Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Interferon Cytokine Res ; 21(11): 931-41, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11747625

ABSTRACT

The epitopes important for receptor binding and activation of human interferon-beta1a (IFN-beta1a) were mapped with monoclonal antibodies (mAb), grouped on the basis of their specificity and ability to neutralize biologic activity, and alanine scanning mutagenesis (ASM). The binding properties of nine mAb were defined, using ASM-IFN-beta mutants having alanine substituted at targeted, surface-exposed residues. The results were correlated with the mAb neutralizing potency. Of six mAb that bound either at or adjacent to the IFNAR-2 receptor chain binding site defined by the ASM epitopes, only three had measurable neutralizing activity. Two of these inhibited IFN-beta/IFNAR-2 complex formation, suggesting that steric hindrance of receptor binding constitutes their mechanism of neutralization. However, two mAb that bound to sites remote from the IFNAR-2 binding site on IFN-beta also inhibited IFN-beta/IFNAR-2 complex formation and demonstrated potent neutralizing activity. Thus, neutralizing mAb may employ mechanisms other than steric blockade to inhibit directly the binding of receptor by cytokine, limiting their usefulness as tools to define precise receptor-ligand interaction sites.


Subject(s)
Alanine/genetics , Antibodies, Monoclonal/immunology , Epitopes/immunology , Interferon-beta/chemistry , Interferon-beta/immunology , Receptors, Interferon/metabolism , Amino Acid Sequence , Amino Acid Substitution , Antibody Specificity , Binding, Competitive , Dose-Response Relationship, Drug , Epitope Mapping , Humans , Interferon beta-1a , Interferon-beta/genetics , Interferon-beta/metabolism , Membrane Proteins , Models, Molecular , Molecular Sequence Data , Neutralization Tests , Receptor, Interferon alpha-beta , Sequence Alignment
2.
Biochim Biophys Acta ; 1518(1-2): 168-72, 2001 Mar 19.
Article in English | MEDLINE | ID: mdl-11267674

ABSTRACT

HIPK2 (homeodomain-interacting protein kinase 2) is a CD95 binding partner in yeast. Its primary amino acid sequence is highly conserved between human and mouse. The highest HIPK2 mRNA expression is found in neuronal tissue. The HIPK2 gene is located on human chromosome 7q33-35 and the protein is mainly localized in the nucleus. HIPK2 has been described to play a role as a co-repressor for homeodomain transcription factors.


Subject(s)
Carrier Proteins/genetics , Chromosomes, Human, Pair 7 , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , fas Receptor/metabolism , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/metabolism , Codon, Initiator , DNA, Complementary , Female , Gene Expression , Humans , Male , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger , Sequence Homology, Amino Acid , Tissue Distribution , Tumor Cells, Cultured
3.
J Biol Chem ; 276(22): 18819-27, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11279189

ABSTRACT

Mutations in the epithelial morphogen ectodysplasin-A (EDA), a member of the tumor necrosis factor (TNF) family, are responsible for the human disorder X-linked hypohidrotic ectodermal dysplasia (XLHED) characterized by impaired development of hair, eccrine sweat glands, and teeth. EDA-A1 and EDA-A2 are two splice variants of EDA, which bind distinct EDA-A1 and X-linked EDA-A2 receptors. We identified a series of novel EDA mutations in families with XLHED, allowing the identification of the following three functionally important regions in EDA: a C-terminal TNF homology domain, a collagen domain, and a furin protease recognition sequence. Mutations in the TNF homology domain impair binding of both splice variants to their receptors. Mutations in the collagen domain can inhibit multimerization of the TNF homology region, whereas those in the consensus furin recognition sequence prevent proteolytic cleavage of EDA. Finally, a mutation affecting an intron splice donor site is predicted to eliminate specifically the EDA-A1 but not the EDA-A2 splice variant. Thus a proteolytically processed, oligomeric form of EDA-A1 is required in vivo for proper morphogenesis.


Subject(s)
Ectodermal Dysplasia/genetics , Genetic Linkage , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutation , Tumor Necrosis Factor-alpha/chemistry , X Chromosome/genetics , Alternative Splicing , Amino Acid Sequence , Cell Line , Chromatography, Gel , Dimerization , Dose-Response Relationship, Drug , Ectodysplasins , Enzyme-Linked Immunosorbent Assay , Exons , Furin , Glycosylation , Humans , Introns , Ligands , Molecular Sequence Data , Phenotype , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Structure-Activity Relationship , Subtilisins/metabolism
4.
Biochemistry ; 39(10): 2538-51, 2000 Mar 14.
Article in English | MEDLINE | ID: mdl-10704203

ABSTRACT

A systematic mutational analysis of human interferon-beta-1a (IFN-beta) was performed to identify regions on the surface of the molecule that are important for receptor binding and for functional activity. The crystal structure of IFN-beta-1a was used to design a panel of 15 mutant proteins, in each of which a contiguous group of 2-8 surface residues was mutated, in most instances to alanine. The mutants were analyzed for activity in vitro in antiviral and in antiproliferation assays, and for their ability to bind to the type I IFN (ifnar1/ifnar2) receptor on Daudi cells and to a soluble ifnar2 fusion protein (ifnar2-Fc). Abolition of binding to ifnar2-Fc for mutants A2, AB1, AB2, and E established that the ifnar2 binding site on IFN-beta comprises parts of the A helix, the AB loop, and the E helix. Mutations in these areas, which together define a contiguous patch of the IFN-beta surface, also resulted in reduced affinity for binding to the receptor on cells and in reductions in activity of 5-50-fold in functional assays. A second receptor interaction site, concluded to be the ifnar1 binding site, was identified on the opposite face of the molecule. Mutations in this region, which encompasses parts of the B, C, and D helices and the DE loop, resulted in disparate effects on receptor binding and on functional activity. Analysis of antiproliferation activity as a function of the level of receptor occupancy allowed mutational effects on receptor activation to be distinguished from effects on receptor binding. The results suggest that the binding energy from interaction of IFN-beta with ifnar2 serves mainly to stabilize the bound IFN/receptor complex, whereas the binding energy generated by interaction of certain regions of IFN-beta with ifnar1 is not fully expressed in the observed affinity of binding but instead serves to selectively stabilize activated states of the receptor.


Subject(s)
Interferon-beta/genetics , Interferon-beta/metabolism , Receptors, Interferon/metabolism , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Antiviral Agents/metabolism , COS Cells , DNA Mutational Analysis , Genetic Vectors/metabolism , Growth Inhibitors/physiology , Humans , Interferon beta-1a , Interferon-beta/biosynthesis , Interferon-beta/physiology , Membrane Proteins , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Mapping , Protein Binding/genetics , Protein Structure, Tertiary , Receptor, Interferon alpha-beta , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Protein Sci ; 8(9): 1867-77, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10493588

ABSTRACT

The extracellular portions of the chains that comprise the human type I interferon receptor, IFNAR1 and IFNAR2, have been expressed and purified as recombinant soluble His-tagged proteins, and their interactions with each other and with human interferon-beta-1a (IFN-beta-1a) were studied by gel filtration and by cross-linking. By gel filtration, no stable binary complexes between IFN-beta-1a and IFNAR1, or between IFNAR1 and IFNAR2 were detected. However, a stable binary complex formed between IFN-beta-1a and IFNAR2. Analysis of binary complex formation using various molar excesses of IFN-beta-1a and IFNAR2 indicated that the complex had a 1:1 stoichiometry, and reducing SDS-PAGE of the binary complex treated with the cross-linking reagent dissucinimidyl glutarate (DSG) indicated that the major cross-linked species had an apparent Mr consistent with the sum of its two individual components. Gel filtration of a mixture of IFNAR1 and the IFN-beta-1a/IFNAR2 complex indicated that the three proteins formed a stable ternary complex. Analysis of ternary complex formation using various molar excesses of IFNAR1 and the IFN-beta-1a/IFNAR2 complex indicated that the ternary complex had a 1:1:1 stoichiometry, and reducing SDS-PAGE of the ternary complex treated with DSG indicated that the major cross-linked species had an apparent Mr consistent with the sum of its three individual components. We conclude that the ternary complex forms by the sequential association of IFN-beta-1a with IFNAR2, followed by the association of IFNAR1 with the preformed binary complex. The ability to produce the IFN-beta-1a/IFNAR2 and IFN-beta-1a/IFNAR1/IFNAR2 complexes make them attractive candidates for X-ray crystallography studies aimed at determining the molecular interactions between IFN-beta-1a and its receptor.


Subject(s)
Interferon-beta/chemistry , Interferon-beta/metabolism , Receptors, Interferon/chemistry , Receptors, Interferon/metabolism , Chromatography, Gel , Dimerization , Histidine/chemistry , Humans , Interferon beta-1a , Interferon-beta/isolation & purification , Macromolecular Substances , Membrane Proteins , Receptor, Interferon alpha-beta , Receptors, Interferon/isolation & purification , Solubility
6.
J Interferon Cytokine Res ; 19(1): 15-26, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10048764

ABSTRACT

The type I interferon receptor (IFNAR) is composed of two subunits, IFNAR-1 and IFNAR-2, encoding transmembrane polypeptides. IFNAR-2 has a dominant role in ligand binding, but IFNAR-1 contributes to binding affinity and to differential ligand recognition. A panel of five monoclonal antibodies (mAb) to human IFNAR-1 (HuIFNAR-1) was produced and characterized. The reactivity of each mAb toward HuIFNAR-1 on native and transfected cells and in Western blot and ELISA formats was determined. In functional assays, one mAb, EA12, blocked IFN-a2 binding to human cells and interfered with Stat activation and antiviral activity. Epitopes for the mAb were localized to subdomains of the HuIFNAR-1 extracellular domain by differential reactivity of the mAb to a series of human/bovine IFNAR-1 chimeras. The antibody EA12 seems to require native HuIFNAR-1 for reactivity and does not map to a single subdomain, perhaps recognizing an epitope containing noncontiguous sequences in at least two subdomains. In contrast, the epitopes of the non-neutralizing mAb FB2, AA3, and GB8 mapped, respectively, to the first, second, and third subdomains of HuIFNAR-1. The mAb DB2 primarily maps to the fourth subdomain, although its reactivity may be affected by other determinants.


Subject(s)
Antibodies, Monoclonal/analysis , Epitope Mapping , Animals , Antiviral Agents/metabolism , COS Cells , Humans , Interferon-alpha/metabolism , Protein Structure, Tertiary , Signal Transduction/physiology
7.
Cell Mol Life Sci ; 54(11): 1203-16, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9849615

ABSTRACT

Interferons (IFNs) are potent extracellular protein mediators of host defence and homoeostasis. This article reviews the structure of human IFN-beta (HuIFN-beta), in particular in relation to its activity. The recently determined crystal structure of HuIFN-beta provides a framework for understanding of the mechanism of differentiation of type I IFNs by their common receptor. Insights are generated by comparison with the structures of other type I IFNs and from the interpretation of existing mutagenesis data. The details of the observed carbohydrate structure, together with biochemical data, implicate the glycosylation of HuIFN-beta, which is uncommon among type I IFNs, as an important factor in the solubility, stability and, consequently, activity of the protein. Finally, these structural implications are discussed in the context of the clinical use of HuIFN-beta.


Subject(s)
Interferon-beta/chemistry , Crystallography, X-Ray , Dimerization , Glycosylation , Humans , Interferon-beta/therapeutic use , Models, Molecular , Protein Binding , Protein Structure, Secondary , Receptors, Interferon/chemistry
8.
J Biol Chem ; 273(14): 8003-8, 1998 Apr 03.
Article in English | MEDLINE | ID: mdl-9525899

ABSTRACT

Type I interferon (IFN) subtypes alpha and beta share a common multicomponent, cell surface receptor and elicit a similar range of biological responses, including antiviral, antiproliferative, and immunomodulatory activities. However, alpha and beta IFNs exhibit key differences in several biological properties. For example, IFN-beta, but not IFN-alpha, induces the association of tyrosine-phosphorylated receptor components ifnar1 and ifnar2, and has activity in cells lacking the IFN receptor-associated, Janus kinase tyk2. To define the structural basis for these functional differences we produced human IFN-beta with point mutations and compared them to wild-type IFN-beta in assays that distinguish alpha and beta IFN subtypes. IFN-beta mutants with charged residues (N86K, N86E, or Y92D) introduced at two positions in the C helix lost the ability to induce the association of tyrosine-phosphorylated receptor chains and had reduced activity on tyk2-deficient cells. The combination of negatively charged residues N86E and Y92D (homologous with IFN-alpha8) increased the cross-species activity of the mutant IFN-betas on bovine cells to a level comparable to that of human IFN-alphas. In contrast, point mutations in the AB loop and D helix had no significant effect on these subtype-specific activities. A subset of these latter mutations did, however, reduce activity in a manner analogous to IFN-alpha mutations. The effects of these mutations on IFN-beta activity are discussed in the context of a family of related ligands acting through a common receptor and signaling pathway.


Subject(s)
Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Amino Acid Sequence , Animals , Cattle , DNA Mutational Analysis , Humans , Interferon-alpha/chemistry , Interferon-beta/chemistry , Molecular Sequence Data , Point Mutation , Structure-Activity Relationship
9.
Pharm Res ; 15(4): 641-9, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9587963

ABSTRACT

PURPOSE: Two recombinant IFN-beta products have been approved for the treatment of multiple sclerosis, a glycosylated form with the predicted natural amino acid sequence (IFN-beta-1a) and a non-glycosylated form that has a Met-1 deletion and a Cys-17 to Ser mutation (IFN-beta-1b). The structural basis for activity differences between IFN-beta-1a and IFN-beta-1b, is determined. METHODS: In vitro antiviral, antiproliferative and immunomodulatory assays were used to directly compare the two IFN-beta products. Size exclusion chromatography (SEC), SDS-PAGE, thermal denaturation, and X-ray crystallography were used to examine structural differences. RESULTS: IFN-beta-1a was 10 times more active than IFN-beta-1b with specific activities in a standard antiviral assay of 20 x 10(7) IU/mg for IFN-beta-1a and 2 x 10(7) IU/mg for IFN-beta-1b. Of the known structural differences between IFN-beta-1a and IFN-beta-1b, only glycosylation affected in vitro activity. Deglycosylation of IFN-beta-1a produced a decrease in total activity that was primarily caused by the formation of an insoluble disulfide-linked IFN precipitate. Deglycosylation also resulted in an increased sensitivity to thermal denaturation. SEC data for IFN-beta-1b revealed large, soluble aggregates that had reduced antiviral activity (approximated at 0.7 x 10(7) IU/mg). Crystallographic data for IFN-beta-1a revealed that the glycan formed H-bonds with the peptide backbone and shielded an uncharged surface from solvent exposure. CONCLUSIONS: Together these results suggest that the greater biological activity of IFN-beta-1a is due to a stabilizing effect of the carbohydrate on structure.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antiviral Agents/pharmacology , Interferon-beta/pharmacology , Recombinant Proteins/pharmacology , Adjuvants, Immunologic/chemistry , Base Sequence , Cardiovirus Infections/drug therapy , Cell Division/drug effects , Cell Line , Cysteine/chemistry , Encephalomyocarditis virus/drug effects , Glycosylation , Humans , Interferon beta-1a , Interferon beta-1b , Interferon-beta/chemistry , Major Histocompatibility Complex/drug effects , Molecular Sequence Data , Mutation , Recombinant Proteins/chemistry , Serine/chemistry
10.
J Interferon Cytokine Res ; 17(6): 355-67, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9198003

ABSTRACT

Interferons (IFNs) generally have been characterized as antiproliferative cytokines. The cell cycle arrest in G1/G0 phase induced by type I IFNs, especially IFN-alpha, was recognized as a manifestation of their antiproliferative effects. In this article, we report that the cell cycle block in G1/G0 is observed mainly in certain cell types, such as Daudi Burkitt's lymphoma cells. In a variety of human transformed cells, but not nontransformed primary cells, IFN-beta and IFN-alpha induced a significant increase in the S phase population. The increase appeared to be due to a continued S phase entry and subsequently a failure of S phase cells to transit efficiently into G2 and M phases. The ability of tumor cells to exhibit the S phase effect correlated with proper IFN signaling and loss or inactivation of the normal G1 checkpoint conferred by the retinoblastoma protein (pRB). Overriding the G1 checkpoint switched human nontransformed primary cells from nonresponsive to sensitive to the IFN-induced effect. Therefore, the cell cycle regulatory machinery could function, at least in part, as a determining factor that affects the IFN-induced cell cycle effect. The IFN effect in transformed cells may suggest intriguing prospects for combinatorial therapies for cancer.


Subject(s)
Interferon-beta/pharmacology , S Phase/drug effects , Cell Cycle/drug effects , Cell Line, Transformed/cytology , Cell Line, Transformed/drug effects , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/physiology , G1 Phase/drug effects , Humans , Retinoblastoma Protein/drug effects , Retinoblastoma Protein/physiology , Signal Transduction/drug effects , Trans-Activators/drug effects , Trans-Activators/physiology , Tumor Cells, Cultured
11.
J Exp Med ; 182(5): 1223-30, 1995 Nov 01.
Article in English | MEDLINE | ID: mdl-7595193

ABSTRACT

Apoptosis occurs in the normal liver and in various forms of liver disease. The CD95 (APO-1/Fas) (CD95) receptor mediates apoptosis, and liver cells in animal models are acutely sensitive to apoptosis initiated by this receptor. We have used primary human hepatocytes as a model system to investigate CD95-mediated apoptotic liver damage. Treatment of fresh human hepatocytes with low concentrations of agonistic antibodies against CD95 resulted in apoptosis of > 95% of the cultured liver cells within 4 and 7.5 h. Immunohistology of a panel of explanted liver tissues revealed that hepatocytes in normal livers (n = 5) and in alcoholic cirrhosis (n = 13) expressed low constitutive levels of CD95. CD95 receptor expression was highly elevated in hepatocytes in hepatitis B virus-related cirrhosis (n = 9) and in acute liver failure (n = 8). By in situ hybridization CD95 ligand messenger RNA expression was absent in normal liver but detected at high levels in livers with ongoing liver damage. In cases of hepatitis B virus-related cirrhosis and acute hepatic failure, ligand expression was found primarily in areas with lymphocytic infiltration. In contrast, in patients with alcoholic liver damage, high CD95 ligand messenger RNA expression was found in hepatocytes. These findings suggest that liver destruction in hepatitis B may primarily involve killing of hepatocytes by T lymphocytes using the CD95 receptor-ligand system. In alcoholic liver damage, death of hepatocytes might occur by fratricide and paracrine or autocrine mechanisms mediated by the hepatocytes themselves.


Subject(s)
Apoptosis/physiology , Liver/pathology , Membrane Glycoproteins/physiology , T-Lymphocytes, Cytotoxic/immunology , fas Receptor/physiology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Fas Ligand Protein , Hepatitis, Viral, Human/complications , Hepatitis, Viral, Human/pathology , Humans , In Situ Hybridization, Fluorescence , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Liver Cirrhosis, Alcoholic/pathology , Liver Failure/pathology , Lymphocyte Activation , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , RNA, Messenger/analysis , fas Receptor/immunology
12.
Virus Genes ; 10(1): 99-102, 1995.
Article in English | MEDLINE | ID: mdl-7483296

ABSTRACT

Two-hybrid protein interaction screening in yeast was used to identify proteins that interact with the HBx nonstructural protein of hepatitis B virus (HBV). A new human member of the proteasome alpha-subunit family was obtained. Its protein sequence closely resembles the 28 kD subunits from other organisms. The interaction with HBx was abolished by a two amino-acid insertion behind position 128 in HBx, in a region previously found to be essential for its transcriptional transactivation function. These data support a model of HBx acting indirectly on transcriptional processes. By binding to a specific proteasome alpha-subunit, HBx might interfere with degradative processes, thereby enhancing the half-life of different transcription factors and other nuclear regulatory proteins. Interaction with the Hu 28k proteasome subunit could thus provide a unifying explanation for the markedly pleiotropic effects of HBx.


Subject(s)
Cysteine Endopeptidases/metabolism , Hepatitis B Antigens/metabolism , Multienzyme Complexes/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , Animals , HeLa Cells , Humans , Molecular Sequence Data , Proteasome Endopeptidase Complex , Protein Binding , Recombinant Fusion Proteins/metabolism , Viral Regulatory and Accessory Proteins
13.
Proc Natl Acad Sci U S A ; 91(9): 3764-8, 1994 Apr 26.
Article in English | MEDLINE | ID: mdl-8170985

ABSTRACT

During hepatitis B virus (HBV) infection, distinct host-virus interactions may establish the patterns of viral clearance and persistence and the extent of virus-associated pathology. It is generally thought that HBV-specific class I-restricted cytotoxic T lymphocytes (CTLs) play a critical role in this process by destroying infected hepatocytes. This cytopathic mechanism, however, could be lethal if most of the hepatocytes are infected. In the current study, we demonstrate that class I-restricted HBV-specific CTLs profoundly suppress hepatocellular HBV gene expression in HBV transgenic mice by a noncytolytic process, the strength of which greatly exceeds the cytopathic effect of the CTLs in magnitude and duration. We also show that the regulatory effect of the CTLs is initially mediated by interferon gamma and tumor necrosis factor alpha, is delayed in onset, and becomes independent of these cytokines shortly after it begins. The data indicate that the anti-viral CTL response activates a complex regulatory cascade that inhibits hepatocellular HBV gene expression without killing the cell. The extent to which this mechanism contributes to viral clearance or viral persistence during HBV infection remains to be determined.


Subject(s)
Hepatitis B Surface Antigens/immunology , Hepatitis B virus/genetics , Liver Diseases/immunology , T-Lymphocytes, Cytotoxic/microbiology , Animals , Cytokines/genetics , Cytotoxicity, Immunologic , Gene Expression Regulation, Viral , Interferon-gamma/physiology , Liver Diseases/microbiology , Mice , Mice, Transgenic , RNA, Messenger/genetics , RNA, Viral/genetics , Tumor Necrosis Factor-alpha/physiology
14.
Virology ; 197(2): 529-36, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8249276

ABSTRACT

A panel of mutants of the hepatitis B virus X gene (HBx) was constructed by oligonucleotide-directed insertion of two codons (Arg Pro) at 10- or 20-amino-acid intervals along the entire gene. These mutants were tested in transiently transfected HepG2 cells for their effects on HBx trans-activation of an AP1-CAT reporter plasmid. The effects of HBx mutations on the mRNA and protein stability were also determined. Our results reveal two separate internal domains of the HBx, one around amino acid residue 68 and the other between residues 110 and 139, that are necessary for its activity. Mutations in the amino terminus (to position 45), between the two necessary domains and in the extreme carboxyl terminal portion of HBx, have little or no effect on its activity.


Subject(s)
Gene Expression Regulation, Viral , Hepatitis B Antigens/genetics , Mutation , Trans-Activators/genetics , Transcriptional Activation , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Chloramphenicol O-Acetyltransferase/biosynthesis , Codon , DNA Mutational Analysis , Genes, Reporter , Humans , Molecular Sequence Data , Mutagenesis, Insertional , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/biosynthesis , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Sequence Homology, Amino Acid , Viral Regulatory and Accessory Proteins
15.
Mol Cell Biol ; 11(3): 1270-80, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1899906

ABSTRACT

Three cis-acting domains that contribute to the basal promoter activity of the human c-fos gene were identified. One encompasses the serum response element and has been previously described. Another spans an NF1-like site situated at -170. Mutations and in vitro protein binding assays pinpoint this site as the sole basal element of the medial domain. The third, or promoter-proximal, domain can be divided into several distinct sites, one containing a directly repeated GC-rich element and the other consisting of partially overlapping recognition sites for transcription factors ATF/CREB and MLTF/USF. Each of these sites contributes to basal activity as assayed by transient transfections and by in vitro transcription. Consistent with this, several complexes could be visualized between this region and nuclear proteins in vitro and genomic footprinting demonstrated that both elements are constitutively bound in vivo. On the basis of these results, we conclude that all three domains are necessary for full c-fos promoter function.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Base Sequence , Binding Sites , Cloning, Molecular , DNA Mutational Analysis , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-fos , Transcription Factors/metabolism , Tumor Cells, Cultured
16.
Nucleic Acids Res ; 14(18): 7143-58, 1986 Sep 25.
Article in English | MEDLINE | ID: mdl-3763402

ABSTRACT

A sequence of 86 bp within the 5' region of the adult chicken beta-globin gene was found to undergo a DNA conformational transition at elevated levels of negative superhelical stress (- sigma = 0.068). In vitro chemical DNA modification studies which detect purine hyperreactivity (HR) to the alkylating agent diethyl pyrocarbonate (DEP) have identified this 86 bp long DEP-HR element. The DEP-HR element is composed of small, tandem segments with imperfect purine-pyrimidine alternations. Methylation of cytosines within GCGC sequences of the DEP-HR element facilitates this structural change. The binding of a monoclonal anti-Z-DNA antibody to the element has been revealed by chemical footprinting with DEP. These data suggest that the DEP-HR sequence can undergo a conformational transition to Z-DNA. It is unknown whether the conformational flexibility observed here occurs in vivo.


Subject(s)
DNA/genetics , Genes , Globins/genetics , Animals , Antibodies , Base Sequence , Chickens , Chromosome Deletion , DNA/blood , Erythrocytes/metabolism , Nucleic Acid Conformation , Plasmids
17.
J Mol Biol ; 189(3): 487-501, 1986 Jun 05.
Article in English | MEDLINE | ID: mdl-2431152

ABSTRACT

Diethylpyrocarbonate (DEPC) carbethoxylates Z-DNA to an increased extent because the reactive N-7 atoms of purine residues appear structurally more accessible on Z-DNA as opposed to B-DNA. This chemical probe was used in DEPC footprinting experiments, which confirm the specificity of binding of anti-Z-DNA monoclonal antibodies and which probe regions of close contact in this DNA-protein complex. Antibody binding to segments of Z-DNA existing in supercoiled plasmids resulted in specific protection from DEPC hyper-reactivity within the Z-DNA segment and induction of hyper-reactivity in purines lying adjacent to the Z-segment. Two different monoclonal immunoglobulin preparations, Z22 and Z44, are shown to generate specific and distinct footprint patterns when bound to the Z-helix. Binding of these antibodies was also found to affect DNA conformation within the Z-DNA segment by influencing the equilibrium between the B- and Z-helical conformations.


Subject(s)
Antibodies, Antinuclear/immunology , DNA/immunology , Diethyl Pyrocarbonate/pharmacology , Formates/pharmacology , Antibodies, Monoclonal/immunology , Antibody Specificity , Binding Sites, Antibody , Computers , DNA/drug effects , DNA, Superhelical , Epitopes , Nucleic Acid Conformation
18.
J Bacteriol ; 154(3): 1508-12, 1983 Jun.
Article in English | MEDLINE | ID: mdl-6343359

ABSTRACT

We determined the DNA sequence of the D-serine deaminase promoter region and of the N-terminal region of the structural gene. There are possibilities in the promoter for secondary structure and for initiation recognition sequences, and there is an open reading frame. The N-terminal sequence for the structural gene confirms that part of the amino acid sequence previously determined by E. Schlitz and W. Schmitt (FEBS Lett. 134:57-62, 1981), including the active site of the enzyme, and spans the two regions unresolved by their work.


Subject(s)
Escherichia coli/genetics , Genes , L-Serine Dehydratase/genetics , Operon , Amino Acid Sequence , Base Sequence , DNA, Bacterial , Escherichia coli/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...