytes/cytology , Cell Differentiation/immunology , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Disease Models, Animal , Immunity, Innate/immunology , Immunoglobulin D/immunology , Immunoglobulin M/immunology , Mice , Mice, Knockout , Phenotype , Sensitivity and Specificity , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology
4.
Clin Lab Med ; 27(3): 453-68, v, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17658402

ABSTRACT

The demonstration that CD T-cell counts can be used to monitor HIV disease progression opened the way to the first clinical application for fluorescence activated cell sorting (FACS) technology. Modern FACS methodologies such multicolor staining and sorting has opened the way to new and constructive therapeutic and clinical applications. This article outlines approaches in which current users can use to improve the quality of their FACS work without undue effort. FACS technology development and the emergence of new software support for this technology are cooperating in this effort.


Subject(s)
Flow Cytometry , Humans
5.
Proc Natl Acad Sci U S A ; 104(11): 4542-6, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360560

ABSTRACT

B-1a cells reside in both the peritoneal cavity and the spleen. LPS stimulates splenic B-1a to differentiate to plasma cells producing natural IgM specific for microbial and self antigens. However, there are conflicting views as to whether the B-1a cells divide before this differentiation occurs, and hence how the resident B-1a population is maintained in the spleen. Studies here resolve this dispute in favor of both sides: we show that (some or all) B-1a cells resident in the spleen respond to LPS by differentiating to plasma cells immediately, without dividing; however, we also show that additional B-1a cells immigrate into the spleen after LPS stimulation and divide at least once before differentiating. Importantly, the studies we presently describe reveal the complex cell migration and differentiation events that collectively underlie the rapid production of natural antibodies in response to in vivo LPS stimulation. Thus, the studies present a different view of the roles that B-1a cells play in the early phases of the innate immune response.


Subject(s)
Antibody-Producing Cells/cytology , Spleen/cytology , Animals , B-Lymphocytes/cytology , CD5 Antigens/biosynthesis , Cell Differentiation , Cell Division , Cell Movement , Flow Cytometry , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Lipopolysaccharides/metabolism , Mice , Mice, Inbred BALB C , Plasma Cells/metabolism , Spleen/immunology , Spleen/metabolism , Syndecan-1/biosynthesis
6.
Curr Opin Immunol ; 19(2): 150-5, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17303402

ABSTRACT

B-1 cells comprise a small percentage of the B lymphocytes that reside in multiple tissues in the mouse, including the peritoneal and pleural cavities. Functionally, B-1 cells participate in innate immunity by producing the majority of the natural IgM in serum, which protects against invading pathogens before the onset of the adaptive immune response. B-1 cells arise from fetal and neonatal progenitors and are distinct from the adult bone marrow progenitors that give rise to follicular and marginal zone B-2 cells. Recent studies have attempted to delineate the progenitors of B-1 cells from those of B-2 cells. Notably, the identification of CD45R(-/lo)CD19(+) B-1 progenitors and expression of two surface determinants, CD138 and major histocompatibility class II antigens, distinguish developing B-1 cells from B-2 cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Hematopoietic Stem Cells/immunology , Lymphopoiesis , Animals , Antigens, Differentiation/analysis , B-Lymphocyte Subsets/cytology , Biomarkers/analysis , Hematopoietic Stem Cells/chemistry , Mice
7.
Blood ; 109(10): 4191-9, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17244677

ABSTRACT

Pbx1, a homeodomain transcription factor that was originally identified as the product of a proto-oncogene in acute pre-B-cell leukemia, is a global regulator of embryonic development. However, embryonic lethality in its absence has prevented an assessment of its role in B-cell development. Here, using Rag1-deficient blastocyst complementation assays, we demonstrate that Pbx1 null embryonic stem (ES) cells fail to generate common lymphoid progenitors (CLPs) resulting in a complete lack of B and NK cells, and a partial impairment of T-cell development in chimeric mice. A critical role for Pbx1 was confirmed by rescue of B-cell development from CLPs following restoration of its expression in Pbx1-deficient ES cells. In adoptive transfer experiments, B-cell development from Pbx1-deficient fetal liver cells was also severely compromised, but not erased, since transient B lymphopoiesis was detected in Rag-deficient recipients. Conditional inactivation of Pbx1 in pro-B (CD19(+)) cells and thereafter revealed that Pbx1 is not necessary for B-cell development to proceed from the pro-B-cell stage. Thus, Pbx1 critically functions at a stage between hematopoietic stem cell development and B-cell commitment and, therefore, is one of the earliest-acting transcription factors that regulate de novo B-lineage lymphopoiesis.


Subject(s)
B-Lymphocytes/cytology , Homeodomain Proteins/genetics , Lymphopoiesis/genetics , Transcription Factors/genetics , Animals , Blastocyst/immunology , Chimera/genetics , Gene Expression Regulation, Developmental , Genes, RAG-1 , Hematopoietic Stem Cells/cytology , Liver/cytology , Liver/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pre-B-Cell Leukemia Transcription Factor 1
8.
Proc Natl Acad Sci U S A ; 103(16): 6293-8, 2006 Apr 18.
Article in English | MEDLINE | ID: mdl-16606838

ABSTRACT

A recent article by Montecino-Rodriguez et al. [Montecino-Rodriguez, E., Leathers, H. & Dorshkind, K. (2006) Nat. Immunol.7, 293-301] has distinguished the early progenitors for B-1 cells, which principally develop in neonates, from early progenitors for B-2 cells, which principally develop in adult bone marrow. Here we introduce syndecan-1 (CD138) and MHC class II (I-A) as markers of early B cell development [Hardy, R. R., Carmack, C. E., Shinton, S. A., Kemp, J. D. & Hayakawa, K. (1991) J. Exp. Med. 173, 1213-1225; Hardy fractions B-D] and show that the expression of these markers distinguishes the predominant B cell development pathway in neonates from the corresponding predominant pathway in adults (both progenitors are present but differently represented in each case). We show that pre-B cells (Hardy fraction D) in the predominant adult pathway express high levels of CD138 and intermediate levels of I-A, whereas the corresponding pre-B cells in the pathway that predominates in neonates do not express either of these markers. As expected, because most of the pre-B cells in adults express CD138, we find that sorted CD138+ adult pre-B cells differentiate to IgM+ B cells in vitro. Sorted CD138- pre-B cells from neonates, the majority subset at this age, also mature to IgM+ cells (without passing through a CD138+ stage). Importantly, our studies here confirm the differential representation of adult and neonatal progenitor populations and further demonstrate that CD138 expression subdivides the adult CD19+, B220-6B2-/low population shown to contain B-1 progenitors in a way consistent with the predominance of B-1b progenitors in adults. Thus, CD138 expression provides a key route to distinguishing early B cell development pathway for what now are clearly three B cell lineages.


Subject(s)
B-Lymphocyte Subsets/immunology , Lymphocyte Activation , Membrane Glycoproteins/analysis , Proteoglycans/analysis , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Lineage , Histocompatibility Antigens Class II/analysis , Immunoglobulin M/immunology , Lymphocyte Activation/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred Strains , Phenotype , Proteoglycans/genetics , Proteoglycans/metabolism , Syndecan-1 , Syndecans
9.
J Immunol ; 176(9): 5321-8, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16621999

ABSTRACT

CD72, a 45-kDa type II transmembrane glycoprotein carrying an ITIM motif, is believed to be an inhibitory coreceptor of the BCR. Mature B cells lacking CD72 show enhanced Ca(2+) mobilization and are hyperproliferative in response to BCR ligation. However, the signal transduction pathways downstream of BCR signaling that transmit the inhibitory effect of CD72 in mature B cells remain unknown. To address this question, we used hen egg lysozyme-specific BCR transgenic mice to elucidate the differential cell signaling between wild-type and CD72-deficient B cells in response to hen egg lysozyme Ag stimulation. Our results demonstrate that CD72 predominantly down-regulates the major signal transduction pathways downstream of the BCR, including NF-AT, NF-kappaB, ERK, JNK, p38-MAPK, and PI3K/Akt in mature B cells. CD72 ligation with anti-CD72 Ab (K10.6), which mimics the binding of CD100 (a natural ligand for CD72) to release the inhibitory function of CD72, augments cell proliferation, Ca(2+) flux, IkappaBalpha activation, and ERK MAPK activity upon Ag stimulation in wild-type B cells. In addition, we show direct evidence that CD72 promotes cell cycle arrest and apoptosis after Ag stimulation in mature B cells. Taken together, our findings conclude that CD72 plays a dominant role as a negative regulator of BCR signaling in primary mature B lymphocytes.


Subject(s)
Antigens, CD/metabolism , Cell Differentiation , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Animals , Antigens, CD/genetics , B-Lymphocytes , Biomarkers , Calcium/metabolism , Cell Survival , Cells, Cultured , Down-Regulation , Enzyme Activation , Gene Expression Regulation , I-kappa B Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Muramidase/pharmacology , NFATC Transcription Factors/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, B-Cell/immunology , Transcription Factor RelA/metabolism
11.
Springer Semin Immunopathol ; 26(4): 347-62, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15633017

ABSTRACT

Natural antibodies are produced at tightly regulated levels in the complete absence of external antigenic stimulation. They provide immediate, early and broad protection against pathogens, making them a crucial non-redundant component of the humoral immune system. These antibodies are produced mainly, if not exclusively, by a subset of long-lived, self-replenishing B cells termed B-1 cells. We argue here that the unique developmental pattern of these B-1 cells, which rests on positive selection by self antigens, ensures production of natural antibodies expressing evolutionarily important specificities that are required for the initial defense against invading pathogens. Positive selection for reactivity with self antigens could also result in the production of detrimental anti-self antibodies. However, B-1 cells have evolved a unique response pattern that minimizes the risk of autoimmunity. Although these cells respond rapidly and strongly to host-derived innate signals, such as cytokines, and to pathogen-encoded signals, such as lipopolysaccharide and phosphorylcholine, they respond very poorly to receptor-mediated activation. In addition, they rarely enter germinal centers and undergo affinity maturation. Thus, their potential for producing high-affinity antibodies with harmful anti-self specificity is highly restricted. The positive selection of B-1 cells occurs during the neonatal period, during which the long-lived self-renewing B-1 population is constituted. Many of these cells (B-1a) express CD5, although a smaller subset (B-1b) does not express this surface marker. Importantly, B-1a cells should not be confused with short-lived anergic B-2 cells, which originate in the bone marrow in adults and initiate CD5 expression and programmed cell death following self-antigen recognition. In summary, we argue here that the mechanisms that enable natural antibody production by B-1 cells reflect the humoral immune system, which has evolved in layers whose distinct developmental mechanisms generate complementary repertoires that collectively operate to maximize flexibility in responses to invading pathogens. B-2 cells, present in what may be the most highly evolved layer(s), express a repertoire that is explicitly selected against self recognition and directed towards the generation of high-affinity antibody response to external antigenic stimuli. B-1 cells, whose repertoire is selected by recognition of self antigen, belong to what may be earlier layer(s) and inherently maintain production of evolutionarily important antibody specificities that respond to pathogen-related, rather then antigen-specific signals.


Subject(s)
Immunity, Innate , Animals , Antibodies, Viral/biosynthesis , Antibody Specificity , Autoantibodies/biosynthesis , Autoantigens , Autoimmunity , B-Lymphocyte Subsets/immunology , Humans , Infections/immunology , Influenza, Human/immunology , Mice
12.
Methods Mol Biol ; 271: 37-58, 2004.
Article in English | MEDLINE | ID: mdl-15146111

ABSTRACT

In the last few years, the effectiveness of developmental and functional studies of individual subsets of cells has increased dramatically owing to the identification of additional subset markers and the extension of fluorescence-activated cell sorter (FACS) capabilities to simultaneously measure the expression of more markers on individual cells. For example, introduction of a 6-8 multiparameter FACS instrument resulted in significant advances in understanding B-cell development. In this chapter, we describe 11-color high-dimensional (Hi-D) FACS staining and data analysis methods that provide greater clarity in identifying the B-cell subsets in bone marrow, spleen, and peritoneal cavity. Further, we show how a single Hi-D FACS antibody reagent combination is sufficient to unambiguously identify most of the currently defined B-cell developmental subsets in the bone marrow (Hardy fractions A-F) and the functional B-cell subsets (B-1a, B-1b, B-2, and marginal zone [MZ] B cells) in the periphery. Although we focus on murine B-cell subsets, the methods we discuss are relevant to FACS studies conducted with all types of cells and other FACS instruments. We introduce a new method for scaling axes for histograms or contour plots of FACS data. This method, which we refer to as Logicle visualization, is particularly useful in promoting correct interpretations of fluorescence-compensated FACS data and visual confirmation of correct compensation values. In addition, it facilitates discrimination of valid subsets. Application of Logicle visualization tools in the Hi-D FACS studies discussed here creates a strong new base for in-depth analysis of B-cell development and function.


Subject(s)
B-Lymphocyte Subsets/cytology , Bone Marrow/immunology , Hematopoietic Stem Cells/cytology , Spleen/cytology , Animals , B-Lymphocyte Subsets/immunology , Cell Separation/methods , Flow Cytometry , Hematopoietic Stem Cells/immunology , Immunophenotyping/methods , Mice , Mice, Inbred BALB C , Software , Spleen/immunology
13.
Clin Immunol ; 110(3): 277-83, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15047205

ABSTRACT

The Fluorescence Activated Cell Sorter (FACS) is an invaluable tool for clinicians and researchers alike in phenotyping and sorting individual cells. With the advances in FACS methodology, notably intracellular staining for cytokines, transcription factors and phosphoproteins, and with increases in the number of fluorescence detection channels, researchers now have the opportunity to study individual cells in far greater detail than previously possible. In this chapter, we discuss High-Definition (Hi-D) FACS methods that can improve analysis of lymphocyte subsets in mouse and man. We focus on the reasons why fluorescence compensation, which is necessary to correct for spectral overlap between two or more fluorochromes used in the same staining combination, is best done as a computed transformation rather than using the analog circuitry available on many flow cytometers. In addition, we introduce a new data visualization method that scales the axes on histograms and two-dimensional contour (or dot) plots to enable visualization of signals from all cells, including those that have minimal fluorescence values and are not properly represented with traditional logarithmic axes. This "Logicle" visualization method, we show, provides superior representations of compensated data and makes correctly compensated data look correct. Finally, we discuss controls that facilitate recognition of boundaries between positive and negative subsets.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Image Processing, Computer-Assisted/methods , Lymphocytes/immunology , Animals , Fluorescence , Fluorescent Dyes , Humans
14.
Proc Natl Acad Sci U S A ; 101(11): 3898-902, 2004 Mar 16.
Article in English | MEDLINE | ID: mdl-15001710

ABSTRACT

The expression of CD5 increases progressively as thymocytes mature. We have shown that CD5 expression is controlled by a tissue-specific regulatory promoter located upstream of the CD5 translation start sites. Deletion of this regulatory promoter, which contains three potential transcription factor binding sites (CCAAT, kappa E2, and ets) reduces the promoter activity to basal level. Of these sites, only ets proved essential for CD5 expression in T cell lines. Here, we introduce a role for the E47 transcription factor and the CD5 promoter kappa E2 site in regulating CD5 expression during thymocyte development. Using T cell lines, we show that (i) mutation of the kappa E2 site in the CD5 regulatory promoter results in a significant elevation of CD5 promoter activity; (ii) the E47 transcription factor binds to the kappa E2 site; and (iii) overexpression of E47 inhibits CD5 expression. We then show, in high-dimensional fluorescence-activated cell sorting studies with primary thymocytes at successive developmental stages, that (i) intracellular E47 levels decrease as surface CD5 expression increases; (ii) E47 expression is down-regulated and CD5 expression is correspondingly up-regulated in DN3 thymocytes in RAG-2-deficient mice injected with anti-CD3 to mimic pre-T cell receptor stimulation; and (iii) E47 expression is down-regulated and CD5 expression is up-regulated when double positive thymocytes are stimulated in vitro with anti-CD3. Based on these data, we propose that E47 negatively regulates CD5 expression by interacting with the kappa E2 site in the CD5 regulatory promoter and that decreases in E47 in response to developmental signals are critical to the progressive increase in CD5 expression as thymocytes mature.


Subject(s)
CD5 Antigens/genetics , DNA-Binding Proteins/metabolism , Promoter Regions, Genetic , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , CD5 Antigens/biosynthesis , Genes, Reporter , Mice , TCF Transcription Factors , Thymus Gland/metabolism , Transcription Factor 7-Like 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...