22 results on '"John H. Ludes‐Meyers"'
Search Results
2. Generation and characterization of mice carrying a conditional allele of the Wwox tumor suppressor gene.
- Author
-
John H Ludes-Meyers, Hyunsuk Kil, Jan Parker-Thornburg, Donna F Kusewitt, Mark T Bedford, and C Marcelo Aldaz
- Subjects
Medicine ,Science - 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
3. Targeting FGFR2 with alofanib (RPT835) shows potent activity in tumour models
- Author
-
Evgenia Stepanova, Jean-Baptiste Joose, Frits Daeyaert, Ilya Tsimafeyeu, Nina Peretolchina, Dmitry Khochenkov, Koen Van Akene, Sergei Tjulandin, Eliso Solomko, Oxana Ryabaya, Mikhail Byakhov, Wei Yin, and John H. Ludes-Meyers
- Subjects
0301 basic medicine ,Cancer Research ,Bevacizumab ,Angiogenesis ,Angiogenesis Inhibitors ,Antineoplastic Agents ,Pharmacology ,Biology ,Benzoates ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Receptor, Fibroblast Growth Factor, Type 2 ,IC50 ,Cell Proliferation ,Sulfonamides ,Cell growth ,Fibroblast growth factor receptor 2 ,Endothelial Cells ,medicine.disease ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Growth inhibition ,Ovarian cancer ,medicine.drug - Abstract
Alofanib (RPT835) is a novel selective allosteric inhibitor of fibroblast growth factor receptor 2 (FGFR2). We showed previously that alofanib could bind to the extracellular domain of FGFR2 and has an inhibitory effect on FGF2-induced phoshphorylation of FRS2α. In the present study, we further showed that alofanib inhibited phosphorylation of FRS2α with the half maximal inhibitory concentration (IC50) values of 7 and 9 nmol/l in cancer cells expressing different FGFR2 isoforms. In a panel of four cell lines representing several tumour types (triple-negative breast cancer, melanoma, and ovarian cancer), alofanib inhibited FGF-mediated proliferation with 50% growth inhibition (GI50) values of 16-370 nmol/l. Alofanib dose dependently inhibited the proliferation and migration of human and mouse endothelial cells (GI50 11-58 nmol/l) compared with brivanib and bevacizumab. Treatment with alofanib ablated experimental FGF-induced angiogenesis in vivo. In a FGFR-driven human tumour xenograft model, oral administration of alofanib was well tolerated and resulted in potent antitumour activity. Importantly, alofanib was effective in FGFR2-expressing models. These results show that alofanib is a potent FGFR2 inhibitor and provide strong rationale for its evaluation in patients with FGFR2-driven cancers.
- Published
- 2016
- Full Text
- View/download PDF
4. WWOXhypomorphic mice display a higher incidence of B-cell lymphomas and develop testicular atrophy
- Author
-
Jan Parker-Thornburg, Maria I. Nunez, Mark T. Bedford, John H. Ludes-Meyers, Hyunsuk Kil, C. Marcelo Aldaz, and Claudio J. Conti
- Subjects
Male ,WWOX ,Cancer Research ,Lymphoma, B-Cell ,Tumor suppressor gene ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Article ,law.invention ,Mice ,law ,Cell Line, Tumor ,Testis ,Gene expression ,Genetics ,medicine ,Animals ,B cell ,Mice, Knockout ,Chromosomal fragile site ,Embryo, Mammalian ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,WW Domain-Containing Oxidoreductase ,Models, Animal ,Knockout mouse ,Cancer research ,Suppressor ,Female ,Atrophy ,Oxidoreductases ,Carcinogenesis - 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 (Wwoxgt/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 Wwoxgt/gt males had high numbers of atrophic seminiferous tubules and reduced fertility when compared with wild-type counterparts. We observed that the Wwoxgt/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. © 2007 Wiley-Liss, Inc.
- Published
- 2007
- Full Text
- View/download PDF
5. Molecular Modeling, de novo Design and Synthesis of a Novel, Extracellular Binding Fibroblast Growth Factor Receptor 2 Inhibitor Alofanib (RPT835)
- Author
-
Koen Jeanne Alfons Van Aken, Ilya Tsimafeyeu, Frits Daeyaert, Mikhail Byakhov, Jean-Baptiste Joos, John H. Ludes-Meyers, and Sergei Tjulandin
- Subjects
0301 basic medicine ,Angiogenesis ,Allosteric regulation ,Antineoplastic Agents ,Biology ,Fibroblast growth factor ,Benzoates ,03 medical and health sciences ,Structure-Activity Relationship ,Allosteric Regulation ,Cell Line, Tumor ,Drug Discovery ,Structure–activity relationship ,Humans ,Receptor, Fibroblast Growth Factor, Type 2 ,Receptor ,Genetics ,Sulfonamides ,Fibroblast growth factor receptor 2 ,Ligand (biochemistry) ,Cell biology ,Molecular Docking Simulation ,030104 developmental biology ,Models, Chemical ,Fibroblast growth factor receptor ,Drug Screening Assays, Antitumor - Abstract
Background: Fibroblast growth factor (FGF) receptors (FGFRs) play a key role in tumor growth and angiogenesis. The present report describes our search for an extracellularly binding FGFR inhibitor using a combined molecular modeling and de novo design strategy. Methods: Based upon crystal structures of the receptor with its native ligand and knowledge of inhibiting peptides, we have developed a computational protocol that predicts the putative binding of a molecule to the extracellular domains of the receptor. This protocol, or scoring function, was used in combination with the de novo synthesis program 'SYNOPSIS' to generate high scoring and synthetically accessible compounds. Results: Eight compounds belonging to 3 separate chemical classes were synthesized. One of these compounds, alofanib (RPT835), was found to be an effective inhibitor of the FGF/FGFR2 pathway. The preclinical in vitro data support an allosteric inhibition mechanism of RPT835. RPT835 potently inhibited growth of KATO III gastric cancer cells expressing FGFR2, with GI50 value of 10 nmol/L. Conclusion: These results provide strong rationale for the evaluation of compound in advanced cancers.
- Published
- 2015
6. 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
-
C. Marcelo Aldaz, Elangovan Thavathiru, Michael C. MacLeod, and John H. Ludes-Meyers
- Subjects
WWOX ,Cancer Research ,Cell cycle checkpoint ,Ultraviolet Rays ,DNA damage ,7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide ,Down-Regulation ,Biology ,medicine.disease_cause ,Article ,FHIT ,Cell Line, Tumor ,Radiation, Ionizing ,medicine ,Humans ,Molecular Biology ,Chromosome Fragile Sites ,Tumor Suppressor Proteins ,Chromosomal fragile site ,Cell cycle ,Molecular biology ,Carcinogens, Environmental ,Acid Anhydride Hydrolases ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,WW Domain-Containing Oxidoreductase ,Chromosome Fragile Site ,Tumor Suppressor Protein p53 ,Oxidoreductases ,Carcinogenesis - 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.
- Published
- 2005
- Full Text
- View/download PDF
7. Frequent downregulation and loss of WWOX gene expression in human hepatocellular carcinoma
- Author
-
Nicholas C. Popescu, Marian E. Durkin, S. W. Park, Claudio M Aldaz, John H. Ludes-Meyers, and Drazen B. Zimonjic
- Subjects
WWOX ,Cancer Research ,Carcinoma, Hepatocellular ,Tumor suppressor gene ,Down-Regulation ,Loss of Heterozygosity ,Apoptosis ,Biology ,WWOX gene expression ,Loss of heterozygosity ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Gene expression ,Tumor Cells, Cultured ,Humans ,Gene ,030304 developmental biology ,0303 health sciences ,Reverse Transcriptase Polymerase Chain Reaction ,Chromosome Fragile Sites ,Tumor Suppressor Proteins ,Chromosomal fragile site ,Liver Neoplasms ,Genetics and Genomics ,hepatocellular carcinoma ,Gene rearrangement ,tumour suppressor gene ,Molecular biology ,chromosome rearrangements ,3. Good health ,Gene Expression Regulation, Neoplastic ,WW Domain-Containing Oxidoreductase ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,fragile sites ,Oxidoreductases ,Chromosomes, Human, Pair 16 ,Gene Deletion ,Microsatellite Repeats - Abstract
The WWOX (WW-domain containing oxidoreductase) is a candidate tumour suppressor gene spanning the same chromosome region, 16q23, as the second most common fragile site (FS), FRA16D. Deletions detected by comparative genomic hybridisation (CGH) and loss of heterozygosity at microsatellite markers on chromosome 16q are common in many human cancers including hepatocellular carcinoma (HCC). The development of human HCC is closely associated with exposure to oncogenic viruses and chemical carcinogens, agents known to frequently target common FS. We examined the status of WWOX genomic DNA, RNA and protein in 18 cell lines derived from human HCC and found recurrent alterations of the gene. Loss of DNA copy-number confined to band 16q23 was detected by CGH in several cell lines. Although homozygous deletions of the WWOX gene were not detected, WWOX mRNA expression was absent or lower in 60% of cell lines. The occurrence of aberrant WWOX reverse transcription-PCR products with deletion of exons 6-8 correlated significantly with altered WWOX expression. All of the cell lines showing mRNA downregulation had a decreased or undetectable level of WWOX protein as demonstrated by Western blotting with antibody to WWOX. Furthermore, 13 out of the 18 cell lines expressed decreased levels or no WWOX protein when compared with normal liver. These results show that WWOX gene is frequently altered in HCC and raise the possibility that this gene is implicated in hepatocarcinogenesis.
- Published
- 2004
- Full Text
- View/download PDF
8. Inhibition of AP-1 transcription factor causes blockade of multiple signal transduction pathways and inhibits breast cancer growth
- Author
-
Rachel Schiff, Yun Zhang, Susan G. Hilsenbeck, Powel H. Brown, Yongmin Liu, Gouqing Ge, Chunhua Lu, Debbie Munoz-Medellin, John H. Ludes-Meyers, C. Kent Osborne, and Hee Tae Kim
- Subjects
Cancer Research ,medicine.drug_class ,Mice, Nude ,Apoptosis ,Breast Neoplasms ,Biology ,Culture Media, Serum-Free ,Mice ,Tumor Cells, Cultured ,Genetics ,medicine ,Animals ,Humans ,Growth Substances ,Molecular Biology ,Transcription factor ,Cell growth ,Kinase ,JNK Mitogen-Activated Protein Kinases ,Estrogens ,Xenograft Model Antitumor Assays ,Molecular biology ,Transcription Factor AP-1 ,AP-1 transcription factor ,Estrogen ,Cell culture ,Mutation ,Cancer research ,Female ,Mitogen-Activated Protein Kinases ,Signal transduction ,Cell Division ,Signal Transduction - Abstract
AP-1 transcription factors play a critical role in signal transduction pathways in many cells. We have investigated the role of AP-1 in controlling proliferative signals in breast cells, and have previously shown that AP-1 complexes are activated by peptide and steroid growth factors in both normal and malignant breast cells. In this study, we investigated the role of AP-1 in transducing proliferative signals induced by peptide and steroid growth factors. We used MCF-7 clones that express a specific inhibitor of AP-1, a dominant-negative cJun mutant (TAM67), under the control of an inducible promoter to investigate the role of AP-1 in regulating breast cancer growth. In the presence of doxycycline (Dox), the AP-1 inhibitor was not expressed, and the MCF-7 clones proliferated normally in response to serum stimulation. However, when Dox was withdrawn, TAM67 was expressed, AP-1 activity was inhibited, and serum-induced proliferation was blocked. We next investigated whether the mitogenic response to specific growth factors also requires AP-1. MCF-7 Tet-Off-TAM67 cells were grown in the presence of increasing concentrations of IGF-1, EGF, heregulin-beta, bFGF, or estrogen under un-induced and induced conditions. These studies showed that the AP-1 inhibitor completely blocked proliferation in response to the peptide growth factors (IGF-1, EGF, heregulin-beta, and bFGF), and partially blocked the response to estrogen. To investigate the effect of AP-1 blockade on in vivo tumor growth, we injected the MCF-7 Tet-Off TAM67 cells into nude mice receiving doxycycline to suppress the expression of the AP-1 inhibitor. After the mice developed tumors, they were randomized to either continue to receive Dox or not. In mice not receiving Dox, the expression of TAM67 was induced, and tumor growth was inhibited, while the tumors in mice receiving Dox continued to grow. Analysis of the tumors from these mice showed that the expression of TAM67 caused reduced proliferation of the breast cancer cells without inducing apoptosis. These results demonstrate that AP-1 blockade supresses mitogenic signals from multiple different peptide growth factors as well as estrogen, and inhibits the growth of MCF-7 breast cancer cells both in vitro and in vivo. These results suggest that novel agents specifically targeting AP-1 or its activating kinases could be promising agents for the treatment of breast cancer.
- Published
- 2002
- 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
-
Rafael E. Herrera, Michael C. Yang, Powel H. Brown, Zafar Nawaz, Holly Hong Zhao, Suzanne A. W. Fuqua, Douglas Yee, Ester Coronado-Heinsohn, Karen J. Seybold-Tilson, Sami Diab, C. Kent Osborne, John H. Ludes-Meyers, and Frederic G. Barr
- Subjects
Transcriptional Activation ,DNA, Complementary ,Recombinant Fusion Proteins ,Blotting, Western ,Molecular Sequence Data ,Mice, Nude ,Estrogen receptor ,Breast Neoplasms ,Biology ,Ligands ,Transfection ,Biochemistry ,Mice ,Transactivation ,Two-Hybrid System Techniques ,Rhabdomyosarcoma ,Tumor Cells, Cultured ,Animals ,Humans ,Tissue Distribution ,Amino Acid Sequence ,Luciferases ,Molecular Biology ,Gene Library ,Glutathione Transferase ,Cell Nucleus ,Thyroid hormone receptor ,Sequence Homology, Amino Acid ,Forkhead Box Protein O1 ,Forkhead Transcription Factors ,Cell Biology ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Retinoic acid receptor ,Receptors, Estrogen ,Nuclear receptor ,COS Cells ,Nuclear receptor coactivator 3 ,Cancer research ,Nuclear receptor coactivator 2 ,Estrogen-related receptor gamma ,Plasmids ,Protein Binding ,Signal Transduction ,Transcription Factors - 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. 'Gain of function' phenotype of tumor-derived mutant p53 requires the oligomerization/nonsequence-specific nucleic acid-binding domain
- Author
-
Mark A. Subler, Sumitra Deb, Debabrita Deb, Seymour Rc, Arpad Lanyi, and John H. Ludes-Meyers
- Subjects
Transcriptional Activation ,Cancer Research ,Mutant ,Plasma protein binding ,Biology ,medicine.disease_cause ,Cell Line ,Mice ,Transactivation ,Biopolymers ,Proliferating Cell Nuclear Antigen ,Gene expression ,Genetics ,medicine ,Animals ,Humans ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,Sequence Deletion ,Mutation ,Promoter ,DNA ,Cell biology ,ErbB Receptors ,Phenotype ,Tumor Suppressor Protein p53 ,Protein Binding ,Binding domain - 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
11. Corrigendum to ‘Targeting FGFR2 with alofanib (RPT835) shows potent activity in tumour models’ [Eur J Cancer 61 (2016) 20–28]
- Author
-
Wei Yin, Nina Peretolchina, Evgenia Stepanova, Mikhail Byakhov, Eliso Solomko, Oxana Ryabaya, Koen Van Akene, Jean-Baptiste Joose, John H. Ludes-Meyers, Frits Daeyaert, Dmitry Khochenkov, Sergei Tjulandin, and Ilya Tsimafeyeu
- Subjects
Cancer Research ,Oncology ,business.industry ,Cancer research ,Medicine ,Cancer ,business ,medicine.disease - Published
- 2017
- Full Text
- View/download PDF
12. Partite expression of the bovine papillomavirus E1 open reading frame in Escherichia coli
- Author
-
John H. Ludes-Meyers and Van G. Wilson
- Subjects
Recombinant Fusion Proteins ,Biophysics ,medicine.disease_cause ,Biochemistry ,Open Reading Frames ,Viral Proteins ,Structural Biology ,Gene expression ,Escherichia coli ,Genetics ,medicine ,Gene ,Bovine papillomavirus 1 ,Bovine papillomavirus ,Antiserum ,biology ,Immune Sera ,beta-Galactosidase ,biology.organism_classification ,Precipitin Tests ,Fusion protein ,Virology ,Molecular biology ,Enterobacteriaceae ,DNA-Binding Proteins ,Open reading frame ,Bacterial Outer Membrane Proteins - Abstract
Six recombinants were constructed which expressed portions of the bovine papillomavirus E1 open reading frame as OmpF/E1/beta-galactosidase tribrid fusion proteins in Escherichia coli. Rabbit sera containing E1-specific antibodies were generated against five of these six fusion proteins (which together constitute 74% of the full-length E1 open reading frame). The individual fusion proteins and their cognate antisera will be useful reagents for defining the structure and function of the BPV E1 protein(s).
- Published
- 1992
- Full Text
- View/download PDF
13. Abstract P6-02-07: Targeting triple-negative breast cancer with fibroblast growth factor receptor 2 allosteric inhibitor RPT835
- Author
-
Frits Daeyaert, Sergey A Tjulandin, Ilya Tsimafeyeu, John H. Ludes-Meyers, Mikhail Y Byakhov, and Wei Yin
- Subjects
Cancer Research ,business.industry ,Fibroblast growth factor receptor 2 ,Xenotransplantation ,medicine.medical_treatment ,Mrna expression ,Allosteric regulation ,Cancer ,medicine.disease ,Breast cancer ,Oncology ,Immunology ,medicine ,Cancer research ,business ,Triple-negative breast cancer ,Tumor xenograft - Abstract
Background: FGFR2 is amplified in 4% of triple-negative breast cancers (TNBC). In addition, the mRNA expression levels of FGFR2 were significantly increased in amplified vs nonamplified tumor samples, suggesting a potential role for FGFR2 in TNBC (Turner et al. 2010). We evaluated efficacy of allosteric inhibition of FGFR2 in high- and low-expressing TNBC xenografts. Methods: Immunocompromised mice were used for xenotransplantation of FGFR2 high-expressing TNBC cells (SUM52PE) or FGFR2 low-expressing TNBC cells (HS578T). Forty animals with measurable tumors were selected on day 10 and randomized into treatment groups (low-molecular weight allosteric inhibitor RPT835, 30 mg/kg; gavage, daily) or vehicle (water; gavage, daily). Measurements of tumor volume (mm3) were performed by digital calipers every 3 days during 40 days after tumor inoculation. Results: RPT835 significantly inhibited aggressive growth of SUM52PE tumor xenograft (P Conclusions: The allosteric FGFR2 inhibitor RPT835 significantly impacts on growth of FGFR2-expressing TNBC. Citation Format: Sergey A Tjulandin, Ilya V Tsimafeyeu, Mikhail Y Byakhov, Wei Yin, John Ludes-Meyers, Frits Daeyaert. Targeting triple-negative breast cancer with fibroblast growth factor receptor 2 allosteric inhibitor RPT835 [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr P6-02-07.
- Published
- 2015
- Full Text
- View/download PDF
14. WWOX protein expression varies among ovarian carcinoma histotypes and correlates with less favorable outcome
- Author
-
Andres J. Klein-Szanto, John H. Ludes-Meyers, Jinsong Liu, Gordon B. Mills, Andrew K. Godwin, Maria I. Nunez, Robert E. Page, Daniel G. Rosen, C. Marcelo Aldaz, Martín Carlos Abba, and Hyunsuk Kil
- Subjects
WWOX ,Cancer Research ,Time Factors ,Blotting, Western ,Immunoblotting ,Loss of Heterozygosity ,Ovary ,Biology ,lcsh:RC254-282 ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Ovarian carcinoma ,Genetics ,medicine ,Humans ,Genes, Tumor Suppressor ,10. No inequality ,Oligonucleotide Array Sequence Analysis ,030304 developmental biology ,Ovarian Neoplasms ,0303 health sciences ,Tissue microarray ,Tumor Suppressor Proteins ,Chromosomal fragile site ,Homozygote ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,medicine.anatomical_structure ,WW Domain-Containing Oxidoreductase ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Oxidoreductases ,Receptors, Progesterone ,Gene Deletion ,Clear cell ,Research Article - 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 χ2 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
15. WWOX binds the specific proline-rich ligand PPXY: identification of candidate interacting proteins
- Author
-
Andrzej K. Bednarek, Mark T. Bedford, John H. Ludes-Meyers, Hyunsuk Kil, C. Marcelo Aldaz, and Jeff Drake
- Subjects
WWOX ,Cancer Research ,Proline ,Molecular Sequence Data ,Golgi Apparatus ,Plasma protein binding ,Biology ,Ligands ,Article ,Protein–protein interaction ,WW domain ,Genetics ,Tumor Cells, Cultured ,Humans ,Amino Acid Sequence ,Molecular Biology ,Transcription factor ,Late endosome ,Binding Sites ,Chromosomal fragile site ,Tumor Suppressor Proteins ,Nuclear Proteins ,Cell biology ,Protein Structure, Tertiary ,Membrane protein ,WW Domain-Containing Oxidoreductase ,Cancer research ,biology.protein ,Oxidoreductases ,Protein Binding ,Transcription Factors - 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 peri-nuclear 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
16. WWOX, the common chromosomal fragile site, FRA16D, cancer gene
- Author
-
Claudio M Aldaz, Andrzej K. Bednarek, Nicholas C. Popescu, Mark T. Bedford, and John H. Ludes-Meyers
- Subjects
WWOX ,Mitotic crossover ,Tumor suppressor gene ,Blotting, Western ,Molecular Sequence Data ,Golgi Apparatus ,Loss of Heterozygosity ,Breast Neoplasms ,medicine.disease_cause ,Article ,WW domain ,Exon ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Genes, Tumor Suppressor ,Amino Acid Sequence ,RNA, Messenger ,Cloning, Molecular ,Molecular Biology ,Genetics (clinical) ,In Situ Hybridization, Fluorescence ,Short-chain dehydrogenase ,biology ,Chromosomal fragile site ,Chromosome Fragile Sites ,Chromosome Mapping ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Alternative Splicing ,Review Literature as Topic ,biology.protein ,Carcinogenesis ,Carrier Proteins ,Chromosomes, Human, Pair 16 - 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.
- Published
- 2003
17. 476 FGFR2 targeting with allosteric inhibitor RPT835
- Author
-
Sergey Tjulandin, Frits Daeyaert, Mikhail Byakhov, John H. Ludes-Meyers, Wei Yin, and Ilya Tsimafeyeu
- Subjects
Cancer Research ,Oncology ,Chemistry ,Allosteric regulation ,Pharmacology - Published
- 2014
- Full Text
- View/download PDF
18. Isolation of an amino-terminal region of bovine papillomavirus type 1 E1 protein that retains origin binding and E2 interaction capacity
- Author
-
Van G. Wilson, X Leng, and John H. Ludes-Meyers
- Subjects
DNA Replication ,HMG-box ,Immunology ,Replication Origin ,Microbiology ,Single-stranded binding protein ,Viral Proteins ,SeqA protein domain ,Virology ,Animals ,Replication protein A ,Bovine papillomavirus 1 ,Telomere-binding protein ,biology ,Binding protein ,DNA ,Molecular biology ,DNA binding site ,DNA-Binding Proteins ,Biochemistry ,Mutagenesis ,Insect Science ,biology.protein ,Cattle ,Binding domain ,Research Article - 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
19. Transcriptional activation of the human epidermal growth factor receptor promoter by human p53
- Author
-
R. M. Munoz, P. Jiang, S Deb, Mark A. Subler, J. E. Bigger, Sumitra Deb, D. R. Brown, C. V. Shivakumar, and John H. Ludes-Meyers
- Subjects
Chloramphenicol O-Acetyltransferase ,Transcriptional Activation ,TATA box ,Response element ,Mutant ,Molecular Sequence Data ,Restriction Mapping ,Biology ,Regulatory Sequences, Nucleic Acid ,Polymerase Chain Reaction ,chemistry.chemical_compound ,Transactivation ,Humans ,Amino Acid Sequence ,Binding site ,Promoter Regions, Genetic ,Molecular Biology ,Peptide sequence ,Sequence Deletion ,Base Composition ,Binding Sites ,Base Sequence ,Cell Biology ,Molecular biology ,TATA Box ,Recombinant Proteins ,ErbB Receptors ,chemistry ,Regulatory sequence ,Tumor Suppressor Protein p53 ,DNA ,Research Article - 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
20. CtIP, a BRCA1 binding protein, is a transcriptional corepressor of estrogen receptor α
- Author
-
Claudio M Aldaz, Min Wu, and John H. Ludes-Meyers
- Subjects
Cancer Research ,medicine.medical_specialty ,Tumor suppressor gene ,Estrogen receptor ,Biology ,Chromatin remodeling ,Endocrinology ,Oncology ,Internal medicine ,medicine ,Transcriptional regulation ,Cancer research ,Gene silencing ,skin and connective tissue diseases ,Corepressor ,Estrogen receptor alpha ,Tamoxifen ,medicine.drug - Abstract
Abstract #606 The role of estrogen and estrogen receptor (ER) transcriptional regulation in breast carcinogenesis is indisputable. In the last few years there has been dramatic progress in understanding how receptor transcriptional coactivator and corepressor complexes operate. We demonstrated that CtIP (CtBP interacting protein) is downregulated in tamoxifen resistant breast cancer lines. Furthermore, CtIP protein expression correlates with response to neo-adjuvant endocrine therapy and patients with progressive disease express lower CtIP protein in their primary breast carcinomas than those who respond. CtIP expression directly correlates with ER alpha expression, and inversely correlates with disease free survival and breast cancer metastasis. Importantly, silencing endogenous CtIP confers tamoxifen resistance in tamoxifen sensitive breast cancer cells and re-expression restored tamoxifen sensitivity in resistant breast cancer cells (Wu et al. Molec. Cancer Res. 2007; 5: 1285-1295). Our findings indicate that CtIP silencing is a novel mechanism for the development of tamoxifen resistance in breast cancer. CtIP interacts with the BRCT domains of BRCA1 where most mutations occur in familial BRCA1 breast cancers. CtIP also interacts with the transcriptional corepressor CtBP. Emerging evidence suggests that CtIP is involved in transcriptional repression and may be a tumor suppressor gene. We hypothesize that CtIP plays a key role in repression of ER transcription and such repressive function is linked to the inhibitory growth effects exerted by tamoxifen. We further speculate that CtIP inhibition of ER transcription is mediated by its ability to bridge BRCA1 with CtBP to form a transcriptional corepressor complex able to modulate ER activity via the interaction between BRCA1 and ER and that CtIP is in fact required for BRCA1 to exert its transcriptional repression activity on ER. We will present our work showing that CtIP, BRCA1 and CtBP form a multiprotein complex, that requires CtIP, and associates with ER in breast cancer cells. More importantly, we will show that BRCA1, CtIP and CtBP associate with ER on the ER-dependent promoter of the pS2 gene in the absence of ligand. Stimulation by ligand leads to dissociation of the complex and ER-dependent gene expression. Furthermore, silencing CtIP expression using RNAi in E2-starved MCF-7 cells led to an increase of basal transcription of the pS2 gene. CtIP was also observed to repress E2 stimulation of ERE-luciferase in transfected MCF-7 cells. We are currently characterizing the role of the CtIP corepressor complex in ER-transcriptional activity. We will present the results of our efforts to identify specific ER-dependent promoters repressed by the CtIP corepressor. In addition, we will present results of studies aimed at characterizing CtIP-specific changes in chromatin remodeling and ER-cofactor recruitment at ER-dependent promoters. Citation Information: Cancer Res 2009;69(2 Suppl):Abstract nr 606.
- Published
- 2009
- Full Text
- View/download PDF
21. A bovine papillomavirus E1-related protein binds specifically to bovine papillomavirus DNA
- Author
-
John H. Ludes-Meyers and Van G. Wilson
- Subjects
Genes, Viral ,viruses ,Recombinant Fusion Proteins ,Immunology ,Blotting, Western ,Microbiology ,Binding, Competitive ,law.invention ,Cell Line ,chemistry.chemical_compound ,Open Reading Frames ,Viral Proteins ,Recognition sequence ,law ,Virology ,Escherichia coli ,Animals ,Binding site ,Cloning, Molecular ,Codon ,Bovine papillomavirus ,Bovine papillomavirus 1 ,Binding Sites ,biology ,Bovine Papillomavirus-1 ,Immune Sera ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Fusion protein ,Molecular biology ,DNA binding site ,DNA-Binding Proteins ,Kinetics ,chemistry ,Insect Science ,DNA, Viral ,Recombinant DNA ,DNA ,Plasmids ,Research Article - Abstract
The E1 open reading frame of bovine papillomavirus (BPV) was expressed as a RecA-E1 fusion protein in Escherichia coli. The bacterially expressed RecA-E1 protein exhibited sequence-specific DNA binding activity; strong binding to the region from nucleotides 7819 to 93 on the BPV genome (designated region A) and weak binding to the adjacent region from nucleotides 7457 to 7818 (region B) were observed. The interaction between the BPV-derived RecA-E1 protein and region A appeared to be highly specific for BPV DNA, as no comparable binding was detected with heterologous papillomavirus DNAs. Binding to region A was eliminated by digestion of region A at the unique HpaI site, which suggests that the RecA-E1 binding site(s) was at or near the HpaI recognition sequence. Binding to region B but not region A was observed when nuclear extracts from ID13 cells were used as a source of E1 proteins. The absence of region A binding by ID13 extracts may reflect a negative regulation of E1 DNA binding activity.
- Published
- 1991
22. 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
-
Elangovan Thavathiru, John H. Ludes‐Meyers, Michael C. MacLeod, and C. Marcelo Aldaz
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.