Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Infect Immun ; 69(3): 1593-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179332

ABSTRACT

Microbial targets for protective humoral immunity are typically surface-localized proteins and contain common sequence motifs related to their secretion or surface binding. Exploiting the whole genome sequence of the human bacterial pathogen Streptococcus pneumoniae, we identified 130 open reading frames encoding proteins with secretion motifs or similarity to predicted virulence factors. Mice were immunized with 108 of these proteins, and 6 conferred protection against disseminated S. pneumoniae infection. Flow cytometry confirmed the surface localization of several of these targets. Each of the six protective antigens showed broad strain distribution and immunogenicity during human infection. Our results validate the use of a genomic approach for the identification of novel microbial targets that elicit a protective immune response. These new antigens may play a role in the development of improved vaccines against S. pneumoniae.


Subject(s)
Genomics/methods , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/therapeutic use , Streptococcus pneumoniae/genetics , Technology, Pharmaceutical/methods , Amino Acid Sequence , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/therapeutic use , Bacterial Vaccines , Conserved Sequence , Convalescence , Female , Humans , Mice , Mice, Inbred C3H , Molecular Sequence Data , Pneumococcal Infections/mortality , Pneumococcal Vaccines/genetics , Sepsis/mortality , Sepsis/prevention & control , Serotyping , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/immunology
2.
Exp Cell Res ; 257(2): 310-9, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10837145

ABSTRACT

The mammalian gamma subunit family consists of a minimum of 12 members. Analysis of the amino acid sequence conservation suggests that the gamma subunit family can be divided into three distinct subclasses. The division of the gamma subunit family into these classes is based not only on amino acid homology, but also to some extent on functional similarities. In the present study, two new members of the gamma subunit family, the gamma(11) and gamma(14) subunits, are identified and characterized in terms of their expression and function. The gamma(11) and gamma(14) subunits are most closely related to the gamma(1) subunit and share similar biochemical properties, suggesting their inclusion in class I. However, despite their close phylogenetic relationship and similar biochemical properties, the gamma(1), gamma(11), and gamma(14) subunits exhibit very distinct expression patterns, suggesting that class I should be further subdivided and that the signaling functions of each subgroup are distinct. In this regard, the gamma(11) and gamma(14) subunits represent a new subgroup of farnesylated gamma subunits that are expressed outside the retina and have functions other than phototransduction.


Subject(s)
GTP-Binding Proteins/classification , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cattle , Cell Line , Cloning, Molecular , DNA, Complementary , GTP-Binding Proteins/genetics , GTP-Binding Proteins/physiology , Gene Expression , Humans , Molecular Sequence Data , RNA, Messenger
3.
J Biol Chem ; 275(2): 1216-25, 2000 Jan 14.
Article in English | MEDLINE | ID: mdl-10625666

ABSTRACT

Prostate cancer, the most frequent solid cancer in older men, is a leading cause of cancer deaths. Although proliferation and differentiation of normal prostate epithelia and the initial growth of prostate cancer cells are androgen-dependent, prostate cancers ultimately become androgen-independent and refractory to hormone therapy. The prostate-specific antigen (PSA) gene has been widely used as a diagnostic indicator for androgen-dependent and -independent prostate cancer. Androgen-induced and prostate epithelium-specific PSA expression is regulated by a proximal promoter and an upstream enhancer via several androgen receptor binding sites. However, little progress has been made in identifying androgen-independent regulatory elements involved in PSA gene regulation. We report the isolation of a novel, prostate epithelium-specific Ets transcription factor, PDEF (prostate-derived Ets factor), that among the Ets family uniquely prefers binding to a GGAT rather than a GGAA core. PDEF acts as an androgen-independent transcriptional activator of the PSA promoter. PDEF also directly interacts with the DNA binding domain of androgen receptor and enhances androgen-mediated activation of the PSA promoter. Our results, as well as the critical roles of other Ets factors in cellular differentiation and tumorigenesis, strongly suggest that PDEF is an important regulator of prostate gland and/or prostate cancer development.


Subject(s)
Gene Expression Regulation , Prostate-Specific Antigen/genetics , Prostate/metabolism , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites , Cell Line , Cells, Cultured , Humans , Keratinocytes , Male , Molecular Sequence Data , Promoter Regions, Genetic , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/chemistry , Transcriptional Activation , Tumor Cells, Cultured
4.
Circ Res ; 85(8): 753-66, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10521248

ABSTRACT

Oxidative stress and the production of intracellular reactive oxygen species (ROS) have been implicated in the pathogenesis of a variety of diseases. In excess, ROS and their byproducts that are capable of causing oxidative damage may be cytotoxic to cells. However, it is now well established that moderate amounts of ROS play a role in signal transduction processes such as cell growth and posttranslational modification of proteins. Oxidants, antioxidants, and other determinants of the intracellular reduction-oxidation (redox) state play an important role in the regulation of gene expression. Recent insights into the etiology and pathogenesis of atherosclerosis suggest that this disease may be viewed as an inflammatory disease linked to an abnormality in oxidation-mediated signals in the vasculature. In this review, we summarize the evidence supporting the notion that oxidative stress and the production of ROS function as physiological regulators of vascular gene expression mediated via specific redox-sensitive signal transduction pathways and transcriptional regulatory networks. Elucidating, at the molecular level, the regulatory processes involved in redox-sensitive vascular gene expression represents a foundation not only for understanding the pathogenesis of atherosclerosis and other inflammatory diseases but also for the development of novel therapeutic treatment strategies.


Subject(s)
Blood Vessels/physiology , Gene Expression Regulation , Oxidative Stress/genetics , Animals , Arteriosclerosis/etiology , Blood Vessels/pathology , Blood Vessels/physiopathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Risk Factors , Signal Transduction
5.
J Biol Chem ; 274(41): 29439-52, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10506207

ABSTRACT

Epithelial cell differentiation is tightly controlled by distinct sets of transcription factors that regulate the expression of stage-specific genes. We recently isolated the first epithelium-specific Ets transcription factor (ESE-1). Here we describe the characterization of ESE-2, a second epithelium-restricted ESE-1-related Ets factor. Like ESE-1, ESE-2 is induced during keratinocyte differentiation. However, whereas ESE-1 is expressed in the majority of epithelial cell types, ESE-2 expression is restricted to differentiated keratinocytes and glandular epithelium such as salivary gland, prostate, mammary gland, and kidney. In contrast to ESE-1, full-length ESE-2 binds poorly to DNA due to the presence of a negative regulatory domain at the amino terminus. Furthermore, although ESE-1 and the amino-terminally deleted ESE-2 bind with similar affinity to the canonical E74 Ets site, ESE-2 and ESE-1 differ strikingly in their relative affinity toward binding sites in the c-MET and PSMA promoters. Similarly, ESE-1 and ESE-2 drastically differ in their ability to transactivate epithelium-specific promoters. Thus, ESE-2, but not ESE-1, transactivates the parotid gland-specific PSP promoter and the prostate-specific PSA promoter. In contrast, ESE-1 transactivates the keratinocyte-specific SPRR2A promoter Ets site and the prostate-specific PSMA promoter significantly better than ESE-2. Our results demonstrate the existence of a unique class of related epithelium-specific Ets factors with distinct functions in epithelial cell gene regulation.


Subject(s)
Proto-Oncogene Proteins , Transcription Factors/genetics , Transcriptional Activation/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Differentiation , Cell Line , Cloning, Molecular , Cornified Envelope Proline-Rich Proteins , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Humans , Keratinocytes , Membrane Proteins/genetics , Molecular Sequence Data , Parotid Gland , Promoter Regions, Genetic , Prostate-Specific Antigen/genetics , Protein Precursors/genetics , Proto-Oncogene Proteins c-ets , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid , Salivary Proteins and Peptides/genetics , Sequence Alignment , Trans-Activators/chemistry , Transcription Factors/chemistry
6.
J Bacteriol ; 181(5): 1436-43, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10049373

ABSTRACT

The acquisition of iron by pathogenic bacteria is often a crucial step in establishing infection. To accomplish this, many bacteria, including Staphylococcus aureus, produce low-molecular-weight iron-chelating siderophores. However, the secretion and transport of these molecules in gram-positive organisms are poorly understood. The sequence, organization, and regulation of genes involved in siderophore transport are conserved among gram-negative bacteria. We used this information to identify a putative siderophore transport locus from an S. aureus genomic sequence database. This locus contains three predicted open reading frames with a high degree of homology to genes involved in siderophore uptake in several bacterial species, in particular the cbr locus of the plant pathogen Erwinia chrysanthemi. The first gene in the locus, which we have designated sir for staphylococcal iron regulated, encodes a putative lipoprotein with a molecular mass of 37 kDa. The open reading frame is preceded by a 19-bp region of dyad symmetry with homology for operator sequences controlling iron-regulated expression of genes in other bacteria. Fur titration experiments indicate that this region of dyad symmetry is sufficient for Fur-dependent regulation in Escherichia coli. The expression of this gene was repressed, in a dose-dependent manner, by the addition of iron to the S. aureus culture medium. sir-encoded proteins may be involved in iron acquisition in vivo and therefore may be targets for antimicrobial agents.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Iron/metabolism , Staphylococcus aureus/metabolism , Amino Acid Sequence , Bacterial Proteins/isolation & purification , Base Sequence , Binding Sites , Cloning, Molecular , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/drug effects , Humans , Iron/pharmacology , Kinetics , Molecular Sequence Data , Plasmids , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/isolation & purification , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism
8.
Mol Cell Biol ; 17(8): 4419-33, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9234700

ABSTRACT

We report here the isolation of a novel, highly tissue-restricted member of the ets transcription factor/oncogene family, ESE-1 (for epithelium-specific Ets), which has features distinct from those of any other ets-related factor. ESE-1 contains two putative DNA binding domains: an ETS domain, which is unique in that the 5' half shows relatively weak homology to known ets factors, and an A/T hook domain, found in HMG proteins and various other nuclear factors. In contrast to any known ets factors, ESE-1 is expressed exclusively in epithelial cells. ESE-1 expression is induced during terminal differentiation of the epidermis and in a primary human keratinocyte differentiation system. The keratinocyte terminal differentiation marker gene, SPRR2A, is a putative target for ESE-1, since SPRR2A expression during keratinocyte differentiation correlates with induction of ESE-1 expression, and ESE-1 binds with high affinity to and transactivates the ets binding site in the SPRR2A promoter. ESE-1 also binds to and transactivates the enhancer of the Endo A gene, a potential target for ESE-1 in simple epithelia. Due to the important role that other ets factors play in cellular differentiation, ESE-1 is expected to be a critical regulator of epithelial cell differentiation.


Subject(s)
Keratinocytes/chemistry , Trans-Activators/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Cells, Cultured , Cloning, Molecular , Cornified Envelope Proline-Rich Proteins , DNA-Binding Proteins/genetics , Epidermal Cells , Epithelial Cells , Epithelium/chemistry , Gene Expression Regulation, Developmental , High Mobility Group Proteins/genetics , Humans , Keratinocytes/cytology , Keratins/genetics , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Nuclear Proteins , Organ Specificity , Protein Precursors/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets , RNA, Messenger/analysis , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Trans-Activators/chemistry , Transcription Factors/genetics
10.
Mol Cell Biol ; 16(9): 5091-106, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8756667

ABSTRACT

We have cloned the gene for a novel Ets-related transcription factor, new Ets-related factor (NERF), from human spleen, fetal liver, and brain. Comparison of the deduced amino acid sequence of NERF with those of other members of the Ets family reveals that the level of homology to ELF-1, which is involved in the regulation of several T- and B-cell-specific genes, is highest. Homologies are clustered in the putative DNA binding domain in the middle of the protein, a basic domain just upstream of this domain, and several shorter stretches of homology towards the amino terminus. The presence of two predominant NERF transcripts in various fetal and adult human tissues is due to at least three alternative splice products, NERF-1a, NERF-1b, and NERF-2, which differ in their amino termini and their expression in different tissues. Only NERF-2 and ELF-1, and not NERF-1a and NERF-1b, function as transcriptional activators of the lyn and blk gene promoters, although all isoforms of NERF bind with affinities similar to those of ELF-1 to a variety of Ets binding sites in, among others, the blk, lck, lyn, mb-1, and immunoglobulin H genes and are expressed at similar levels. Since NERF and ELF-1 are coexpressed in B and T cells, both might be involved in the regulation of the same genes.


Subject(s)
DNA-Binding Proteins/chemistry , Genes , Transcription Factors/chemistry , Transcription Factors/isolation & purification , Adult , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Cell Line, Transformed , Chlorocebus aethiops , Cloning, Molecular , Fetal Proteins/isolation & purification , Fibroblasts/metabolism , Gene Expression Regulation , HeLa Cells/metabolism , Humans , Mice , Molecular Sequence Data , Multigene Family , Nuclear Proteins , Organ Specificity , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Regulatory Sequences, Nucleic Acid , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/physiology , Transcriptional Activation , src-Family Kinases/genetics
11.
J Biol Chem ; 270(37): 21765-71, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7665596

ABSTRACT

With the growing awareness that the G protein beta and gamma subunits directly regulate the activities of various enzymes and ion channels, the importance of identifying and characterizing these subunits is underscored. In this paper, we report the isolation of cDNA clones encoding eight different human gamma subunits, including three novel forms designated gamma 4, gamma 10, and gamma 11. The predicted protein sequence of gamma 4 shares the most identity (60-77%) with gamma 2, gamma 3, and gamma 7 and the least identity (38%) with gamma 1. The gamma 4 is modified by a geranylgeranyl group and is capable of interacting with both beta 1 and beta 2 but not with beta 3. The predicted protein sequence of gamma 10 shows only modest to low identity (35-53%) with the other known gamma subunits, with most of the differences concentrated in the N-terminal region, suggesting gamma 10 may interact with a unique subclass of alpha. The gamma 10 is modified by a geranylgeranyl group and is capable of interacting with beta 1 and beta 2 but not with beta 3. Finally, the predicted protein sequence of gamma 11 shows the most identity to gamma 1 (76% identity) and the least identity to the other known gamma (33-44%). Unlike most of the other known gamma subunits, gamma 11 is modified by a farnesyl group and is not capable of interacting with beta 2. The close resemblance of gamma 11 to gamma 1 raises intriguing questions regarding its function since the mRNA for gamma 11 is abundantly expressed in all tissues tested except for brain, whereas the mRNA for gamma 1 is expressed only in the retina where the protein functions in phototransduction.


Subject(s)
GTP-Binding Proteins/biosynthesis , GTP-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cattle , Cloning, Molecular/methods , DNA, Complementary/isolation & purification , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , GTP-Binding Proteins/isolation & purification , Humans , Macromolecular Substances , Molecular Sequence Data , Plasmids , Protein Biosynthesis , Protein Prenylation , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Homology, Amino Acid , Transcription, Genetic
12.
Mol Cell Neurosci ; 6(3): 230-51, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7496629

ABSTRACT

Cerebellar Purkinje cells were ablated by the specific expression of diphtheria toxin in these cells in transgenic mice. Purkinje cell degeneration during early postnatal development shows a zonally restricted pattern which has been exploited in order to look for local secondary effects. The most obvious early effect is the alignment of gaps in the Purkinje cell layer with dramatically thinned zones in the overlying EGL, the germinal layer from which granule cells are generated. Within these EGL zones in the transgenic mutant, markers that distinguish matrix from mantle cells demonstrate a preferential loss of the proliferative cells. Comparison of BrdU incorporation in the mutant vs wild-type confirms the reduction in proliferation. In the mutant, in situ labeling of DNA fragmentation associated with apoptotic cell death shows abundant labeling of granule cells that have exited the EGL, but not of progenitor cells in the EGL. Thus, although a trophic role for Purkinje cells has been well documented, these observations further suggest a mitogenic role which can be exerted locally.


Subject(s)
Cell Division , Cerebellum/drug effects , Purkinje Cells/physiology , Animals , Autoradiography , Cerebellum/ultrastructure , Diphtheria , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Transgenic , Mitogens
13.
J Immunol ; 153(1): 153-64, 1994 Jul 01.
Article in English | MEDLINE | ID: mdl-8207232

ABSTRACT

Transcriptional activation of the IL-8 gene by several inflammatory mediators, including the cytokines IL-1 and TNF-alpha, is mediated through sequences located between nucleotide -94 and -71 of the IL-8 promoter. Because adjacent binding sites for the inducible transcription factors NF-kappa B and NF-IL-6 are located within this region, we examined the functional interaction of these two transcription factor families in IL-8 gene regulation. Maximal transcriptional activation by PMA in Jurkat T lymphocytes was shown to require intact binding sites for both NF-kappa B and NF-IL-6. Electrophoretic mobility shift analysis indicates that NF-IL-6, as well as other related members of this family, bind specifically to the NF-IL-6 site in the IL-8 promoter. In addition, NF-kappa B p65 (RelA), but not NF-kappa B p50 (NFKB1), binds specifically to the NF-kappa B site. When incubated together, RelA and NF-IL-6/C/EBP form a ternary complex with this region of the IL-8 promoter; this binding is dependent on intact binding sites for both NF-IL-6 and RelA. Transient cotransfection analyses indicate that the cooperative association of NF-IL-6 and RelA with the IL-8 promoter results in synergistic transcriptional activation. Mutational analyses of RelA demonstrate that the C-terminal transactivation domain and the DNA binding domain are required for synergistic activation with NF-IL-6. In addition, overexpression of the NF-kappa B inhibitor molecule, I kappa B, abolished the RelA- and RelA/NF-IL-6-dependent synergistic activation. These data demonstrate that RelA and members of the C/EBP/NF-IL-6 family can functionally cooperate in transcriptional activation of the IL-8 gene and suggest a common mechanism for inducible regulation of cytokine gene expression.


Subject(s)
DNA-Binding Proteins/administration & dosage , Gene Expression Regulation , Interleukin-8/genetics , NF-kappa B/administration & dosage , Nuclear Proteins/administration & dosage , Promoter Regions, Genetic , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Drug Synergism , Humans , In Vitro Techniques , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/pharmacology , Oligodeoxyribonucleotides/chemistry , RNA, Messenger/genetics , Recombinant Proteins , Structure-Activity Relationship , Transcription, Genetic , Tumor Cells, Cultured
14.
Mol Cell Biol ; 14(6): 3772-81, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8196620

ABSTRACT

Exposure of monocytic cells to bacterial lipopolysaccharide (LPS) activates the NF-kappa B/Rel family of proteins and leads to the rapid induction of inflammatory gene products, including tissue factor (TF). TF is the primary cellular initiator of the coagulation protease cascades. Here we report the characterization of a nuclear complex from human monocytic cells that bound to a kappa B-like site, 5'-CGGAGTTTCC-3', in the 5'-flanking region of the human TF gene. This nuclear complex was activated by LPS with kinetics that preceded induction of the TF gene. In vitro binding studies demonstrated that the TF site bound translated c-Rel and p65 homodimers but not p50/p65 heterodimers or p50 homodimers. Base-pair substitutions in the TF site indicated that the presence of a cytosine at position 1 precluded binding of NF-kappa B. In fact, under low-ionic-strength conditions, the TF complex did not migrate with translated p50/p65 dimers but instead comigrated with c-Rel/p65 dimers. Antibodies against the NF-kappa B and Rel proteins and UV cross-linking studies revealed the presence of c-Rel and p65 and the absence of p50 in the TF complex and further showed that c-Rel/p65 heterodimers selectively bound to the TF kappa B-like site. Functional studies indicated that the TF site conferred LPS inducibility on a heterologous promoter and was transactivated by c-Rel or p65. Taken together, our results demonstrated that binding of c-Rel/p65 heterodimers to a novel kappa B-like site mediated LPS induction of TF gene expression in monocytic cells.


Subject(s)
Gene Expression/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides/metabolism , Proto-Oncogene Proteins/metabolism , Thromboplastin/biosynthesis , Animals , Base Sequence , Binding Sites , Cell Nucleus/metabolism , Consensus Sequence , Humans , Leukocytes, Mononuclear/drug effects , Macromolecular Substances , Mice , Molecular Sequence Data , NF-kappa B/isolation & purification , Nuclear Proteins/isolation & purification , Proto-Oncogene Proteins/isolation & purification , Proto-Oncogene Proteins c-rel , Swine , Transcription Factors , Transfection
15.
J Clin Invest ; 92(4): 1866-74, 1993 Oct.
Article in English | MEDLINE | ID: mdl-7691889

ABSTRACT

Oxidative stress and expression of the vascular cell adhesion molecule-1 (VCAM-1) on vascular endothelial cells are early features in the pathogenesis of atherosclerosis and other inflammatory diseases. Regulation of VCAM-1 gene expression may be coupled to oxidative stress through specific reduction-oxidation (redox) sensitive transcriptional or posttranscriptional regulatory factors. In cultured human umbilical vein endothelial (HUVE) cells, the cytokine interleukin 1 beta (IL-1 beta) activated VCAM-1 gene expression through a mechanism that was repressed approximately 90% by the antioxidants pyrrolidine dithiocarbamate (PDTC) and N-acetylcysteine (NAC). Furthermore, PDTC selectively inhibited the induction of VCAM-1, but not intercellular adhesion molecule-1 (ICAM-1), mRNA and protein accumulation by the cytokine tumor necrosis factor-alpha (TNF alpha) as well as the noncytokines bacterial endotoxin lipopolysaccharide (LPS) and double-stranded RNA, poly(I:C) (PIC). PDTC also markedly attenuated TNF alpha induction of VCAM-1-mediated cellular adhesion. In a distinct pattern, PDTC partially inhibited E-selectin gene expression in response to TNF alpha but not to LPS, IL-1 beta, or PIC. TNF alpha and LPS-mediated transcriptional activation of the human VCAM-1 promoter through NF-kappa B-like DNA enhancer elements and associated NF-kappa B-like DNA binding proteins was inhibited by PDTC. These studies suggest a molecular linkage between an antioxidant sensitive transcriptional regulatory mechanism and VCAM-1 gene expression that expands on the notion of oxidative stress as an important regulatory signal in the pathogenesis of atherosclerosis.


Subject(s)
Antioxidants/pharmacology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Endothelium, Vascular/metabolism , Gene Expression Regulation/physiology , Interleukin-1/pharmacology , Base Sequence , Binding Sites , Blotting, Northern , Cell Adhesion Molecules/analysis , Cell Nucleus/metabolism , Cells, Cultured , DNA Probes , E-Selectin , Endothelium, Vascular/drug effects , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/biosynthesis , Humans , Intercellular Adhesion Molecule-1 , Molecular Sequence Data , NF-kappa B/metabolism , Oligodeoxyribonucleotides/chemical synthesis , Oligodeoxyribonucleotides/metabolism , Promoter Regions, Genetic , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins , Vascular Cell Adhesion Molecule-1
16.
Mol Cell Biol ; 13(10): 6137-46, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8413215

ABSTRACT

Interleukin-8 (IL-8), a chemotactic cytokine for T lymphocytes and neutrophils, is induced in several cell types by a variety of stimuli including the inflammatory cytokines IL-1 and tumor necrosis factor alpha TNF-alpha. Several cis elements, including a binding site for the inducible transcription factor NF-kappa B, have been identified in the regulatory region of the IL-8 gene. We have examined the ability of various NF-kappa B subunits to bind to, and activate transcription from, the IL-8 promoter. A nuclear complex was induced in phorbol myristate acetate-treated Jurkat T cells which bound specifically to the kappa B site of the IL-8 promoter and was inhibited by addition of purified I kappa B alpha to the reaction mixture. Only antibody to RelA (p65), but not to NFKB1 (p50), NFKB2 (p50B), c-Rel, or RelB was able to abolish binding, suggesting that RelA is a major component in these kappa B binding complexes. Gel mobility shift analysis with in vitro-translated and purified proteins indicated that whereas the kappa B element in the human immunodeficiency virus type 1 long terminal repeat bound to all members of the kappa B/Rel family examined, the IL-8 kappa B site bound only to RelA and to c-Rel and NFKB2 homodimers, but not to NFKB1 homodimers or heterodimers of NFKB1-RelA. Transient transfection analysis demonstrated a kappa B-dependent expression of the IL-8 promoter in a human fibrosarcoma cell line (8387) and in Jurkat T lymphocytes. Cotransfection with various NF-kappa B subunits indicated that RelA and c-Rel, but neither NFKB1 nor heterodimeric NFKB1-RelA, was able to activate transcription from the IL-8 promoter. Furthermore, cotransfection of NFKB1 and RelA, although able to support activation from the human immunodeficiency virus type 1 long terminal repeat, failed to activate expression from the IL-8 promoter. Antisense oligonucleotides to RelA, but not NFKB1, inhibited phorbol myristate acetate-induced IL-8 production in Jurkat T lymphocytes. These data demonstrate the differential ability of members of the kappa B/Rel family to bind to, and activate transcription from, the IL-8 promoter. Furthermore, while providing a novel example of a kappa B-regulated promoter in which the classical NF-kappa B complex is unable to activate transcription from the kappa B element, these data provide direct evidence for the role of RelA in regulation of IL-8 gene expression.


Subject(s)
Gene Expression Regulation , Interleukin-8/genetics , NF-kappa B/physiology , Promoter Regions, Genetic , Base Sequence , Cell Line , DNA , Enzyme-Linked Immunosorbent Assay , HIV Long Terminal Repeat , Humans , Molecular Sequence Data , NF-kappa B/chemistry , NF-kappa B/genetics , Oligonucleotides, Antisense/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelA , Transcriptional Activation
17.
Mol Cell Biol ; 13(7): 3850-9, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8321192

ABSTRACT

The subunits of NF-kappa B, NFKB1 (formerly p50) and RelA (formerly p65), belong to a growing family of transcription factors that share extensive similarity to the c-rel proto-oncogene product. The homology extends over a highly conserved stretch of approximately 300 amino acids termed the Rel homology domain (RHD). This region has been shown to be involved in both multimerization (homo- and heterodimerization) and DNA binding. It is now generally accepted that homodimers of either subunit are capable of binding DNA that contains a kappa B site originally identified in the immunoglobulin enhancer. Recent studies have demonstrated that the individual subunits of the NF-kappa B transcription factor complex can be distinguished by their ability to bind distinct DNA sequence motifs. By using NFKB1 and RelA subunit fusion proteins, different regions within the RHD were found to confer DNA-binding and multimerization functions. A fusion protein that contains 34 N-terminal amino acids of NFKB1 and 264 amino acids of RelA displayed preferential binding to an NFKB1-selective DNA motif while dimerizing with the characteristics of RelA. Within the NFKB1 portion of this fusion protein, a single amino acid change of His to Arg altered the DNA-binding specificity to favor interaction with the RelA-selective DNA motif. Furthermore, substitution of four amino acids from NFKB1 into RelA was able to alter the DNA-binding specificity of the RelA protein to favor interaction with the NFKB1-selective site. Taken together, these findings demonstrate the presence of a distinct subdomain within the RHD involved in conferring the DNA-binding specificity of the Rel family of proteins.


Subject(s)
DNA/metabolism , NF-kappa B/metabolism , Amino Acid Sequence , Base Sequence , Cell Line , Cloning, Molecular , Humans , Molecular Sequence Data , Mutagenesis , NF-kappa B/chemistry , Proto-Oncogene Mas , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transcription Factor RelA
18.
Antisense Res Dev ; 3(4): 309-22, 1993.
Article in English | MEDLINE | ID: mdl-8155973

ABSTRACT

Antisense oligonucleotides have proved effective in achieving targeted inhibition of gene expression. In such experiments, sense oligonucleotides have frequently been used as a control for nonspecific effects, but the results have been variable, raising questions about the reliability of sense oligomers as a control. It is possible that some of the effects of sense oligonucleotides may be specific. We have shown that phosphorothioate antisense oligonucleotides to the p65 subunit of NF-kappa B, a transcription factor, cause a block in cell adhesion. In our efforts to test the efficacy of NF-kappa B p65 oligonucleotides in vivo, we unexpectedly observed that the control p65-sense, but not the p65-antisense, oligonucleotides caused massive splenomegaly in mice. In the current study we demonstrate a sequence-specific stimulation of splenic cell proliferation, both in vivo and in vitro, by treatment with p65-sense oligonucleotides. Cells expanded by this treatment are primarily B-220+, sIg+ B cells. The secretion of immunoglobulins by the p65-sense oligonucleotide-treated splenocytes is also enhanced. In addition, the p65-sense-treated splenocytes, but not several other cell lines, showed an upregulation of NF-kappa B-like activity in the nuclear extracts, an effect not dependent on new protein or RNA synthesis. These results demonstrate that phosphorothioate oligonucleotides can exert sequence-specific effects in vivo, irrespective of sense or antisense orientation.


Subject(s)
B-Lymphocytes/drug effects , Lymphocyte Activation/drug effects , NF-kappa B/genetics , Oligodeoxyribonucleotides/pharmacology , Spleen/drug effects , Thionucleotides/pharmacology , Animals , B-Lymphocytes/cytology , Base Sequence , Binding Sites , Cell Adhesion/drug effects , Cell Division/drug effects , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NF-kappa B/metabolism , Spleen/cytology , Splenomegaly/chemically induced , Up-Regulation
19.
Mol Cell Biol ; 12(10): 4412-21, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1406630

ABSTRACT

Analysis of the p50 and p65 subunits of the NF-kappa B transcription factor complex has revealed that both proteins can interact with related DNA sequences through either homo- or heterodimer formation. In addition, the product of the proto-oncogene c-rel can bind to similar DNA motifs by itself or as a heterodimer with p50 or p65. However, these studies have used a limited number of known kappa B DNA motifs, and the question of the optimal DNA sequences preferred by each homodimer has not been addressed. Using purified recombinant p50, p65, and c-Rel proteins, optimal DNA-binding motifs were selected from a pool of random oligonucleotides. Alignment of the selected sequences allowed us to predict a consensus sequence for binding of the individual homodimeric Rel-related proteins, and DNA-protein binding analysis of the selected DNA sequences revealed sequence specificity of the proteins. Contrary to previous assumptions, we observed that p65 homodimers can interact with a subset of DNA sequences not recognized by p50 homodimers. Differential binding affinities were also obtained with p50- and c-Rel-selected sequences. Using either a p50- or p65-selected kappa B motif, which displayed differential binding with respect to the other protein, little to no binding was observed with the heterodimeric NF-kappa B complex. Similarly, in transfection experiments in which the selective kappa B binding sites were used to drive the expression of a chloramphenicol acetyltransferase reporter construct, the p65- and p50-selected motifs were activated only in the presence of p65 and p50/65 (a chimeric protein with the p50 DNA binding domain and p65 activation domain) expression vectors, respectively, and neither demonstrated a significant response to stimuli that induce NF-kappa B activity. These findings demonstrate that interaction of both subunits of the heterodimeric NF-kappa B complex with DNA is required for DNA binding and transcriptional activation and suggest that transcriptional activation mediated by the individual rel-related proteins will differ dramatically, depending on the specific kappa B motifs present.


Subject(s)
DNA/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Base Sequence , Binding Sites , Cell Line , Consensus Sequence , Gene Expression Regulation , Humans , Molecular Sequence Data , NF-kappa B/chemistry , Proto-Oncogene Mas , Proto-Oncogene Proteins c-rel , T-Lymphocytes
20.
J Leukoc Biol ; 49(5): 505-10, 1991 May.
Article in English | MEDLINE | ID: mdl-2016571

ABSTRACT

Infection of the nervous system by human immunodeficiency virus type-1 (HIV-1) has been implicated in the generation of acquired immunodeficiency syndrome (AIDS)-associated neurologic dysfunction and direct infection of glia has been suggested as one of the potential mechanisms leading to deterioration of nervous system function. We have been examining the interaction of HIV-1 with the developing peripheral nervous system in vitro, and have previously shown that HIV-1 infection of primary human fetal dorsal root ganglia (DRG) neural cells resulted in HIV-1 gag antigen expression in approximately 70% of the glial cell subpopulation with little, if any, cytopathic damage to the infected cells. Accumulation of HIV-1 gag antigens and viral mRNA reached a maximum by 2-3 days postinfection and declined thereafter to minimally detectable levels in the surviving neural cell population. In addition, infection of the fetal DRG neural cells appeared to be abortive or nonproductive, with little if any, infectious progeny virus production. However, we have been able to detect HIV-1-specific proviral DNA as late as 24 days postinfection by polymerase chain reaction amplification and subsequent DNA blot hybridization. These results suggest that accumulation of HIV-1 structural proteins without the assembly and release of mature virus in HIV-1-infected human fetal DRG neural cells results in a nonproductive infection and maintenance of HIV-1 proviral DNA in the infected cell population.


Subject(s)
Cell Transformation, Viral , DNA, Viral/genetics , Ganglia, Spinal/cytology , HIV-1/physiology , Neurons/cytology , Proviruses/physiology , Cells, Cultured , DNA, Viral/analysis , Fetus , Fluorescent Antibody Technique , Gene Products, gag/analysis , HIV-1/genetics , Humans , Kinetics , Proviruses/genetics , RNA, Viral/genetics , RNA, Viral/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...