14 results on '"Ludes-Meyers JH"'
Search Results
2. Discovery of Sisunatovir (RV521), an Inhibitor of Respiratory Syncytial Virus Fusion.
- Author
-
Cockerill GS, Angell RM, Bedernjak A, Chuckowree I, Fraser I, Gascon-Simorte J, Gilman MSA, Good JAD, Harland R, Johnson SM, Ludes-Meyers JH, Littler E, Lumley J, Lunn G, Mathews N, McLellan JS, Paradowski M, Peeples ME, Scott C, Tait D, Taylor G, Thom M, Thomas E, Villalonga Barber C, Ward SE, Watterson D, Williams G, Young P, and Powell K
- Subjects
- Animals, Antiviral Agents chemical synthesis, Antiviral Agents pharmacokinetics, Benzimidazoles chemical synthesis, Benzimidazoles pharmacokinetics, Biological Availability, Cell Line, Tumor, Clinical Trials as Topic, Drug Discovery, Humans, Microbial Sensitivity Tests, Protein Binding, Viral Fusion Proteins metabolism, Antiviral Agents pharmacology, Benzimidazoles pharmacology, Respiratory Syncytial Virus, Human drug effects, Virus Internalization drug effects
- Abstract
RV521 is an orally bioavailable inhibitor of respiratory syncytial virus (RSV) fusion that was identified after a lead optimization process based upon hits that originated from a physical property directed hit profiling exercise at Reviral. This exercise encompassed collaborations with a number of contract organizations with collaborative medicinal chemistry and virology during the optimization phase in addition to those utilized as the compound proceeded through preclinical and clinical evaluation. RV521 exhibited a mean IC
50 of 1.2 nM against a panel of RSV A and B laboratory strains and clinical isolates with antiviral efficacy in the Balb/C mouse model of RSV infection. Oral bioavailability in preclinical species ranged from 42 to >100% with evidence of highly efficient penetration into lung tissue. In healthy adult human volunteers experimentally infected with RSV, a potent antiviral effect was observed with a significant reduction in viral load and symptoms compared to placebo.- Published
- 2021
- Full Text
- View/download PDF
3. Generation and characterization of mice carrying a conditional allele of the Wwox tumor suppressor gene.
- Author
-
Ludes-Meyers JH, Kil H, Parker-Thornburg J, Kusewitt DF, Bedford MT, and Aldaz CM
- Subjects
- Alleles, Animals, Bone and Bones metabolism, Cell Transformation, Neoplastic, Female, Gene Deletion, Genotype, Humans, Integrases metabolism, Kidney Tubules metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, WW Domain-Containing Oxidoreductase, Gene Expression Regulation, Oxidoreductases genetics
- Abstract
WWOX, the gene that spans the second most common human chromosomal fragile site, FRA16D, is inactivated in multiple human cancers and behaves as a suppressor of tumor growth. Since we are interested in understanding WWOX function in both normal and cancer tissues we generated mice harboring a conditional Wwox allele by flanking Exon 1 of the Wwox gene with LoxP sites. Wwox knockout (KO) mice were developed by breeding with transgenic mice carrying the Cre-recombinase gene under the control of the adenovirus EIIA promoter. We found that Wwox KO mice suffered from severe metabolic defect(s) resulting in growth retardation and all mice died by 3 wk of age. All Wwox KO mice displayed significant hypocapnia suggesting a state of metabolic acidosis. This finding and the known high expression of Wwox in kidney tubules suggest a role for Wwox in acid/base balance. Importantly, Wwox KO mice displayed histopathological and hematological signs of impaired hematopoiesis, leukopenia, and splenic atrophy. Impaired hematopoiesis can also be a contributing factor to metabolic acidosis and death. Hypoglycemia and hypocalcemia was also observed affecting the KO mice. In addition, bone metabolic defects were evident in Wwox KO mice. Bones were smaller and thinner having reduced bone volume as a consequence of a defect in mineralization. No evidence of spontaneous neoplasia was observed in Wwox KO mice. We have generated a new mouse model to inactivate the Wwox tumor suppressor gene conditionally. This will greatly facilitate the functional analysis of Wwox in adult mice and will allow investigating neoplastic transformation in specific target tissues.
- Published
- 2009
- Full Text
- View/download PDF
4. WWOX hypomorphic mice display a higher incidence of B-cell lymphomas and develop testicular atrophy.
- Author
-
Ludes-Meyers JH, Kil H, Nuñez MI, Conti CJ, Parker-Thornburg J, Bedford MT, and Aldaz CM
- Subjects
- Animals, Atrophy, Cell Line, Tumor, Embryo, Mammalian metabolism, Female, Gene Expression Regulation, Neoplastic, Male, Mice, Mice, Knockout, Mice, Transgenic, Models, Animal, WW Domain-Containing Oxidoreductase, Lymphoma, B-Cell genetics, Oxidoreductases genetics, Testis pathology
- Abstract
WWOX is a putative tumor suppressor gene encoded within common chromosomal fragile site region FRA16D, in chromosome band 16q23. Multiple studies have demonstrated that WWOX expression is often reduced or lost in various tumor types. WWOX tumor suppressor activity was suggested by re-expressing WWOX in breast, ovarian, and lung tumor cell lines leading to tumor growth inhibition in vivo. To determine whether loss of Wwox gene expression has a role in tumorigenesis, we generated a mouse strain containing a Wwox gene mutated by a gene-trap vector. Homozygous Wwox gene-trap mice (Wwox(gt/gt)) had no detectable Wwox protein in most tissues examined, although, a low level could be detected in a minority of tissues. Because of these observations, we concluded that these mice are Wwox hypomorphs. Remarkably, Wwox hypomorphic mice are viable in contrast to the recently reported postnatal lethality of Wwox knockout mice. Testes from Wwox(gt/gt) males had high numbers of atrophic seminiferous tubules and reduced fertility when compared with wild-type counterparts. We observed that the Wwox(gt/gt) mice had a significantly shorter lifespan, and female hypomorphs had a higher incidence of spontaneous B-cell lymphomas. In conclusion, we describe a novel Wwox hypomorphic mouse model that overcomes postnatal lethality that was recently observed in Wwox knockout mice. Therefore, tumorigenesis studies using this model more closely recapitulates the loss of WWOX expression observed in human cancers. Importantly, our observation that Wwox hypomorphs had an increased incidence of B-cell lymphomas supports a role of Wwox as a tumor suppressor., ((c) 2007 Wiley-Liss, Inc.)
- Published
- 2007
- Full Text
- View/download PDF
5. Expression of common chromosomal fragile site genes, WWOX/FRA16D and FHIT/FRA3B is downregulated by exposure to environmental carcinogens, UV, and BPDE but not by IR.
- Author
-
Thavathiru E, Ludes-Meyers JH, MacLeod MC, and Aldaz CM
- Subjects
- Acid Anhydride Hydrolases genetics, Cell Line, Tumor, Down-Regulation radiation effects, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic radiation effects, Humans, Neoplasm Proteins genetics, Oxidoreductases genetics, Radiation, Ionizing, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Proteins, WW Domain-Containing Oxidoreductase, 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide pharmacology, Acid Anhydride Hydrolases metabolism, Carcinogens, Environmental pharmacology, Chromosome Fragile Sites genetics, Down-Regulation drug effects, Neoplasm Proteins metabolism, Oxidoreductases metabolism, Ultraviolet Rays
- Abstract
Common chromosomal fragile sites are unstable genomic loci susceptible to breakage, rearrangement, and are highly recombinogenic. Frequent alterations at these loci in tumor cells led to the hypothesis that they may contribute to cancer development. The two most common chromosomal fragile sites FRA16D and FRA3B which harbor WWOX and FHIT genes, respectively, are frequently altered in human cancers. Here we report that environmental carcinogens, ultraviolet (UV) light, and Benzo[a]pyrene diol epoxide (BPDE), significantly downregulate expression of both genes. On the other hand, we observe that ionizing radiation (IR) does not affect expression of these genes, suggesting that the effect of repression exerted by UV and BPDE is not just a consequence of DNA damage but may be a result of different signaling pathways triggered by specific DNA lesions. Such downregulation correlates with an induction of an S-phase delay in the cell cycle. Treatment of UV-irradiated cells with caffeine abrogates the S-phase delay while concomitantly overcoming the repression phenomenon. This suggests the involvement of unique cell cycle checkpoint mechanisms in the observed repression. Therefore, it is hypothesized that protracted downregulation of the putative tumor suppressor genes WWOX and FHIT by environmental carcinogens may constitute an additional mechanism of relevance in the initiation of tumorigenesis., (Copyright 2005 Wiley-Liss, Inc)
- Published
- 2005
- Full Text
- View/download PDF
6. WWOX protein expression varies among ovarian carcinoma histotypes and correlates with less favorable outcome.
- Author
-
Nunez MI, Rosen DG, Ludes-Meyers JH, Abba MC, Kil H, Page R, Klein-Szanto AJ, Godwin AK, Liu J, Mills GB, and Aldaz CM
- Subjects
- Blotting, Western, Female, Gene Deletion, Genes, Tumor Suppressor, Homozygote, Humans, Immunoblotting, Immunohistochemistry, Loss of Heterozygosity, Oligonucleotide Array Sequence Analysis, Receptors, Progesterone biosynthesis, Time Factors, Treatment Outcome, Tumor Suppressor Proteins, WW Domain-Containing Oxidoreductase, Gene Expression Regulation, Neoplastic, Ovarian Neoplasms metabolism, Ovary metabolism, Oxidoreductases biosynthesis
- Abstract
Background: The putative tumor suppressor WWOX gene spans the common chromosomal fragile site 16D (FRA16D) at chromosome area 16q23.3-24.1. This region is a frequent target for loss of heterozygosity and chromosomal rearrangement in ovarian, breast, hepatocellular, prostate carcinomas and other neoplasias. The goal of these studies was to evaluate WWOX protein expression levels in ovarian carcinomas to determine if they correlated with clinico-pathological parameters, thus providing additional support for WWOX functioning as a tumor suppressor., Methods: We performed WWOX protein expression analyses by means of immunobloting and immunohistochemistry on normal ovaries and specific human ovarian carcinoma Tissue Microarrays (n = 444). Univariate analysis of clinical-pathological parameters based on WWOX staining was determined by chi2 test with Yates' correction. The basic significance level was fixed at p < 0.05., Results: Immunoblotting analysis from normal ovarian samples demonstrated consistently strong WWOX expression while 37% ovarian carcinomas showed reduced or undetectable WWOX protein expression levels. The immunohistochemistry of normal human ovarian tissue sections confirmed strong WWOX expression in ovarian surface epithelial cells and in epithelial inclusion cysts within the cortex. Out of 444 ovarian carcinoma samples analyzed 30% of tumors showed lack of or barely detectable WWOX expression. The remaining ovarian carcinomas (70%) stained moderately to strongly positive for this protein. The two histotypes showing significant loss of WWOX expression were of the Mucinous (70%) and Clear Cell (42%) types. Reduced WWOX expression demonstrated a significant association with clinical Stage IV (FIGO) (p = 0.007), negative Progesterone Receptor (PR) status (p = 0.008) and shorter overall survival (p = 0.03)., Conclusion: These data indicate that WWOX protein expression is highly variable among ovarian carcinoma histotypes. It was also observed that subsets of ovarian tumors demonstrated loss of WWOX expression and is potentially associated with patient outcome.
- Published
- 2005
- Full Text
- View/download PDF
7. WWOX binds the specific proline-rich ligand PPXY: identification of candidate interacting proteins.
- Author
-
Ludes-Meyers JH, Kil H, Bednarek AK, Drake J, Bedford MT, and Aldaz CM
- Subjects
- Amino Acid Sequence, Binding Sites, Golgi Apparatus metabolism, Humans, Ligands, Molecular Sequence Data, Nuclear Proteins metabolism, Proline analogs & derivatives, Protein Binding, Protein Structure, Tertiary, Transcription Factors metabolism, Tumor Cells, Cultured, Tumor Suppressor Proteins, WW Domain-Containing Oxidoreductase, Oxidoreductases metabolism, Proline metabolism
- Abstract
WWOX, the gene that maps to common chromosomal fragile site FRA16D, is frequently affected by aberrations in multiple types of cancers. WWOX encodes a 46 kDa protein that contains two WW domains and a short-chain oxidoreductase (SDR) domain. We recently demonstrated that ectopic expression of WWOX inhibits xenograft tumor growth of tumorigenic breast cancer cells. Little is known of the biochemical function(s) of WWOX. The SDR domain is predicted to be involved in sex-steroid metabolism and the WW domains are likely involved in protein-protein interactions. In this report, we identify the specific proline-rich ligand for WWOX as PPXY and show that the amino-terminal WW domain is responsible for this interaction. Using the WWOX WW domains as a probe, we screened high-density protein arrays and identified five candidate-binding partners. The binding to one of these candidates, small membrane protein of the lysosome/late endosome (SIMPLE), was further analysed, and we observed that a specific PPSY motif in the SIMPLE amino-acid sequence was required to interact with the amino-terminal WW domain of WWOX. In addition, immunofluorescence staining demonstrated that endogenous WWOX and SIMPLE co-localize to perinuclear compartments of MCF-7 human breast cancer cells. These studies demonstrate that WWOX contains a Group I WW domain that binds known cellular proteins containing the specific ligand PPXY. Identification and characterization of WWOX interacting proteins will lead to an understanding of the biological functions of WWOX in normal and tumor cells.
- Published
- 2004
- Full Text
- View/download PDF
8. WWOX, the common chromosomal fragile site, FRA16D, cancer gene.
- Author
-
Ludes-Meyers JH, Bednarek AK, Popescu NC, Bedford M, and Aldaz CM
- Subjects
- Alternative Splicing, Amino Acid Sequence, Blotting, Western, Breast Neoplasms pathology, Carrier Proteins metabolism, Cell Line, Tumor, Chromosome Mapping, Cloning, Molecular, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Golgi Apparatus metabolism, Humans, In Situ Hybridization, Fluorescence, Loss of Heterozygosity, Molecular Sequence Data, Neoplasm Proteins metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Review Literature as Topic, Breast Neoplasms genetics, Carrier Proteins genetics, Chromosome Fragile Sites genetics, Chromosomes, Human, Pair 16 genetics, Neoplasm Proteins genetics
- Abstract
Gross chromosomal rearrangements and aneuploidy are among the most common somatic genomic abnormalities that occur during cancer initiation and progression, in particular in human solid tumor carcinogenesis. The loss of large chromosomal regions as consequence of gross rearrangements (e.g. deletions, monosomies, unbalanced translocations and mitotic recombination) have been traditionally associated with the existence of tumor suppressor genes within the areas affected by the loss of genetic material. The long arm of chromosome 16 was identified as being frequently associated with structural abnormalities in multiple neoplasias, that led us to focus attention on the detailed genetic dissection of this region resulting in the cloning of the putative tumor suppressor gene, WWOX (WW domain containing Oxidoreductase). Interestingly, the WWOX gene resides in the very same region as that of the common chromosomal fragile site 16D (FRA16D). The WWOX gene encodes a protein that contains two WW domains, involved in protein-protein interactions, and a short chain dehydrogenase (SDR) domain, possibly involved in sex-steroid metabolism. We have identified the WWOX WW domain ligand as the PPXY motif confirming the biochemical activity of this domain. WWOX normally resides in the Golgi and we will demonstrate that Golgi localization requires an intact SDR. Inactivation of the WWOX gene during tumorigenesis can occur by homozygous deletions and possibly mutation, however, aberrantly spliced forms of WWOX mRNA have been observed even when one allele is still intact. The aberrantly spliced mRNAs have deletions of the exons that encode the SDR and these WWOX protein isoforms display abnormal intracellular localization to the nucleus possibly functioning as dominant negative inhibitors of full length WWOX. Thus, generation of aberrant transcripts of WWOX may represent a novel mechanism to functionally inactivate WWOX without genomic alteration of the remaining allele. In this article we will review the cloning and identification of WWOX as the target of FRA16D. In addition, we will discuss the possible biochemical functions of WWOX and present evidence that ectopic WWOX expression inhibits tumor growth., (Copyright 2003 S. Karger AG, Basel)
- Published
- 2003
- Full Text
- View/download PDF
9. Forkhead homologue in rhabdomyosarcoma functions as a bifunctional nuclear receptor-interacting protein with both coactivator and corepressor functions.
- Author
-
Zhao HH, Herrera RE, Coronado-Heinsohn E, Yang MC, Ludes-Meyers JH, Seybold-Tilson KJ, Nawaz Z, Yee D, Barr FG, Diab SG, Brown PH, Fuqua SA, and Osborne CK
- Subjects
- Amino Acid Sequence, Animals, Blotting, Western, Breast Neoplasms metabolism, COS Cells, DNA, Complementary metabolism, Forkhead Box Protein O1, Forkhead Transcription Factors, Gene Library, Glutathione Transferase metabolism, Humans, Ligands, Luciferases metabolism, Mice, Mice, Nude, Molecular Sequence Data, Plasmids metabolism, Protein Binding, Protein Structure, Tertiary, Receptors, Estrogen metabolism, Recombinant Fusion Proteins metabolism, Sequence Homology, Amino Acid, Signal Transduction, Tissue Distribution, Transcriptional Activation, Transfection, Tumor Cells, Cultured, Two-Hybrid System Techniques, Cell Nucleus metabolism, DNA-Binding Proteins chemistry, Rhabdomyosarcoma metabolism, Transcription Factors chemistry
- Abstract
In a search for novel transcriptional intermediary factors for the estrogen receptor (ER), we used the ligand-binding domain and hinge region of ER as bait in a yeast two-hybrid screen of a cDNA library derived from tamoxifen-resistant MCF-7 human breast tumors from an in vivo athymic nude mouse model. Here we report the isolation and characterization of the forkhead homologue in rhabdomyosarcoma (FKHR), a recently described member of the hepatocyte nuclear factor 3/forkhead homeotic gene family, as a nuclear hormone receptor (NR) intermediary protein. FKHR interacts with both steroid and nonsteroid NRs, although the effect of ligand on this interaction varies by receptor type. The interaction of FKHR with ER is enhanced by estrogen, whereas its interaction with thyroid hormone receptor and retinoic acid receptor is ligand-independent. In addition, FKHR differentially regulates the transactivation mediated by different NRs. Transient transfection of FKHR into mammalian cells dramatically represses transcription mediated by the ER, glucocorticoid receptor, and progesterone receptor. In contrast, FKHR stimulates rather than represses retinoic acid receptor- and thyroid hormone receptor-mediated transactivation. Most intriguingly, overexpression of FKHR dramatically inhibits the proliferation of ER-dependent MCF-7 breast cancer cells. Therefore, FKHR represents a bifunctional NR intermediary protein that can act as either a coactivator or corepressor, depending on the receptor type.
- Published
- 2001
- Full Text
- View/download PDF
10. AP-1 blockade inhibits the growth of normal and malignant breast cells.
- Author
-
Ludes-Meyers JH, Liu Y, Muñoz-Medellin D, Hilsenbeck SG, and Brown PH
- Subjects
- Breast metabolism, Breast Neoplasms metabolism, Cell Division physiology, Female, Genetic Vectors metabolism, Humans, Proto-Oncogene Proteins c-jun genetics, Proto-Oncogene Proteins c-jun metabolism, Transcription Factor AP-1 metabolism, Tumor Cells, Cultured drug effects, Tumor Stem Cell Assay, Breast cytology, Breast Neoplasms pathology, Cell Division drug effects, Transcription Factor AP-1 antagonists & inhibitors
- Abstract
We have previously demonstrated that basal AP-1 transcriptional activity is high in normal human mammary epithelial cells, intermediate in immortal breast cells, and relatively low in breast cancer cells. In this study we investigated whether differences in AP-1 transcriptional activity reflect differences in breast cells' dependence on AP-1 for proliferation. The cJun dominant negative, TAM-67, was used to determine the effect of AP-1 blockade on the growth of normal, immortal and malignant breast cells. We first showed that TAM-67 inhibits AP-1 activity in normal and malignant breast cells. We then determined whether this AP-1 inhibitor affected colony forming efficiency of the immortalized and malignant breast cells. The AP-1 inhibitor reduced colony formation of immortal breast cells by over 50% (by 58% in 184B5 cells and 62% in MCF10A cells), and reduced colony formation in the breast cancer cell line MCF7 by 43%, but did not reduce colony formation in the other breast cancer cell lines (T47D, MDA MB231 and MDA MB 435). We also determined the effect of AP-1 blockade on the growth of normal breast cells using a single cell proliferation assay. Using this assay, the growth of normal breast cells was extremely sensitive to AP-1 blockade, while immortal breast cells were moderately sensitive. We next directly tested the effect of TAM-67 expression on the growth of MCF7 breast cancer cells, using cells stably transfected with TAM-67 under the control of a doxycycline-inducible promoter. Upon induction, TAM-67 was expressed and AP-1 activity was inhibited in these cells. We then measured the growth of these cells in the presence or absence of TAM-67. The results of these studies show that the growth of MCF7 cells was suppressed by the AP-1 inhibitor, TAM-67. These results demonstrate that normal and immortalized breast cells, and some breast cancer cells (such as MCF7), require AP-1 to transduce proliferative signals, while other breast cancer cells (such as T47D, MDA MB 231 and MDA MB 435) do not. These studies suggest that the AP-1 transcription factor is a potential target for future agents for the prevention or treatment of breast cancer.
- Published
- 2001
- Full Text
- View/download PDF
11. 'Gain of function' phenotype of tumor-derived mutant p53 requires the oligomerization/nonsequence-specific nucleic acid-binding domain.
- Author
-
Lányi A, Deb D, Seymour RC, Ludes-Meyers JH, Subler MA, and Deb S
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Animals, Biopolymers, Cell Line, ErbB Receptors genetics, Humans, Mice, Phenotype, Proliferating Cell Nuclear Antigen genetics, Promoter Regions, Genetic, Protein Binding, Sequence Deletion, Transcriptional Activation, DNA metabolism, Mutation, Tumor Suppressor Protein p53 genetics
- Abstract
Tumor-derived p53 mutants can transcriptionally activate a number of promoters of genes involved in cellular proliferation. For this transactivation, mutant p53 does not use the wild-type p53 DNA-binding site, suggesting a mechanism of transactivation that is independent of direct DNA binding. Here we describe our analysis of the domain requirements for mutant p53 to transactivate promoters of the human epidermal growth factor receptor (EGFR), human multiple drug resistance 1 (MDR-1) and human proliferating cell nuclear antigen (PCNA) genes. We also report the identification of a structural domain required for the 'gain of function' property of mutant p53-281G. 'Gain of function' is measured as the tumorigenicity (in nude mice) of 10(3) murine cells expressing mutant p53 constitutively. We have generated internal deletion mutants of p53-281G deleting conserved domains I, II, III, IV and V, individually. We have also generated one deletion mutant eliminating amino acids 100 through 300 that removes four of the five conserved domains (II - V); another mutant, p53-281G del 393-327, deletes the oligomerization and nonsequence-specific nucleic acid-binding domains of p53. For the EGFR and MDR-1 promoters, all these mutants have significantly lower transactivation ability than intact p53-281G. These deletion mutants, however, significantly activated the pCNA promoter, suggesting that the mechanism of transactivation of the PCNA promoter is different from that of the EGFR and MDR-1 promoters. When expressed constitutively in 10(3) cells, p53-281G del 393-327 was found to be defective in inducing tumor formation in nude mice although intact p53-281G was very efficient. Thus, our results suggest that structural domains near the C-terminus are needed for 'gain of function'.
- Published
- 1998
- Full Text
- View/download PDF
12. Isolation of an amino-terminal region of bovine papillomavirus type 1 E1 protein that retains origin binding and E2 interaction capacity.
- Author
-
Leng X, Ludes-Meyers JH, and Wilson VG
- Subjects
- Animals, Bovine papillomavirus 1 genetics, Bovine papillomavirus 1 physiology, Cattle, DNA Replication, DNA-Binding Proteins genetics, Mutagenesis, Viral Proteins genetics, Bovine papillomavirus 1 metabolism, DNA metabolism, DNA-Binding Proteins metabolism, Replication Origin, Viral Proteins metabolism
- Abstract
In vitro DNA binding results from a series of E1 proteins containing amino-terminal or carboxy-terminal truncations indicated that sequences between amino acids 121 and 284 were critical for origin binding. Additional binding experiments with E1 proteins containing internal, in-frame insertions or deletions confirmed the importance of the region defined by truncated E1 proteins and also demonstrated that downstream sequences were not required for binding activity in the context of the full-length E1 protein. On the basis of mapping results from the E1 mutants, a clone (pE1(121-311)) was constructed that expressed E1 amino acids within the approximate boundaries of the critical sequences for DNA binding. The E1(121-311) protein retained origin-specific DNA binding, confirming that this region was not only necessary but was also sufficient for origin recognition. In addition to origin binding, E1(121-311) bound E2 protein in a cold-sensitive manner. Therefore, DNA binding and E2 binding activities colocalize to a 191-amino-acid functional domain derived from the amino-terminal half of the E1 protein. Finally, three E1 proteins with mutations in this region all lacked DNA binding activity and were all defective for in vivo replication. Two of these E1 mutants retained E2 binding capability, demonstrating that origin recognition by E1 is critical for replication and cannot necessarily be rescued by an interaction with E2 protein.
- Published
- 1997
- Full Text
- View/download PDF
13. Transcriptional activation of the human epidermal growth factor receptor promoter by human p53.
- Author
-
Ludes-Meyers JH, Subler MA, Shivakumar CV, Munoz RM, Jiang P, Bigger JE, Brown DR, Deb SP, and Deb S
- Subjects
- Amino Acid Sequence, Base Composition, Base Sequence, Binding Sites, Chloramphenicol O-Acetyltransferase biosynthesis, Humans, Molecular Sequence Data, Polymerase Chain Reaction, Recombinant Proteins biosynthesis, Regulatory Sequences, Nucleic Acid, Restriction Mapping, Sequence Deletion, TATA Box, ErbB Receptors biosynthesis, ErbB Receptors genetics, Promoter Regions, Genetic, Transcriptional Activation, Tumor Suppressor Protein p53 metabolism
- Abstract
The human epidermal growth factor receptor (EGFR) promoter is activated by both wild-type and tumor-derived mutant p53. In this communication, we demonstrate that EGFR promoter sequence requirements for transactivation by wild-type and mutant p53 are different. Transient-expression assays with EGFR promoter deletions identified a wild-type human p53 response element, 5'-AGCTAGACGTCCGGGCAGCCCCCGGCG -3', from positions --265 to --239. Electrophoretic mobility shift analysis and DNase I footprinting assays indicated that wild-type p53 binds sequence specifically to the response element. Using circularly permuted DNA fragments containing the p53-binding site, we show that wild-type p53 binding induces DNA bending at this site. We further show that the EGFR promoter is also activated by tumor-derived p53 mutants p53-143A, p53-175H, p53-248W, p53-273H, and p53-281G. However, the transactivation by mutant p53 does not require the wild-type p53-binding site. The minimal EGFR promoter from positions --104 to --20 which does not contain the wild-type p53-binding site is transactivated by the p53 mutants but not by the wild-type protein, showing a difference in the mechanism of transactivation by wild-type and mutant p53. Transactivation of the EGFR promoter by p53 may represent a novel mechanism of cell growth regulation.
- Published
- 1996
- Full Text
- View/download PDF
14. Simple procedure for creation of in-frame deletion mutations throughout an open reading frame.
- Author
-
Ludes-Meyers JH and Wilson VG
- Subjects
- Base Sequence, Cloning, Molecular methods, DNA, Recombinant genetics, DNA, Viral genetics, Escherichia coli genetics, Genome, Viral, Molecular Sequence Data, Bovine papillomavirus 1 genetics, Mutagenesis, Open Reading Frames genetics, Sequence Deletion
- Abstract
A general method is presented for randomly mutagenizing open reading frames (ORF) to generate in-frame deletions and insertions. The protocol requires expression of the ORF of interest as a hybrid ORF-beta-galactosidase fusion protein. This allows colorimetric screening for beta-galactosidase activity during subsequent mutagenesis steps. Consequently, proteins with no suitable phenotypic selection or screening properties can be readily screened for mutations that disrupt and subsequently restore the reading frame of the hybrid protein. In addition, this system provides gene expression for subsequent biochemical analysis of the mutant proteins. The bovine papillomavirus type 1 (BPV-1) EI ORF has been mutagenized using this method as an example.
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.