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











Publication year range
1.
Clin Cancer Res ; 26(13): 3333-3344, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32299814

ABSTRACT

PURPOSE: Although programmed death-ligand 1 (PD-L1) antibody-based therapy has improved the outcome of patients with cancer, acquired resistance to these treatments limits their clinical efficacy. FS118 is a novel bispecific, tetravalent antibody (mAb2) against human lymphocyte activation gene-3 (LAG-3) and PD-L1 with the potential to reinvigorate exhausted immune cells and overcome resistance mechanisms to PD-L1 blockade. Here, using FS118 and a murine surrogate, we characterized the activity and report a novel mechanism of action of this bispecific antibody. EXPERIMENTAL DESIGN: This study characterizes the binding activity and immune function of FS118 in cell lines and human peripheral blood mononuclear cells and further investigates its antitumor activity and mechanism of action using a surrogate murine bispecific antibody (mLAG-3/PD-L1 mAb2). RESULTS: FS118 demonstrated simultaneous binding to LAG-3 and PD-L1 with high affinity and comparable or better activity than the combination of the single component parts of the mAb2 in blocking LAG-3- and PD-L1-mediated immune suppression and enhancing T-cell activity. In syngeneic tumor mouse models, mLAG-3/PD-L1 mAb2 significantly suppressed tumor growth. Mechanistic studies revealed decreased LAG-3 expression on T cells following treatment with the mouse surrogate mLAG-3/PD-L1 mAb2, whereas LAG-3 expression increased upon treatment with the combination of mAbs targeting LAG-3 and PD-L1. Moreover, following binding of mLAG-3/PD-L1 mAb2 to target-expressing cells, mouse LAG-3 is rapidly shed into the blood. CONCLUSIONS: This study demonstrates a novel benefit of the bispecific approach over a combination of mAbs and supports the further development of FS118 for the treatment of patients with cancer.


Subject(s)
Antibodies, Bispecific/pharmacology , Antigens, CD/metabolism , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Lymphocyte Activation/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Animals , Antibody Affinity , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Humans , Immunophenotyping , Lymphocyte Activation/immunology , Mice , Protein Binding , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays , Lymphocyte Activation Gene 3 Protein
2.
Methods ; 154: 60-69, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30208333

ABSTRACT

The immunoglobulin superfamily protein lymphocyte-activation gene 3 (LAG-3) participates in immune suppression and has been identified as a suitable target for cancer therapies. In order to generate bispecific antibodies targeting LAG-3, Fcabs (Fc-region with antigen binding) targeting human and murine LAG-3 were generated from phage libraries. These Fcabs bind to LAG-3, inhibiting its interaction with MHC class II, and induce IL-2 production in a T cell assay. Bispecific antibodies, known as mAb2, were produced by replacing the Fc region of a monoclonal antibody with Fcab sequences in the CH3 domain. mAb2 containing anti-LAG-3 Fcabs have mAb-like biophysical characteristics and retain LAG-3 binding and functional activity. mAb2 can thus be generated using multiple Fabs to investigate bispecific parings and develop novel therapeutics.


Subject(s)
Antibodies, Bispecific , Antigens, CD/immunology , Immunoglobulin Fc Fragments , Animals , Humans , Macaca fascicularis/metabolism , Mice , Protein Engineering , Lymphocyte Activation Gene 3 Protein
3.
Cancer Immunol Res ; 2(2): 121-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24778275

ABSTRACT

The depletion of tumor stromal cells that are marked by their expression of the membrane protein fibroblast activation protein-α (FAP) overcomes immune suppression and allows an anticancer cell immune response to control tumor growth. In subcutaneous tumors established with immunogenic Lewis lung carcinoma cells expressing ovalbumin (LL2/OVA), the FAP(+) population is comprised of CD45(+) and CD45(-) cells. In the present study, we further characterize the tumoral FAP(+)/CD45(+) population as a minor subpopulation of F4/80(hi)/CCR2(+)/CD206(+) M2 macrophages. Using bone marrow chimeric mice in which the primate diphtheria toxin receptor is restricted either to the FAP(+)/CD45(+) or to the FAP(+)/CD45(-) subset, we demonstrate by conditionally depleting each subset that both independently contribute to the immune-suppressive tumor microenvironment. A basis for the function of the FAP(+)/CD45(+) subset is shown to be the immune inhibitory enzyme, heme oxygenase-1 (HO-1). The FAP(+)/CD45(+) cells are the major tumoral source of HO-1, and an inhibitor of HO-1, Sn mesoporphyrin, causes the same extent of immune-dependent arrest of LL2/OVA tumor growth as does the depletion of these cells. Because this observation of immune suppression by HO-1 expressed by the FAP(+)/CD45(+) stromal cell is replicated in a transplanted model of pancreatic ductal adenocarcinoma, we conclude that pharmacologically targeting this enzyme may improve cancer immunotherapy.


Subject(s)
Carcinoma, Lewis Lung/immunology , Gelatinases/metabolism , Heme Oxygenase-1/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , Carcinoma, Lewis Lung/therapy , Endopeptidases , Immune Tolerance , Immunotherapy/methods , Macrophages/immunology , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation
4.
Dev Dyn ; 243(6): 778-90, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24425002

ABSTRACT

BACKGROUND: Protein Phosphatase 2A (PP2A) function is controlled by regulatory subunits that modulate the activity of the catalytic subunit and direct the PP2A complex to specific intracellular locations. To study PP2A's role in signal transduction pathways that control growth and differentiation in vivo, a transgenic mouse lacking the B56γ regulatory subunit of PP2A was made. RESULTS: Lack of PP2A activity specific to the PP2A-B56γ holoenzyme, resulted in the formation of an incomplete ventricular septum and a decrease in the number of ventricular cardiomyocytes. During cardiac development, B56γ is expressed in the nucleus of α-actinin-positive cardiomyocytes that contain Z-bands. The pattern of B56γ expression correlated with the cardiomyocyte apoptosis we observed in B56γ-deficient mice during mid to late gestation. In addition to the cardiac phenotypes, mice lacking B56γ have a decrease in locomotive coordination and gripping strength, indicating that B56γ has a role in controlling PP2A activity required for efficient neuromuscular function. CONCLUSIONS: PP2A-B56γ activity is required for efficient cardiomyocyte maturation and survival. The PP2A B56γ regulatory subunit controls PP2A substrate specificity in vivo in a manner that cannot be fully compensated for by other B56 subunits.


Subject(s)
Embryo, Mammalian/enzymology , Heart Septum/embryology , Heart Ventricles/embryology , Myocytes, Cardiac/enzymology , Protein Phosphatase 2/metabolism , Animals , Embryo, Mammalian/cytology , Heart Septum/cytology , Mice , Mice, Knockout , Mice, Obese , Myocytes, Cardiac/cytology , Protein Phosphatase 2/genetics
5.
Proc Natl Acad Sci U S A ; 110(50): 20212-7, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277834

ABSTRACT

An autochthonous model of pancreatic ductal adenocarcinoma (PDA) permitted the analysis of why immunotherapy is ineffective in this human disease. Despite finding that PDA-bearing mice had cancer cell-specific CD8(+) T cells, the mice, like human patients with PDA, did not respond to two immunological checkpoint antagonists that promote the function of T cells: anti-cytotoxic T-lymphocyte-associated protein 4 (α-CTLA-4) and α-programmed cell death 1 ligand 1 (α-PD-L1). Immune control of PDA growth was achieved, however, by depleting carcinoma-associated fibroblasts (CAFs) that express fibroblast activation protein (FAP). The depletion of the FAP(+) stromal cell also uncovered the antitumor effects of α-CTLA-4 and α-PD-L1, indicating that its immune suppressive activity accounts for the failure of these T-cell checkpoint antagonists. Three findings suggested that chemokine (C-X-C motif) ligand 12 (CXCL12) explained the overriding immunosuppression by the FAP(+) cell: T cells were absent from regions of the tumor containing cancer cells, cancer cells were coated with the chemokine, CXCL12, and the FAP(+) CAF was the principal source of CXCL12 in the tumor. Administering AMD3100, a CXCL12 receptor chemokine (C-X-C motif) receptor 4 inhibitor, induced rapid T-cell accumulation among cancer cells and acted synergistically with α-PD-L1 to greatly diminish cancer cells, which were identified by their loss of heterozygosity of Trp53 gene. The residual tumor was composed only of premalignant epithelial cells and inflammatory cells. Thus, a single protein, CXCL12, from a single stromal cell type, the FAP(+) CAF, may direct tumor immune evasion in a model of human PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Chemokine CXCL12/metabolism , Gelatinases/metabolism , Immunotherapy/methods , Membrane Proteins/metabolism , Pancreatic Neoplasms/therapy , Serine Endopeptidases/metabolism , Tumor Escape/genetics , Analysis of Variance , Animals , Base Sequence , Benzylamines , Carcinoma, Pancreatic Ductal/immunology , Cyclams , Endopeptidases , Enzyme-Linked Immunospot Assay , Fibroblasts/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Heterocyclic Compounds , Immunohistochemistry , Mice , Molecular Sequence Data , Pancreatic Neoplasms/immunology , Sequence Analysis, RNA
6.
J Exp Med ; 210(6): 1137-51, 2013 Jun 03.
Article in English | MEDLINE | ID: mdl-23712428

ABSTRACT

Fibroblast activation protein-α (FAP) identifies stromal cells of mesenchymal origin in human cancers and chronic inflammatory lesions. In mouse models of cancer, they have been shown to be immune suppressive, but studies of their occurrence and function in normal tissues have been limited. With a transgenic mouse line permitting the bioluminescent imaging of FAP(+) cells, we find that they reside in most tissues of the adult mouse. FAP(+) cells from three sites, skeletal muscle, adipose tissue, and pancreas, have highly similar transcriptomes, suggesting a shared lineage. FAP(+) cells of skeletal muscle are the major local source of follistatin, and in bone marrow they express Cxcl12 and KitL. Experimental ablation of these cells causes loss of muscle mass and a reduction of B-lymphopoiesis and erythropoiesis, revealing their essential functions in maintaining normal muscle mass and hematopoiesis, respectively. Remarkably, these cells are altered at these sites in transplantable and spontaneous mouse models of cancer-induced cachexia and anemia. Thus, the FAP(+) stromal cell may have roles in two adverse consequences of cancer: their acquisition by tumors may cause failure of immunosurveillance, and their alteration in normal tissues contributes to the paraneoplastic syndromes of cachexia and anemia.


Subject(s)
Anemia/metabolism , Bone Marrow/metabolism , Cachexia/metabolism , Gelatinases/genetics , Gelatinases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle, Skeletal/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Stromal Cells/metabolism , Adipose Tissue/metabolism , Anemia/genetics , Anemia/pathology , Animals , Cachexia/genetics , Cachexia/pathology , Cell Lineage/genetics , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Endopeptidases , Erythropoiesis/genetics , Follistatin/genetics , Follistatin/metabolism , Hematopoiesis/genetics , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Muscle, Skeletal/cytology , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Pancreas/metabolism , Stromal Cells/cytology , Transcriptome/genetics
7.
J Immunol ; 185(12): 7141-5, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21068412

ABSTRACT

Persistent viral infections induce the differentiation and accumulation of large numbers of senescent CD8(+) T cells, raising the possibility that repetitive stimulation drives clones of T cells to senesce. It is therefore unclear whether T cell responses are maintained by the self-renewal of Ag-experienced peripheral T cell subsets or by the continuous recruitment of newly generated naive T cells during chronic infections. Using a transgenic mouse model that permits the indelible marking of granzyme B-expressing cells, we found that T cells primed during the initial stages of a persistent murine γ-herpes infection persisted and continued to divide during a latent phase of up to 7 mo. Such cells maintained an ability to extensively replicate in response to challenge with influenza virus expressing the same Ag. Therefore, Ag-experienced, virus-specific CD8(+) T cell populations contain a subset that maintains replicative potential, despite long-term, persistent antigenic stimulation.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Granzymes/immunology , Herpesviridae Infections/immunology , Rhadinovirus/immunology , Animals , Antigens, Viral/metabolism , CD8-Positive T-Lymphocytes/metabolism , Granzymes/biosynthesis , Herpesviridae Infections/genetics , Herpesviridae Infections/metabolism , Mice , Mice, Transgenic , Rhadinovirus/genetics , Rhadinovirus/metabolism , Time Factors
8.
Science ; 330(6005): 827-30, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-21051638

ABSTRACT

The stromal microenvironment of tumors, which is a mixture of hematopoietic and mesenchymal cells, suppresses immune control of tumor growth. A stromal cell type that was first identified in human cancers expresses fibroblast activation protein-α (FAP). We created a transgenic mouse in which FAP-expressing cells can be ablated. Depletion of FAP-expressing cells, which made up only 2% of all tumor cells in established Lewis lung carcinomas, caused rapid hypoxic necrosis of both cancer and stromal cells in immunogenic tumors by a process involving interferon-γ and tumor necrosis factor-α. Depleting FAP-expressing cells in a subcutaneous model of pancreatic ductal adenocarcinoma also permitted immunological control of growth. Therefore, FAP-expressing cells are a nonredundant, immune-suppressive component of the tumor microenvironment.


Subject(s)
Carcinoma, Lewis Lung/immunology , Carcinoma, Pancreatic Ductal/immunology , Gelatinases/metabolism , Immune Tolerance , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Stromal Cells/immunology , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Endopeptidases , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Transgenic , Necrosis , Neoplasm Transplantation , Stromal Cells/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
9.
Eur J Immunol ; 39(8): 2083-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19637204

ABSTRACT

CD8(+) T-cell responses must have at least two components, a replicative cell type that proliferates in the secondary lymphoid tissue and that is responsible for clonal expansion, and cytotoxic cells with effector functions that mediate the resolution of the infection in the peripheral tissues. To confer memory, the response must also generate replication-competent T cells that persist in the absence of antigen after the primary infection is cleared. The current models of memory differentiation differ in regards to whether or not memory CD8(+) T cells acquire effector functions during their development. In this review we discuss the existing models for memory development and the consequences that the recent finding that memory CD8(+) T cells may express granzyme B during their development has for them. We propose that memory CD8(+) T cells represent a self-renewing population of T cells that may acquire effector functions but that do not lose the naïve-like attributes of lymphoid homing, antigen-independent persistence or the capacity for self-renewal.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Signal Transduction/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Proliferation , Humans , Lymphocyte Activation/immunology , Models, Immunological , Virus Diseases/immunology
10.
Genesis ; 47(8): 573-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19530136

ABSTRACT

The Notch pathway is an intercellular signaling mechanism frequently used for controlling cell fate during organogenesis. There are four structurally related Notch receptors in mice and humans, and Notch1 and Notch2 are essential genes. In this report we describe the construction of a transgenic mouse strain that expresses the Notch2 intracellular domain in response to cell lineage specific expression of Cre recombinase. This approach bypasses the requirement for ligand- receptor interaction and allows the direct determination of the consequences of Notch2 activation in vivo. Exogenous expression of the Notch2 intracellular domain resulted in the developmental arrest of secondary heart field derived cardiomyocytes during the transition from immature alpha-Smooth Muscle Actin expressing cells to mature alpha-Actinin positive cardiomyocytes. In contrast, a cell nonautonomous mesenchymal expansion was observed in semilunar valves. This new conditionally expressed allele of Notch2 can be used in studies by investigators interested in the effects of Notch2 activation in vivo.


Subject(s)
Receptor, Notch2/genetics , Animals , Base Sequence , Cell Proliferation , DNA Primers , Female , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Pregnancy
11.
Science ; 323(5913): 505-9, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19164749

ABSTRACT

Models of the differentiation of memory CD8+ T cells that replicate during secondary infections differ over whether such cells had acquired effector function during primary infections. We created a transgenic mouse line that permits mapping of the fate of granzyme B (gzmB)-expressing CD8+ T cells and their progeny by indelibly marking them with enhanced yellow fluorescent protein (EYFP). Virus-specific CD8+ T cells express gzmB within the first 2 days of a primary response to infection with influenza, without impairment of continued primary clonal expansion. On secondary infection, virus-specific CD8+ T cells that became EYFP+ during a primary infection clonally expand as well as all virus-specific CD8+ T cells. Thus, CD8+ T cells that have acquired an effector phenotype during primary infection may function as memory cells with replicative function.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Orthomyxoviridae Infections/immunology , Animals , Cell Differentiation , Cell Division , Granzymes/genetics , Granzymes/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lung/immunology , Mice , Mice, Transgenic , Phenotype , Spleen/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology
12.
Dev Dyn ; 237(4): 1144-52, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18330927

ABSTRACT

Mutations in Notch receptors and their ligands have been identified as the cause of human congenital heart diseases, indicating the importance of the Notch signaling pathway during heart development. In our study, we use Cre-Lox technology to inactivate Notch2 in several cardiac cell lineages to determine the functional requirements for Notch2 during mammalian heart development. Inactivation of Notch2 in cardiac neural crest cells resulted in abnormally narrow aortas and pulmonary arteries due to a decrease in smooth muscle tissue. The reduction in smooth muscle tissue was not due to cell migration defects but instead was found to be caused by less proliferation in smooth muscle cells during mid to late gestation. Our findings demonstrate that Notch2 is required cell autonomously for proper formation of the heart outflow tract and provides insights into the role of Notch2 in vascular smooth muscle development and the cardiovascular defects associated with Alagille syndrome.


Subject(s)
Cell Proliferation , Heart , Myocytes, Smooth Muscle/physiology , Neural Crest/cytology , Receptor, Notch2/metabolism , Actins/metabolism , Animals , Arteries/anatomy & histology , Arteries/diagnostic imaging , Arteries/metabolism , Cell Lineage , Cell Movement/physiology , Female , Genes, Reporter , Genotype , Heart/anatomy & histology , Heart/embryology , Humans , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/cytology , Phenotype , Receptor, Notch2/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ultrasonography
13.
Proc Natl Acad Sci U S A ; 103(51): 19454-9, 2006 Dec 19.
Article in English | MEDLINE | ID: mdl-17159138

ABSTRACT

The clonal expansion of antigen-specific CD8+ T cells in response to microbial infections is essential for adaptive immunity. Although IL-2 has been considered to be primarily responsible for this process, quantitatively normal expansion occurs in the absence of IL-2 receptor signaling. Here, we show that ligating CD27 on CD8+ T cells that have been stimulated through the T cell receptor causes their expansion in the absence of IL-2 by mediating two distinct cellular processes: enhancing cell cycling and promoting cell survival by maintaining the expression of IL-7 receptor alpha. This pathway for clonal expansion of the CD8+ T cell is not associated with the development of a capacity either for production of IFN-gamma or for cytotoxic T lymphocyte function and, therefore, is uncoupled from differentiation. Furthermore, ligating CD27 increases the threshold concentration at which IL-2 induces IFN-gamma-producing capability by the CD8+ T cell, suggesting that CD27 signaling may suppress effector differentiation. Finally, CD8+ T cells that have been stimulated by the TCR/CD27 pathway maintain their capacity for subsequent expansion and effector differentiation in response to a viral challenge in vivo. Thus, the TCR/CD27 pathway enables the CD8+ T cell to replicate by a process of self-renewal, which may contribute to the continuous generation of new effector CD8+ T cells in persistent viral infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Proliferation , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Mice , Receptors, Interleukin-7/metabolism
14.
FASEB J ; 19(10): 1311-3, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15897231

ABSTRACT

The Notch receptor is a key component of a highly conserved signaling pathway that regulates cell fate determination during development. In Drosophila, where Notch signaling was first identified and studied, there is only one Notch receptor. In contrast, mammals have four Notch receptor genes, Notch1-4. Notch1 and Notch2 are both required for embryo viability, are widely expressed in mammals, and are structurally conserved. It is presently unknown if these two receptors are functionally redundant or if they have unique capabilities related to differences in their amino acid sequences. In contrast to the rest of the molecule, the amino acid sequences of a large region of the Notch intracellular domain are not highly conserved and thus may be able to interact with distinct transcription factors and mediate the expression of different sets of genes. To determine if the function of this region is conserved, the last 426 amino acids of the Notch2 receptor have been replaced with the corresponding region of Notch1 in mice by using gene targeting. We have determined that even though the amino acid sequences of this region are only 37% identical (137/426), the C-terminal region of the Notch1 intracellular domain can functionally replace that of Notch2 in vivo.


Subject(s)
Receptor, Notch1/physiology , Receptor, Notch2/physiology , Alleles , Animals , Conserved Sequence , Female , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Phenotype , Protein Structure, Tertiary , RNA, Messenger/analysis , Receptor, Notch1/chemistry , Receptor, Notch1/genetics , Receptor, Notch2/chemistry , Receptor, Notch2/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
15.
Oncogene ; 22(36): 5572-8, 2003 Aug 28.
Article in English | MEDLINE | ID: mdl-12944904

ABSTRACT

Appropriately timed proliferation, differentiation and apoptosis are essential to the normal functions of the mammary epithelium. Here, we report that the transcription factor BCL-6 is expressed in mammary epithelium in nonpregnant animals as well as during early pregnancy. When overexpressed in the nontransformed EpH4 mammary epithelial cell line, BCL-6 prevents the STAT-driven expression of the milk protein beta-casein and duct formation, and prevents apoptosis. Consistent with an antiapoptotic function, we demonstrate that BCL-6 is expressed in 68% of histologically high-grade ductal breast carcinomas, which are clinically the most aggressive. BCL-6 has previously been characterized as a regulator of B lymphocyte growth and development, but our work identifies a novel role for it in mammary epithelial differentiation, which may also implicate it in carcinogenesis.


Subject(s)
Breast Neoplasms/chemistry , DNA-Binding Proteins/physiology , Mammary Glands, Animal/cytology , Milk Proteins , Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , Animals , Apoptosis , Breast Neoplasms/etiology , Cell Differentiation , Cell Division , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Female , Humans , Mice , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-6 , RNA, Messenger/analysis , STAT5 Transcription Factor , Trans-Activators/metabolism , Transcription Factors/analysis , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL