48 results on '"Rosalyn B, Irby"'
Search Results
2. The Akt inhibitor ISC-4 synergizes with cetuximab in 5-FU-resistant colon cancer.
- Author
-
Joshua E Allen, Jean-Nicolas Gallant, David T Dicker, Shantu Amin, Rosalyn B Irby, Arun K Sharma, and Wafik S El-Deiry
- Subjects
Medicine ,Science - Abstract
Phenylbutyl isoselenocyanate (ISC-4) is an Akt inhibitor with demonstrated preclinical efficacy against melanoma and colon cancer. In this study, we sought to improve the clinical utility of ISC-4 by identifying a synergistic combination with FDA-approved anti-cancer therapies, a relevant and appropriate disease setting for testing, and biomarkers of response. We tested the activity of ISC-4 and 19 FDA-approved anticancer agents, alone or in combination, against the SW480 and RKO human colon cancer cell lines. A synergistic interaction with cetuximab was identified and validated in a panel of additional colon cancer cell lines, as well as the kinetics of synergy. ISC-4 in combination with cetuximab synergistically reduced the viability of human colon cancer cells with wild-type but not mutant KRAS genes. Further analysis revealed that the combination therapy cooperatively decreased cell cycle progression, increased caspase-dependent apoptosis, and decreased phospho-Akt in responsive tumor cells. The synergism between ISC-4 and cetuximab was retained independently of acquired resistance to 5-FU in human colon cancer cells. The combination demonstrated synergistic anti-tumor effects in vivo without toxicity and in the face of resistance to 5-FU. These results suggest that combining ISC-4 and cetuximab should be explored in patients with 5-FU-resistant colon cancer harboring wild-type KRAS.
- Published
- 2013
- Full Text
- View/download PDF
3. Correction: The Akt Inhibitor ISC-4 Synergizes with Cetuximab in 5-FU-Resistant Colon Cancer.
- Author
-
Joshua E. Allen, Jean-Nicolas Gallant, David T. Dicker, Shantu Amin, Rosalyn B. Irby, Arun K. Sharma, and Wafik S. El-Deiry
- Subjects
Medicine ,Science - Published
- 2013
- Full Text
- View/download PDF
4. Overexpression Of Prostate Apoptosis Response Protein-4 In Colon Cancer Cells Can Inhibit Metastasis By Upregulating E-cadherin Expression
- Author
-
Christina Leah B. Kline, Rosalyn B. Irby, Jeffrey Q. Nguyen, and Natalia Caballero
- Subjects
Oncology ,medicine.medical_specialty ,biology ,Cadherin ,business.industry ,Colorectal cancer ,Lewis lung carcinoma ,Vimentin ,Cell migration ,medicine.disease ,Metastasis ,Downregulation and upregulation ,Apoptosis ,Internal medicine ,medicine ,biology.protein ,Cancer research ,business - Abstract
Colon cancer has a five-year survival of 64.7%, and about 50,000 people are expected to die from colon cancer this year. Patients with metastatic colorectal cancer have a significantly worse prognosis, a 12.9% five -year survival. This emphasizes the need for strategies to inhibit the growth and metastases of colorectal cancer. Prostate apoptosis response protein 4 (Par-4) is a pro-apoptotic protein that has been shown to mediate apoptosis in response to stimuli, such as chemotherapeutics and radiation. Recombinant Par-4 protein has been shown to reduce the occurrence of Lewis lung carcinoma metastases in-vivo; however, the mechanism by which Par-4 can inhibit metastasis has not been elucidated. In this study, human colon cancer cell lines SW480 and SW620 were transfected with Par-4 plasmid or anti-Par-4 shRNA, and the effect on metastasis was examined. Par-4 overexpression inhibited cell migration and invasion, while Par-4 knockdown promoted it. Moreover, the morphology of SW620 cells was altered when Par-4 levels were increased. The change was characteristic of a mesenchymal-to-epithelial transition (MET) in these cells. MET can be induced by upregulation of E-cadherin expression, and RT-PCR and Western blot analyses showed that E-cadherin mRNA and protein levels, respectively, were increased in the Par-4 overexpressing cells concomitant with a decrease in vimentin. The results of this study demonstrate the potential of Par-4 in colon cancer therapy, not only in primary tumors but also in metastatic cells. DOI : 10.14302/issn.2471-7061.jcrc-14-574 Corresponding Author: Rosalyn B. Irby, Ph.D. 500 University Drive Hershey, PA 17033 Tel: 717-531-5035 Email: rirby@hmc.psu.edu
- Published
- 2015
- Full Text
- View/download PDF
5. TRIM21 is a novel regulator of Par-4 in colon and pancreatic cancer cells
- Author
-
Jeffrey Q. Nguyen and Rosalyn B. Irby
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,pancreatic cancer ,Regulator ,Transfection ,Mass Spectrometry ,03 medical and health sciences ,Pancreatic cancer ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,Immunoprecipitation ,Pharmacology ,Cisplatin ,Mechanism (biology) ,business.industry ,medicine.disease ,Prognosis ,Pancreatic Neoplasms ,030104 developmental biology ,proteasome ,Proteasome ,Ribonucleoproteins ,colon cancer ,Apoptosis ,Colonic Neoplasms ,Cancer research ,Molecular Medicine ,CA19-9 ,business ,Apoptosis Regulatory Proteins ,Par-4 ,TRIM21 ,medicine.drug ,Research Paper - Abstract
The prostate apoptosis response protein 4 (Par-4) is a tumor-suppressor that has been shown to induce cancer-cell selective apoptosis in a variety of cancers. The regulation of Par-4 expression and activity is a relatively understudied area, and identifying novel regulators of Par-4 may serve as novel therapeutic targets. To identify novel regulators of Par-4, a co-immunoprecipitation was performed in colon cancer cells, and co-precipitated proteins were identified by mass-spectometry. TRIM21 was identified as a novel interacting partner of Par-4, and further shown to interact with Par-4 endogenously and through its PRY-SPRY domain. Additional studies show that TRIM21 downregulates Par-4 levels in response to cisplatin, and that TRIM21 can increase the resistance of colon cancer cells to cisplatin. Furthermore, forced Par-4 expression can sensitize pancreatic cancer cells to cisplatin. Finally, we demonstrate that TRIM21 expression predicts survival in pancreatic cancer patients. Our work highlights a novel mechanism of Par-4 regulation, and identifies a novel prognostic marker and potential therapeutic target for pancreatic cancer.
- Published
- 2016
6. The pro-apoptotic protein Prostate Apoptosis Response Protein-4 (Par-4) can be activated in colon cancer cells by treatment with Src inhibitor and 5-FU
- Author
-
Christina Leah B. Kline and Rosalyn B. Irby
- Subjects
Exonucleases ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Apoptosis ,Biology ,Prostate cancer ,Cell Line, Tumor ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Kinase activity ,Protein Kinase Inhibitors ,Pharmacology ,Biochemistry (medical) ,Cell Biology ,medicine.disease ,Gene Expression Regulation, Neoplastic ,src-Family Kinases ,Endocrinology ,14-3-3 Proteins ,Tumor progression ,Colonic Neoplasms ,Exoribonucleases ,Cancer research ,Phosphorylation ,Fluorouracil ,Apoptosis Regulatory Proteins ,Proto-Oncogene Proteins c-akt ,Tyrosine kinase ,Protein Binding ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The overexpression of the pro-apoptotic protein Prostate Apoptosis Response Protein-4 in colon cancer has been shown to increase response to the chemotherapeutic agent 5-fluorouracil (5-FU). Although colon cancer cells endogenously express Par-4, the presence or overexpression of Par-4 alone does not cause apoptosis. We hypothesize that Par-4 is inactivated in colon cancer. In colon cancer, the levels and the kinase activity of the nonreceptor tyrosine kinase c-Src increase with tumor progression. One of the downstream effectors of c-Src is Akt1. Akt1 has been shown to inhibit the pro-apoptotic activity of Par-4 in prostate cancer cells. We therefore investigated the potential of activating Par-4 by inhibiting c-Src. Colon carcinoma cell lines were treated with the Src kinase inhibitor 4-amino-5-(4-chlorophenyl)-7-(dimethylethyl)pyrazolo[3,4-d]pyrimidine (PP2) in combination with the chemotherapeutic agent 5-FU. Treating cells with PP2 and 5-FU resulted in reduced interaction of Par-4 with Akt1 and with the scaffolding protein 14-3-3σ, and mobilization of Par-4 to the nucleus. Par-4 was shown to interact not only with Akt1 and 14-3-3σ, but also with c-Src. Overexpression of c-Src induced the phosphorylation of Par-4 at tyrosine site/s. Thus, in this study, we have shown that Par-4 can be activated by inhibiting Src with a pharmacological inhibitor and adding a chemotherapeutic agent. The activation of the pro-apoptotic protein Par-4 as reported in this study is a novel mechanism by which apoptosis occurs with a Src kinase inhibitor and 5-FU. In addition, we have demonstrated that the pro-apoptotic activity of endogenously expressed Par-4 can be increased in colon cancer cells.
- Published
- 2011
- Full Text
- View/download PDF
7. The Akt Inhibitor ISC-4 Activates Prostate Apoptosis Response Protein-4 and Reduces Colon Tumor Growth in a Nude Mouse Model
- Author
-
Christina Leah B. Kline, Arun Sharma, Arthur Berg, Shantu Amin, and Rosalyn B. Irby
- Subjects
Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,Mice, Nude ,AKT1 ,Antineoplastic Agents ,Apoptosis ,Article ,Mice ,HT29 Cells ,Nude mouse ,Cell Line, Tumor ,Organoselenium Compounds ,Internal medicine ,medicine ,Animals ,Humans ,Endoplasmic Reticulum Chaperone BiP ,Protein Kinase Inhibitors ,Protein kinase B ,Cell Death ,biology ,Cancer ,Bystander Effect ,HCT116 Cells ,biology.organism_classification ,medicine.disease ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Endocrinology ,Oncology ,Cell culture ,Colonic Neoplasms ,Cancer research ,Female ,Apoptosis Regulatory Proteins ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Purpose: Prostate apoptosis response protein-4 (Par-4) sensitizes cells to chemotherapy; however, Akt1 inactivates Par-4. Previously we showed that Par-4–overexpressing colon cancer cells responded more readily to 5-fluorouracil (5-FU) than their wild-type counterparts. In this study we investigated (i) the effects of the Akt inhibitor, phenylbutyl isoselenocyanate (ISC-4), on tumor growth in nude mice and (ii) bystander effect of Par-4–overexpressing cells on wild-type tumor growth. Experimental Design: Mice (n = 80) were injected with wild-type HT29 human colon cancer cells in the right flank. Forty of the mice were also injected in the left flank with HT29 cells engineered to overexpress Par-4. The mice were treated with 5-FU, ISC-4, a combination, or vehicle. Results: ISC-4 reduced tumor growth, with or without 5-FU. When Par-4–overexpressing tumors were present, wild-type tumors grew more slowly compared to when no Par-4–overexpressing tumors were present. The level of Par-4 protein as well as the Par-4 binding protein, GRP78, was increased in wild-type cells growing in the same mouse as Par-4–overexpressing tumors compared with wild-type tumors growing without Par-4–overexpressing tumors. Conclusions: Par-4–overexpressing tumors exhibited a bystander effect on wild-type tumors growing distally in the same mouse. This suggests that gene therapy need not achieve total penetration to have a positive effect on tumor treatment. Inhibition of Akt with ISC-4 inhibited tumor growth and had a greater effect on cells overexpressing Par-4. The data indicate ISC-4 alone or in combination with Par-4 can greatly reduce tumor growth. Clin Cancer Res; 17(13); 4474–83. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
8. Tyrosine Kinase Update: Role and Response in Cancer Therapy
- Author
-
Yixing Jiang and Rosalyn B. Irby
- Subjects
Cancer Research ,Oncology ,business.industry ,Cancer therapy ,Cancer research ,Molecular Medicine ,Medicine ,business ,Tyrosine kinase - Published
- 2011
- Full Text
- View/download PDF
9. Tumor Necrosis Factor α Induces p53 Up-regulated Modulator of Apoptosis Expression in Colorectal Cancer Cell Lines
- Author
-
Lisa S. Poritz, Danielle M. Pastor, and Rosalyn B. Irby
- Subjects
medicine.medical_treatment ,Immunoblotting ,Fluorescent Antibody Technique ,Apoptosis ,medicine.disease_cause ,HT29 Cells ,Cell Line, Tumor ,Puma ,Humans ,Medicine ,Analysis of Variance ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Necrosis Factor-alpha ,business.industry ,Gastroenterology ,Wild type ,General Medicine ,biology.organism_classification ,digestive system diseases ,Up-Regulation ,Cytokine ,Cell culture ,Immunology ,Cancer research ,Tumor necrosis factor alpha ,Tumor Suppressor Protein p53 ,biological phenomena, cell phenomena, and immunity ,Apoptosis Regulatory Proteins ,Colorectal Neoplasms ,business ,Carcinogenesis - Abstract
Purpose Inflammatory bowel disease (IBD)-associated colorectal carcinogenesis involves dysregulation of multiple cellular pathways, including p53 signaling and cytokine action. The purpose of the current study was to evaluate the effects of tumor necrosis factor alpha (TNF-alpha) on p53 and p53 up-regulated modulator of apoptosis (PUMA), a downstream effector of p53 in the apoptotic pathway in colorectal cancer cells. Methods The cell lines HT29 (which express mutant p53) and HCT116 (which express wild-type p53) were treated with TNF-alpha (0, 50, 100, or 500 ng/mL) for 1, 12, 24, or 48 hours. Protein expression and subcellular localization of p53 and PUMA were determined by immunoblot and immunofluorescence. Changes in p53 and PUMA mRNA expression were determined by quantitative real time polymerase chain reaction. Results Nuclear p53 expression was increased in TNF-alpha-treated HT29 cells; in contrast, expression was decreased or minimally changed in TNF-alpha-treated HCT116 cells, as determined by immunoblot and immunofluorescence. At 24 hours, p53 mRNA transcript levels were minimally increased in HT29 cells, whereas PUMA increased 34-fold. Conclusions TNF-alpha increased nuclear p53 expression in HT29 cells, which express p53 mutation, but not in HCT116 cells, which are wild type for p53. In addition, TNF-alpha markedly up-regulated PUMA mRNA levels in HT29 cells. Our findings suggest that TNF-alpha may be a factor in carcinogenesis in IBD in cells carrying a p53 mutation.
- Published
- 2010
- Full Text
- View/download PDF
10. Platelet-derived growth factor mediates survival of leukemic large granular lymphocytes via an autocrine regulatory pathway
- Author
-
Kathleen Broeg, Kendall Thomas Baab, Xin Liu, Lindsay Ryland, Ranran Zhang, Jun Yang, Thomas P. Loughran, Susan B. Nyland, Rosalyn B. Irby, and Nancy Ruth Jarbadan
- Subjects
Platelet-derived growth factor ,Cell Survival ,Immunology ,Becaplermin ,Apoptosis ,Enzyme-Linked Immunosorbent Assay ,Biochemistry ,Receptor, Platelet-Derived Growth Factor beta ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,Growth factor receptor ,Cell Line, Tumor ,medicine ,Humans ,Cytotoxic T cell ,Lymphocytes ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Autocrine signalling ,Protein kinase B ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,Platelet-Derived Growth Factor ,Lymphoid Neoplasia ,Staining and Labeling ,biology ,Gene Expression Regulation, Leukemic ,Proto-Oncogene Proteins c-sis ,Cell Biology ,Hematology ,medicine.disease ,Antibodies, Neutralizing ,Immunohistochemistry ,Leukemia, Large Granular Lymphocytic ,Autocrine Communication ,Leukemia ,src-Family Kinases ,chemistry ,biology.protein ,Cancer research ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Platelet-derived growth factor receptor ,Signal Transduction - Abstract
Large granular lymphocyte (LGL) leukemia results from chronic expansion of cytotoxic T cells or natural killer (NK) cells. Apoptotic resistance resulting from constitutive activation of survival signaling pathways is a fundamental pathogenic mechanism. Recent network modeling analyses identified platelet-derived growth factor (PDGF) as a key master switch in controlling these survival pathways in T-cell LGL leukemia. Here we show that an autocrine PDGF regulatory loop mediates survival of leukemic LGLs of both T- and NK-cell origin. We found high levels of circulating PDGF-BB in platelet-poor plasma samples from LGL leukemia patients. Production of PDGF-BB by leukemic LGLs was demonstrated by immunocytochemical staining. Leukemic cells expressed much higher levels of PDGFR-β transcripts than purified normal CD8+ T cells or NK cells. We observed that phosphatidylinositol-3-kinase (PI3 kinase), Src family kinase (SFK), and downstream protein kinase B (PKB)/AKT pathways were constitutively activated in both T- and NK-LGL leukemia. Pharmacologic blockade of these pathways led to apoptosis of leukemic LGLs. Neutralizing antibody to PDGF-BB inhibited PKB/AKT phosphorylation induced by LGL leukemia sera. These results suggest that targeting of PDGF-BB, a pivotal regulator for the long-term survival of leukemic LGLs, may be an important therapeutic strategy.
- Published
- 2010
- Full Text
- View/download PDF
11. Delivery of PAR-4 plasmid in vivo via nanoliposomes sensitizes colon tumor cells subcutaneously implanted into nude mice to 5-FU
- Author
-
Christina Leah B. Kline, Rosalyn B. Irby, Mark Kester, and Sriram S. Shanmugavelandy
- Subjects
Antimetabolites, Antineoplastic ,Cancer Research ,Colorectal cancer ,medicine.medical_treatment ,Mice, Nude ,Apoptosis ,Biology ,Transfection ,Mice ,HT29 Cells ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Pharmacology ,Chemotherapy ,Genetic Therapy ,medicine.disease ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Molecular biology ,Oncology ,Cell culture ,Colonic Neoplasms ,Liposomes ,Cancer cell ,Nanoparticles ,Molecular Medicine ,Receptors, Thrombin ,Fluorouracil ,HeLa Cells ,Plasmids - Abstract
The prostate apoptosis response protein 4 (Par-4), a tumor suppressor, has been shown to induce apoptosis in cancer cells. While reduced Par-4 expression has been linked to survival of some cancers, its involvement in colon cancer has not been well documented. To explore the feasibility of increasing Par-4 in colon cancer to induce apoptosis, the human colon cancer cell line, HT29, was transfected to overexpress Par-4. In these cells, overexpressed Par-4 led to increased apoptosis in the presence of 5-fluorouracil. Subsequently, PAR-4 cDNA was packaged in nanoliposomal particles. Treating cells with the Par-4 nanoliposomes also increased susceptibility to 5-FU. These nanoliposomes were used to deliver Par-4 plasmid to tumors growing in nude mice from wild type HT29 cells. Results showed that nanoliposomes effectively delivered plasmid DNA to tumors in vivo. Again, tumors in mice treated with the Par-4 nanoliposomes were more susceptible to 5-FU treatment. This suggests that upregulation of Par-4 expression is a potentially useful mechanism to enhance the current chemotherapeutic regimen for colon cancer. Packaging Par-4 cDNA in nanoliposomal particles is a promising delivery method to increase response to chemotherapy.
- Published
- 2009
- Full Text
- View/download PDF
12. Molecular profiling of LGL leukemia reveals role of sphingolipid signaling in survival of cytotoxic lymphocytes
- Author
-
Ranran Zhang, Ravi Kothapalli, Rosalyn B. Irby, Mithun Vinod Shah, Ty Arrington, Thomas P. Loughran, Xin Liu, Bryan Frank, and Norman H. Lee
- Subjects
Programmed cell death ,Cell Survival ,Lymphocyte ,CD3 ,Immunology ,Apoptosis ,Biology ,Biochemistry ,medicine ,Humans ,Cytotoxic T cell ,Sphingolipids ,Neoplasia ,Gene Expression Profiling ,Cell Biology ,Hematology ,medicine.disease ,Cell biology ,Gene Expression Regulation, Neoplastic ,Leukemia, Large Granular Lymphocytic ,Receptors, Lysosphingolipid ,Leukemia ,medicine.anatomical_structure ,Case-Control Studies ,Galactosylgalactosylglucosylceramidase ,biology.protein ,Signal transduction ,CD8 ,Signal Transduction ,T-Lymphocytes, Cytotoxic - Abstract
T-cell large granular lymphocyte (LGL) leukemia is characterized by clonal expansion of CD3+CD8+ cells. Leukemic LGLs correspond to terminally differentiated effector-memory cytotoxic T lymphocytes (CTLs) that escape Fas-mediated activation-induced cell death (AICD) in vivo. The gene expression signature of peripheral blood mononuclear cells from 30 LGL leukemia patients showed profound dysregulation of expression of apoptotic genes and suggested uncoupling of activation and apoptotic pathways as a mechanism for failure of AICD in leukemic LGLs. Pathway-based microarray analysis indicated that balance of proapoptotic and antiapoptotic sphingolipid-mediated signaling was deregulated in leukemic LGLs. We further investigated sphingolipid pathways and found that acid ceramidase was constitutively overexpressed in leukemic LGLs and that its inhibition induced apoptosis of leukemic LGLs. We also showed that S1P5 is the predominant S1P receptor in leukemic LGLs, whereas S1P1 is down-regulated. FTY720, a functional antagonist of S1P-mediated signaling, induced apoptosis in leukemic LGLs and also sensitized leukemic LGLs to Fas-mediated death. Collectively, these results show a role for sphingolipid-mediated signaling as a mechanism for long-term survival of CTLs. Therapeutic targeting of this pathway, such as use of FTY720, may have efficacy in LGL leukemia.
- Published
- 2008
- Full Text
- View/download PDF
13. Role of Stat3 in Regulating p53 Expression and Function
- Author
-
Rosalyn B. Irby, Gabriela Wright, James G. Karras, Guilian Niu, Drew M. Pardoll, Yihong Ma, Jon Briggs, W. Douglas Cress, Hua Yu, Richard Jove, Kenneth L. Wright, Mei Huang, and Jiangdong Chen
- Subjects
STAT3 Transcription Factor ,Transcription, Genetic ,Gene Expression ,Apoptosis ,Electrophoretic Mobility Shift Assay ,Biology ,Response Elements ,medicine.disease_cause ,Mice ,Neoplasms ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,STAT3 ,Molecular Biology ,Gene ,Cell Proliferation ,Regulation of gene expression ,Mutation ,Cell growth ,Cancer ,Cell Biology ,medicine.disease ,DNA-Binding Proteins ,Gene Expression Regulation ,Cancer cell ,Trans-Activators ,Cancer research ,biology.protein ,Tumor Suppressor Protein p53 ,Signal transduction ,Signal Transduction - Abstract
Loss of p53 function by mutation is common in cancer. However, most natural p53 mutations occur at a late stage in tumor development, and many clinically detectable cancers have reduced p53 expression but no p53 mutations. It remains to be fully determined what mechanisms disable p53 during malignant initiation and in cancers without mutations that directly affect p53. We show here that oncogenic signaling pathways inhibit the p53 gene transcription rate through a mechanism involving Stat3, which binds to the p53 promoter in vitro and in vivo. Site-specific mutation of a Stat3 DNA-binding site in the p53 promoter partially abrogates Stat3-induced inhibition. Stat3 activity also influences p53 response genes and affects UV-induced cell growth arrest in normal cells. Furthermore, blocking Stat3 in cancer cells up-regulates expression of p53, leading to p53-mediated tumor cell apoptosis. As a point of convergence for many oncogenic signaling pathways, Stat3 is constitutively activated at high frequency in a wide diversity of cancers and is a promising molecular target for cancer therapy. Thus, repression of p53 expression by Stat3 is likely to have an important role in development of tumors, and targeting Stat3 represents a novel therapeutic approach for p53 reactivation in many cancers lacking p53 mutations.
- Published
- 2005
- Full Text
- View/download PDF
14. Osteopontin regulates multiple functions contributing to human colon cancer development and progression
- Author
-
Susan McCarthy, Rosalyn B. Irby, and Timothy J. Yeatman
- Subjects
Cancer Research ,Colorectal cancer ,Sialoglycoproteins ,Integrin ,Mice, Nude ,Metastasis ,Mice ,stomatognathic system ,Cell Movement ,Cell Adhesion ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Osteopontin ,Cell adhesion ,Cell Proliferation ,Regulation of gene expression ,Neovascularization, Pathologic ,biology ,Microcirculation ,CD44 ,General Medicine ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Platelet Endothelial Cell Adhesion Molecule-1 ,Hyaluronan Receptors ,Oncology ,Colonic Neoplasms ,Cancer cell ,Immunology ,Disease Progression ,biology.protein ,Cancer research - Abstract
Osteopontin (OPN) is a secreted phosphoglycoprotein known to interact with a number of integrin receptors. While increased OPN expression has been reported in a number of human cancers, and its cognate receptors (alphav-beta3, alphav-beta5, and alphav-beta1 integrins and CD44) have been identified, its role in colon cancer development and progression has not been extensively studied. We previously identified, using a combination of gene expression and tissue microarrays, that increased OPN expression is concordant with tumor stage. The current study examined the functional role of OPN in colon cancer progression and metastatic potential. The principal findings of this study were that both endogenous OPN expression (via stable transfection) as well as exogenous OPN (added to culture medium) enhanced the motility and invasive capacity of human colon cancer cells in vitro. OPN appeared to regulate motility though interaction with CD44. OPN expression also reduced intercellular (homotypic) adhesion, an important characteristic of metastatic cancer cells. Stable transfection of four poorly tumorigenic human colon cancer cell lines with OPN also resulted in enhanced tumorigenicity in vivo with increased proliferation and increased CD31 positive microvessel counts, concordant with the degree of OPN expression. Collectively, these results suggest that OPN may affect multiple functional components contributing to human colon cancer progression and solidifies its role in this process.
- Published
- 2004
- Full Text
- View/download PDF
15. Peroxisome proliferator-activated receptor α induces rat sterol carrier protein x promoter activity through two peroxisome proliferator-response elements
- Author
-
Rosalyn B. Irby, Dayami Lopez, and Mark P. McLean
- Subjects
Transcriptional Activation ,Protein Conformation ,Receptors, Retinoic Acid ,Molecular Sequence Data ,Receptors, Cytoplasmic and Nuclear ,Peroxisome proliferator-activated receptor ,Biology ,Retinoid X receptor ,Response Elements ,Biochemistry ,Endocrinology ,Genes, Reporter ,Transcription (biology) ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Clofibrate ,Acetyl-CoA C-Acetyltransferase ,Promoter Regions, Genetic ,Receptor ,Molecular Biology ,Gene ,chemistry.chemical_classification ,Binding Sites ,Base Sequence ,Fatty acid ,Peroxisome ,Molecular biology ,Rats ,Retinoid X Receptors ,chemistry ,Carrier Proteins ,Transcription Factors ,medicine.drug - Abstract
Sterol carrier protein x (SCPx) plays a critical role in the peroxisomal oxidation of fatty acids. It has been previously demonstrated in streptozotocin-induced diabetic rats that SCPx expression is induced in association with an elevation in serum fatty acid and triglyceride levels. To elucidate the mechanisms underlying the expression of this gene during diabetes, the rat SCPx promoter was cloned and analyzed for regulatory motifs. Sequence analysis of this TATA-less promoter revealed two putative peroxisomal-proliferator-response element (PPRE) binding motifs at positions -134 and -869 relative to the translation start site. To examine peroxisomal-proliferator-activated receptor alpha (PPARalpha) effects on this gene, 935 bp of the SCPx promoter containing both PPRE motifs was cloned in front of the chloramphenicol acetyl-transferase gene or the luciferase gene and co-transfected into HTB-9 cells with vectors that encoded for PPARalpha and retinoid X receptor (RXR). The results indicate that PPARalpha was able to induce SCPx promoter activity in both cases, an effect that was enhanced by RXR and clofibrate. In addition, mutational analysis studies demonstrated that both PPREs contributed to the PPARalpha/RXRalpha-dependent activation of the SCPx promoter. Mobility shift assays and supershift analysis showed that nuclear extracts containing PPARalpha bound to the two PPRE motifs. This investigation indicates that similar to other genes involved in beta-oxidation, SCPx transcription may be controlled by fatty acid levels via PPARalpha.
- Published
- 2003
- Full Text
- View/download PDF
16. Osteopontin Identified as Lead Marker of Colon Cancer Progression, Using Pooled Sample Expression Profiling
- Author
-
Ann F. Chambers, Marianna Szabo, Deepak Agrawal, Timothy J. Yeatman, Tingan Chen, Domenico Coppola, Alan B. Cantor, John Quackenbush, and Rosalyn B. Irby
- Subjects
Adenoma ,Cancer Research ,Pathology ,medicine.medical_specialty ,Colorectal cancer ,Sialoglycoproteins ,Gene Expression ,Biology ,Metastasis ,Immunoenzyme Techniques ,Biomarkers, Tumor ,medicine ,Humans ,RNA, Messenger ,Northern blot ,Osteopontin ,Neoplasm Staging ,Oligonucleotide Array Sequence Analysis ,Tumor marker ,Gene Expression Profiling ,Cancer ,Blotting, Northern ,medicine.disease ,Neoplasm Proteins ,Gene expression profiling ,Oncology ,Tumor progression ,Colonic Neoplasms ,Disease Progression ,Cancer research ,biology.protein - Abstract
Background: New tumor markers and markers of tumor progression are needed for improved staging and for better assessment of treatment of many cancers. Gene expression profiling techniques offer the opportunity to discover such markers. We investigated the feasibility of sample pooling strategy in combination with a novel analysis algorithm to identify markers. Methods: Total RNA from human colon tumors (n = 60) of multiple stages (adenomas; cancers with modified Astler Collier stages B, C, and D; and liver metastases) were pooled within stages and compared with pooled normal mucosal specimens (n = 10) by using oligonucleotide expression arrays. Genes that showed consistent increases or decreases in their expression through tumor progression were identified. Northern blot analysis was used to validate the findings. All statistical tests were two-sided. Results: More than 300 candidate tumor markers and more than 100 markers of tumor progression were identified. Northern analysis of 11 candidate tumor markers confirmed the gene expression changes. The gene for the secreted integrinbinding protein osteopontin was most consistently differentially expressed in conjunction with tumor progression. Its potential as a progression marker was validated (Spearman’s = 0.903; P
- Published
- 2002
- Full Text
- View/download PDF
17. Par-4: how far will it go?
- Author
-
Rosalyn B. Irby
- Subjects
education.field_of_study ,Chemotherapy ,biology ,business.industry ,medicine.medical_treatment ,Population ,Cancer ,General Medicine ,medicine.disease ,Radiation therapy ,Cancer cell ,medicine ,Adjuvant therapy ,Cancer research ,biology.protein ,Occult cancer ,Antibody ,business ,education - Abstract
The mainstay of cancer treatment typically includes surgery, chemotherapy, and radiation. If cancer is confined to one area, surgery alone can be curative, although it is often combined with adjuvant therapy (1). Radiation is limited to treatment of cells within the scope of the radiation beam, although radio immunotherapy has enhanced radiation therapy particularly in B-cell lymphomas (2). Chemotherapy has a number of forms, including systemically toxic drugs and both drugs and antibodies targeted to specific proteins that are driving the growth of the cancer cells. Chemotherapy can be used to treat systemically both visible and occult cancer cells. However, therapy resistance in tumors is a continuing problem for patients and oncologists alike despite an aggressive treatment regimen. In some cases, early mutations give resistance to tumors, or to a sub-population of tumor cells within a heterogeneous tumor, even prior to treatment. In this case treatment can cause death to sensitive cells allowing the few resistant cells to become the dominant population and lead to tumor recurrence. On the other hand, tumors that initially respond to treatment often become resistant over time as new mutations develop that allow cells to evade death. Such resistance, which may lead to multi-drug resistance, results in tumor recurrence and, ultimately, death (3).
- Published
- 2017
- Full Text
- View/download PDF
18. Abstract 1166: Identification of a novel quinoxaline-isoselenourea targeting STAT3 pathway as a potential melanoma therapeutic
- Author
-
Manoj Pandey, Verónica Alcolea, Carmen Sanmartín, Deepkamal N. Karelia, Daniel Plano, Arun K. Sharma, Rosalyn B. Irby, Shantu Amin, Juan Antonio Palop, and Parvesh Singh
- Subjects
Cancer Research ,biology ,business.industry ,Melanoma ,medicine.disease ,chemistry.chemical_compound ,Quinoxaline ,Oncology ,chemistry ,Cancer research ,biology.protein ,medicine ,Identification (biology) ,STAT3 ,business - Abstract
The prognosis for patients with metastatic melanoma remains very poor. Constitutive STAT3 activation has been correlated to larger tumor size, metastasis, acquired resistance against vemurafenib (PLX-4032), and poor patient survival suggesting its potential as a molecular target. We recently designed a series of isoseleno- and isothio-urea derivatives of several biologically active heterocyclic scaffolds. The cytotoxic effects of lead isoseleno-/isothio-urea derivatives (compounds 1/3) were studied in a panel of five melanoma cell lines, including B-RAFV600E mutant and wild type (WT) cells. Compound 1 (IC50 range 0.8-3.8 µM) showed lower IC50 values than 3 (IC50 range 8.1-38.7 µM) and the mutant B-RAF specific inhibitor PLX-4032 (IC50 range 0.4->50 µM), especially at shorter treatment time (24 h). These effects are long-lasting since melanoma cells did not recover their proliferative potential after 14 days of the treatment. In addition, we confirmed that compound 1 induces cell death by apoptosis using Live and Dead, Annexin V and Caspase3/7 apoptosis assays. Furthermore, compound 1 reduces protein levels of STAT3 and its phosphorylation, as well as decreases the expression of STAT3 regulated genes involved in survival and metastasis such as survivin and c-myc. Compound 1 also upregulates the cell cycle inhibitor p21. Docking studies further revealed the favorable binding of 1 with SH2 domain of STAT3 suggesting its activity through STAT3 inhibition. Taken together, our results suggest that compound 1 induces apoptosis by means of the inhibition of STAT3 pathway to non-specifically target both B-RAF mutant and WT melanoma cells with much better cytotoxicity than the current therapy PLX-4032. Citation Format: Verónica Alcolea, Deepkamal Karelia, Manoj K. Pandey, Daniel Plano, Parvesh Singh, Rosalyn B. Irby, Juan Palop, Shantu Amin, Carmen Sanmartín, Arun K. Sharma. Identification of a novel quinoxaline-isoselenourea targeting STAT3 pathway as a potential melanoma therapeutic [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1166. doi:10.1158/1538-7445.AM2017-1166
- Published
- 2017
- Full Text
- View/download PDF
19. v-Src Transformation Is Mediated through Farnesylated Proteins
- Author
-
Jiazhi Sun, Steven Teng, Timothy J. Yeatman, Andrew D. Hamilton, Said M. Sebti, and Rosalyn B. Irby
- Subjects
Biology ,Transfection ,medicine.disease_cause ,Cell Line ,Oncogene Protein pp60(v-src) ,Prenylation ,Western blot ,medicine ,Animals ,Farnesyltranstransferase ,Enzyme Inhibitors ,Alkyl and Aryl Transferases ,medicine.diagnostic_test ,rap1 GTP-Binding Proteins ,Fibroblasts ,Molecular biology ,Rats ,Blot ,Transformation (genetics) ,Cell Transformation, Neoplastic ,Cell culture ,v-Src ,ras Proteins ,Surgery ,Carcinogenesis ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Src is an oncoprotein which has been implicated in a number of human malignancies in which it has been shown to be overexpressed and highly activated. The precise mechanism of Src transformation, however, is still poorly understood. We hypothesized that Ras and other farnesylated proteins may mediate Src transformation. To test this hypothesis, v-Src-transfected rat fibroblasts (3Y1) were treated every 72 h with a 15 μM concentration of a farnesyl-transferase inhibitor (FTI). At 2 weeks, a focus formation assay was performed to assess transformation potential. Untreated and FTI-treated v-Src-transfected 3Y1 cells formed a mean of 39 (±2.6) and 29.8 (±2.9) foci per well, respectively. This 24% decrease was judged to be statistically significant ( P = 0.02). Moreover, foci (>90%) in the FTI-treated wells were also consistently smaller than foci in the untreated wells. Western blots with antibody directed toward H-Ras confirmed complete inhibition of Ras farnesylation in the treated cell lines. The specificity of this inhibition was verified by Western blot using antibody specific for Rap1A. The transforming potential of v-Src is inhibited, but not eliminated by FTI treatment. This suggests that v-Src transformation is mediated in part by farnesylated proteins, one of which may be Ras.
- Published
- 2001
- Full Text
- View/download PDF
20. Activating SRC mutation in a subset of advanced human colon cancers
- Author
-
Timothy J. Yeatman, Rosalyn B. Irby, Jean Marc Loubeau, Jimmy J. Kang, Richard C. Karl, Richard Jove, Walter L. Trudeau, Donald J. Fujita, Domenico Coppola, and Weiguang Mao
- Subjects
Lung Neoplasms ,Proto-Oncogene Proteins pp60(c-src) ,Mice, Nude ,Biology ,Transfection ,medicine.disease_cause ,Polymerase Chain Reaction ,Cell Line ,Metastasis ,Mice ,Genetics ,medicine ,Animals ,Humans ,Kinase activity ,Mice, Inbred BALB C ,Mutation ,Rous sarcoma virus ,Liver Neoplasms ,3T3 Cells ,medicine.disease ,biology.organism_classification ,Rats ,Gene Expression Regulation, Neoplastic ,Genes, src ,Tumor progression ,Colonic Neoplasms ,Cancer research ,Phosphorylation ,Tyrosine kinase ,Neoplasm Transplantation ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The discovery of Rous sarcoma virus (RSV) led to the identification of cellular Src (c–Src), a non-receptor tyrosine kinase, which has since been implicated in the development of numerous human cancers1,2,3,4. c-Src has been found to be highly activated in colon cancers, particularly in those metastatic to the liver5,6,7,8,9,10,11. Studies of the mechanism of c-Src regulation have suggested that c-Src kinase activity is downregulated by phosphorylation of a critical carboxy-terminal tyrosine (Tyr 530 in human c-Src, equivalent to Tyr 527 in chicken Src) and have implied the existence of activating mutations in this C-terminal regulatory region12,13,14,15,16,17,18. We report here the identification of a truncating mutation in SRC at codon 531 in 12% of cases of advanced human colon cancer tested and demonstrate that the mutation is activating, transforming, tumorigenic and promotes metastasis. These results provide, for the first time, genetic evidence that activating SRC mutations may have a role in the malignant progression of human colon cancer.
- Published
- 1999
- Full Text
- View/download PDF
21. Intra-cellular tyrosine kinase
- Author
-
Timothy J. Yeatman and Rosalyn B. Irby
- Subjects
Dasatinib ,ABL ,Chemistry ,medicine ,Cancer research ,Phosphorylation ,Imatinib ,Signal transduction ,Molecular oncology ,Tyrosine kinase ,medicine.drug ,Proto-oncogene tyrosine-protein kinase Src - Published
- 2013
- Full Text
- View/download PDF
22. Correction: The Akt Inhibitor ISC-4 Synergizes with Cetuximab in 5-FU-Resistant Colon Cancer
- Author
-
Wafik S. El-Deiry, Arun K. Sharma, Joshua E. Allen, David T. Dicker, Jean-Nicolas Gallant, Rosalyn B. Irby, and Shantu Amin
- Subjects
Multidisciplinary ,Cetuximab ,Colorectal cancer ,business.industry ,Science ,lcsh:R ,Correction ,lcsh:Medicine ,Akt inhibitor ,medicine.disease ,Bioinformatics ,medicine ,Cancer research ,Medicine ,lcsh:Q ,business ,lcsh:Science ,medicine.drug - Abstract
There is an error in the title of Supplemental Table S2. The table describes the combinatorial effects of ISC-4, not TIC10. The correct title reads: Summary of combinatorial effects of ISC-4 with approved antitumor agents.
- Published
- 2013
23. Altered Ovarian Sterol Carrier Protein Expression in the Pregnant Streptozotocin-Treated Diabetic Rat1
- Author
-
Dale B. Hales, Kim J. Warden, Mark P. McLean, Todd W. Sandhoff, and Rosalyn B. Irby
- Subjects
endocrine system ,medicine.medical_specialty ,Fatty acid metabolism ,Triglyceride ,Cholesterol ,Cholesterol side-chain cleavage enzyme ,medicine.medical_treatment ,Cell Biology ,General Medicine ,Biology ,medicine.disease ,Streptozotocin ,chemistry.chemical_compound ,Steroid hormone ,medicine.anatomical_structure ,Endocrinology ,Reproductive Medicine ,chemistry ,Diabetes mellitus ,Internal medicine ,medicine ,Corpus luteum ,medicine.drug - Abstract
Reproductive dysfunction in the diabetic female rat is associated with impaired folliculogenesis, reduced corpus luteum progesterone output, and spontaneous abortion. The underlying mechanism for reduced steroid production remains unresolved. In this study we examined whether or not diabetes alters levels of P450 side-chain cleavage enzyme (P450scc), 3 beta-hydroxysteroid dehydrogenase (3 beta-HSD), or the cholesterol transport proteins, steroidogenic acute regulatory (StAR) protein and sterol carrier protein-2 (SCP2), leading to lower progesterone levels and pregnancy loss. Rats (Day 3 pregnant) received an injection of streptozotocin (STZ, 60 mg/kg; i.v.) to induce a diabetic state; P450scc, 3 beta-HSD, and SCP2 were examined by Western and Northern blot analysis in ovarian tissue 12 days after injection of STZ (diabetic rats, n = 12) or vehicle (nondiabetic rats, n = 12). Serum progesterone, triglyceride, and beta-hydroxybutyrate (beta-HBA) levels were also examined. Results indicate that diabetic rats that aborted (diabetic-fetus [Ft], n = 6) had significantly lower progesterone levels (7.04 +/- 2.6 ng/ml; p < 0.004) than nondiabetic animals (108.6 +/- 5.15 ng/ml) and diabetic +Ft animals (74.3 +/- 8.9 ng/ml, n = 6). Western blot analysis of ovarian P450scc and 3 beta-HSD in the nondiabetic rats and the diabetic rats with fetuses indicated no significant difference. In contrast, ovaries from diabetic animals without fetuses had significantly lower SCP2 levels (p < 0.017) compared to controls. Concomitant with the reduction in SCP2, a 58-kDa SCP2-immunoreactive protein, referred to as sterol carrier protein-X (SCPx), increased significantly (p < 0.001). The C-terminal sequence of SCPx is identical to SCP2, while its N-terminal region is homologous with 3-oxoacyl coenzyme A thiolase, an enzyme involved in fatty acid metabolism. Increased SCPx expression coincided with increased serum triglyceride and beta-HBA levels, suggesting that the enhanced SCPx level may coincide with an ovarian shift to fatty acid metabolism. When SCPx steady-state mRNA levels were measured using an SCPx-specific riboprobe (280-bp protected fragment) in a ribonuclease protection assay, ovarian SCPx mRNA levels in the diabetic animals were increased 4.2-fold compared to control SCPx mRNA levels. Ovarian StAR mRNA levels were increased slightly in the diabetic animals, and ovarian P450scc and 3 beta-HSD mRNA levels were increased 3-fold in the diabetic animals that aborted relative to the nondiabetic animals and the +Ft diabetic animals. Results of this study confirm that SCPx mRNA levels are elevated following diabetes onset and that StAR, P450scc, and 3 beta-HSD mRNA levels do not correspond with the reduced steroid hormone profile associated with diabetes. These results are concordant with the possibility that reduced steroid levels in the diabetic animals reflect a loss of SCP2-mediated cholesterol transport capacity as SCPx/3-oxoacyl coenzyme A thiolase expression is enhanced.
- Published
- 1996
- Full Text
- View/download PDF
24. Prostaglandin F2 alpha mediates ovarian sterol carrier protein-2 expression during luteolysis
- Author
-
J T Billheimer, Rosalyn B. Irby, Mark P. McLean, and K J Warden
- Subjects
endocrine system ,medicine.medical_specialty ,3-Hydroxysteroid Dehydrogenases ,Blotting, Western ,Molecular Sequence Data ,Gene Expression ,Biology ,Dinoprost ,Rats, Sprague-Dawley ,Endocrinology ,Western blot ,Internal medicine ,Luteolysis ,medicine ,Animals ,Cholesterol Side-Chain Cleavage Enzyme ,RNA, Messenger ,Northern blot ,Progesterone ,Plant Proteins ,Messenger RNA ,Base Sequence ,medicine.diagnostic_test ,Cholesterol side-chain cleavage enzyme ,Ovary ,Nuclease protection assay ,Blotting, Northern ,Rats ,Blot ,medicine.anatomical_structure ,Female ,Carrier Proteins ,Corpus luteum - Abstract
In the corpus luteum, prostaglandin F2 alpha (PGF2 alpha) appears to be a physiological agent with both antisteroidogenic and luteolytic actions. It is hypothesized that the antisteroidogenic action of PGF2 alpha acts through altered transport of cholesterol to the mitochondrial cytochrome P450 side-chain cleavage enzyme (P450scc). However, the effect of PGF2 alpha on the expression of the putative cholesterol transport protein, sterol carrier protein-2 (SCP2; 13.2 kilodaltons), has not been examined. In this study, the decline in serum progesterone after PGF2 alpha injection was examined in parallel with altered ovarian SCP2, P450scc, and 3 beta-hydroxysteroid dehydrogenase (3 beta HSD) protein and messenger RNA (mRNA) levels. Rats (28 days old) were treated with 8 IU PMSG to induce follicular development and ovulation. Ten days after ovulation, animals were treated with PGF2 alpha (single or multiple injections; 100-250 micrograms each) or left untreated. Ovarian SCP2, P450scc, and 3 beta HSD protein and mRNA levels were examined 0 (time zero), 4, and 8 h post-PGF2 alpha treatment using Western and Northern blot analysis. SCP2 mRNA levels were also examined using a highly sensitive ribonuclease protection assay that detects a 429-base pair SCP2-mRNA specific sequence. The results indicate that serum progesterone was significantly reduced 4 and 8 h after PGF2 alpha injections (P < 0.001; n = 6/time point). The decline in progesterone paralleled a 50-60% reduction in 3 beta HSD protein and mRNA levels by 4 h post-PGF2 alpha. Protein and mRNA levels for 3 beta HSD returned to control values by 8 h post-PGF2 alpha treatment. P450scc expression was also reduced at 4 h (44-54%), but by 8 h, both protein and mRNA levels had increased above the normal control levels (P < 0.02). In contrast, the 0.8-kilobase SCP2-specific mRNA transcript was reduced to 50% and 80% of the pre-PGF2 alpha treatment level at 4 and 8 h, respectively (P < 0.01). SCP2 ribonuclease protection assay analysis also indicated that SCP2 mRNA levels were reduced 65% (P < 0.03) and 85% (P < 0.01) by 4 and 8 h post-PGF2 alpha treatment compared to those in time zero ovarian tissue. Consistent with the loss of SCP2 mRNA expression, Western blot analysis indicated that a 15-kilodalton SCP2-immunoreactive protein (presumably the pro-SCP2 form) was significantly reduced or absent in the PGF2 alpha treated animals (P < 0.04).(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1995
- Full Text
- View/download PDF
25. Differential expression of hepatic sterol carrier proteins in the streptozotocin-treated diabetic rat
- Author
-
K J Warden, Mark P. McLean, Rosalyn B. Irby, and J T Billheimer
- Subjects
Male ,medicine.medical_specialty ,Time Factors ,Sterol O-acyltransferase ,Biology ,Intracellular cholesterol transport ,Cholesterol 7 alpha-hydroxylase ,Diabetes Mellitus, Experimental ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Endocrinology ,Internal medicine ,medicine ,Animals ,Insulin ,RNA, Messenger ,Acetyl-CoA C-Acetyltransferase ,Plant Proteins ,SCP2 ,Cholesterol ,Reverse cholesterol transport ,Sterol ,Rats ,Sterols ,Sterol carrier protein ,Liver ,chemistry ,Carrier Proteins - Abstract
Sterol carrier protein-2 (SCP2) is a 13.2-kilodalton protein that has been implicated in intracellular cholesterol transport, whereas a related sterol carrier protein, sterol carrier protein-X (SCPx; 58 kilodaltons) has been suggested to function also in the beta-oxidation of fatty acids. Although diabetes-related hyperlipidemia and altered cholesterol metabolism have been extensively studied, the intracellular cholesterol transport capacity during hyperglycemic states has not been examined. The fact that beta-oxidation is increased in diabetes whereas hepatic cholesterol metabolism is reduced suggests that differential expression of these sterol carrier proteins may accompany diabetic dyslipidemia. In this study, SCP2 protein levels were reduced by 60% in mildly hypercholesterolemic (cholesterol,130 and150 mg/dl; P0.01) diabetic rats and by 90% in severely hypercholesterolemic (cholesterol,150 mg/dl; P0.002) diabetic animals. In contrast, hepatic SCPx protein expression increased (3.5-fold) after diabetes induction with streptozotocin (STZ). The decline in SCP2 was inversely related to serum cholesterol levels. Hepatic SCP messenger RNA levels examined by ribonuclease protection assay demonstrated that hepatic SCP messenger RNA was increased 2-fold in diabetic animals. Northern blot analysis indicated that both the 0.8-kilobase SCP2-specific and the 2.1-kilobase SCPx-specific transcripts increased after STZ injection. SCPx protein induction preceded the decline in SCP2 by 4-5 days. Insulin treatment reversed the increase in SCPx and prevented the decline in SCP2. We conclude that SCP2 and SCPx are differentially expressed in the STZ-diabetic rat and suggest that this change in SCP expression should be considered a potential contributing mechanism through which cholesterol metabolism may be altered in diabetes.
- Published
- 1995
- Full Text
- View/download PDF
26. Reduced hepatic sterol carrier protein-2 expression in the streptozotocin treated diabetic rat
- Author
-
K J Warden, J. T. Billheimer, K. Nanjo, Rosalyn B. Irby, and Mark P. McLean
- Subjects
medicine.medical_specialty ,Cholesterol ,Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,Biology ,medicine.disease ,Streptozotocin ,Sterol ,chemistry.chemical_compound ,Endocrinology ,Sterol carrier protein ,chemistry ,Diabetes mellitus ,Internal medicine ,medicine ,Northern blot ,SCP2 ,medicine.drug - Abstract
While a strong relationship between the hypercholesterolemia of diabetes and premature atherosclerosis is established, the etiology for the elevation in serum cholesterol in this disease is unknown. To determine whether diabetic hypercholesterolemia may be related to alterations in hepatic cholesterol transport capacity, sterol carrier protein-2 (SCP2) expression was examined in rats treated with streptozotocin (SZT). Furthermore, this study examined whether 17β-estradiol and insulin confer a protective effect on liver cholesterol homeostasis by maintaining hepatic SCP2 levels. SCP2 protein and mRNA expression were examined 13 days following SZT-induced diabetes onset and in diabetic rats treated with estradiol (1 cm silastic implant) or insulin (12 units/day). Data indicate that SCP2 protein levels were significantly reduced in the diabetic animals and that SCP2 protein expression in the liver was inversely related to the level of serum cholesterol in the diabetic animals. In contrast, SCP2 mRNA levels examined by slot blot, ribonuclease protection assay, and Northern blot analysis were significantly elevated. Both insulin and estradiol were able to enhance the expression of SCP2 protein in the liver following SZT treatment. The results of this investigation clearly indicate that hepatic SCP2 protein levels are significantly altered in the diabetic state suggesting that cholesterol transport capacity is reduced in the SZT-treated diabetic rat. The inverse relationship between serum cholesterol and hepatic SCP2 protein content suggests that the reduction in this protein may be a contributing factor in diabetic hypercholesterolemia.
- Published
- 1995
- Full Text
- View/download PDF
27. Par-4 as a potential target for cancer therapy
- Author
-
Rosalyn B. Irby and Christina Leah B. Kline
- Subjects
Pharmacology ,Programmed cell death ,business.industry ,Clinical Biochemistry ,Cancer ,Apoptosis ,Suicide gene ,medicine.disease ,Metastasis ,In vivo ,Neoplasms ,Drug Discovery ,Cancer cell ,Immunology ,medicine ,Cancer research ,Molecular Medicine ,Animals ,Humans ,business ,Apoptosis Regulatory Proteins ,Cause of death - Abstract
Despite extensive research, cancer continues to be a leading cause of death worldwide and is expected to continue to rise as a result of an aging population. Therefore, new therapies are constantly being developed. Par-4 is a naturally occurring tumor suppressor protein that is capable of inducing apoptosis in cancer, but not normal cells. For this reason, Par-4 offers an attractive target for development of cancer therapy, particularly of difficult to treat cancers.The mechanisms by which Par-4 induces cell death are summarized. The ways that Par-4 is controlled in cancer cells are discussed. We discuss how different research groups have developed ways to overexpress and/or activate Par-4 in vitro and in vivo. The studies described demonstrate that when Par-4 levels and/or activity are increased, susceptibility to apoptosis is enhanced and tumor growth is inhibited.Par-4 is a promising therapeutic protein that can be overexpressed and/or activated to induce apoptosis in a cancer-selective manner. This cancer selectivity is important given that the side-effects of chemotherapeutics can be as debilitating as cancer itself. However, there are key issues that need to be addressed to optimize the effects of Par-4 in patients.
- Published
- 2012
28. Intermediates in the folic acid biosynthetic pathway are incorporated into molybdopterin the yeast, Pichia canadensis
- Author
-
Rosalyn B. Irby and W L Adair
- Subjects
Stereochemistry ,Molybdopterin ,Wild type ,Guanosine ,Cell Biology ,Biology ,Biochemistry ,Yeast ,Cofactor ,chemistry.chemical_compound ,chemistry ,Biosynthesis ,Glyceraldehyde ,Ribose ,biology.protein ,Molecular Biology - Abstract
Biosynthesis of molybdopterin was followed in the yeast, Pichia canadensis, using labeled precursors. High performance liquid chromatography analysis of extracts from cells labeled with [U-14C]guanosine showed that the label was incorporated into the molybdopterin oxidation product, dephospho Form A. Dephospho Form A isolated from cells labeled with [U-14C,5'-3H]guanosine was devoid of tritium, indicating partial loss of the ribose moiety of guanosine during the synthesis of molybdopterin. In vivo labeling of P. canadensis using [7-14C]neopterin and [6,7,1-14C]hydroxymethylpterin led to label from both compounds appearing in dephospho Form A as well as in folic acid in wild type cells. When these labeled precursors were incubated with P. canadensis mutants blocked in molybdopterin synthesis, only folic acid was labeled. These results suggest a shared pathway in the biosyntheses of molybdopterin and folic acid. [6-14C]Glucose labeling experiments led to exclusive incorporation into the 4'-position of dephospho Form A but not in folic acid. It is proposed that molybdopterin synthesis branches from the folic acid biosynthetic pathway at dihydrohydroxymethylpterin and that a 3-carbon phosphorylated compound such as glyceraldehyde 3-phosphate may condense with dihydrohydroxymethylpterin to form the 4-carbon side chain precursor to molybdopterin.
- Published
- 1994
- Full Text
- View/download PDF
29. Seroreactivity to LGL leukemia-specific epitopes in aplastic anemia, myelodysplastic syndrome and paroxysmal nocturnal hemoglobinuria: results of a bone marrow failure consortium study
- Author
-
Nancy Ruth Jarbadan, Ronald Paquette, Michael J. Clemente, Jason Liao, Kendall Thomas Baab, Pearlie K. Epling-Burnette, Lubomir Sokol, Daniel J. Krissinger, Thomas P. Loughran, Eric Schaefer, Jaroslaw P. Maciejewski, Rosalyn B. Irby, Alan F. List, Susan B. Nyland, and David Cuthbertson
- Subjects
Cancer Research ,Molecular Sequence Data ,Hemoglobinuria, Paroxysmal ,chemical and pharmacologic phenomena ,Enzyme-Linked Immunosorbent Assay ,Epitope ,Article ,hemic and lymphatic diseases ,medicine ,Cytotoxic T cell ,Humans ,Amino Acid Sequence ,Aplastic anemia ,business.industry ,Bone marrow failure ,Anemia, Aplastic ,Hematology ,medicine.disease ,Leukemia, Large Granular Lymphocytic ,CTL ,Leukemia ,Oncology ,Myelodysplastic Syndromes ,Immunology ,Paroxysmal nocturnal hemoglobinuria ,Epitopes, B-Lymphocyte ,Hemoglobinuria ,business - Abstract
Large granular lymphocyte (LGL) leukemia is characterized by clonal expansion of antigen-activated cytotoxic T cells (CTL). Patients frequently exhibit seroreactivity against a human T-cell leukemia virus (HTLV) epitope, BA21. Aplastic anemia, paroxysmal nocturnal hemoglobinuria and myelodysplastic syndrome are bone marrow failure diseases that can also be associated with similar aberrant CTL activation (LGL-BMF). We identified a BA21 peptide that was specifically reactive with LGL leukemia sera and found significantly elevated antibody reactivity against the same peptide in LGL-BMF sera. This finding of shared seroreactivity in LGL-BMF conditions and LGL leukemia suggests that these diseases might share a common pathogenesis.
- Published
- 2011
30. Synthesis and biological evaluation of a novel class of isatin analogs as dual inhibitors of tubulin polymerization and Akt pathway
- Author
-
Arun Sharma, Gowdahalli Krishnegowda, Kevin F. Staveley-O’ Carroll, Rosalyn B. Irby, Shantu Amin, Hephzibah Rani S. Tagaram, and A. S. Prakasha Gowda
- Subjects
Isatin ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Article ,Polymerization ,Structure-Activity Relationship ,Tubulin ,Cell Line, Tumor ,Drug Discovery ,medicine ,Humans ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,biology ,Tubulin Modulators ,Chemistry ,Organic Chemistry ,Vinblastine ,Oncogene Protein v-akt ,Mechanism of action ,biology.protein ,Molecular Medicine ,Phosphorylation ,medicine.symptom ,Vinblastine Sulfate ,Drug Screening Assays, Antitumor ,medicine.drug ,Signal Transduction - Abstract
A novel series of 5,7-dibromoisatin analogs were synthesized and evaluated for their cytotoxicities against four human cancer cell lines including colon HT29, breast MCF-7, lung A549 and melanoma UACC903. Analogs 6, 11 and 13 displayed good in vitro anticancer activity on the HT29 human colon cancer cell line in the 1 μM range. Analogs 5, 9 and 12, containing a selenocyanate group in the alkyl chain were the most promising compounds on the breast cancer MCF-7 cell line. Biological assays relating to apoptosis were performed to understand the mechanism of action of these analogs. Compounds 5 and 6 were found to inhibit tubulin polymerization to the same extent as the anticancer drug vinblastine sulfate, but compounds 11 and 13 inhibited significantly better than vinblastine. Further western blot analysis suggested that compound 6 at 2 μM reduced both levels and phosphorylation state of Akt. Compounds 11 and 13 at 1 μM caused reduced Akt protein levels and strongly suppressed the phosphorylation of Akt. Therefore, 11 and 13 were demonstrated as efficient dual inhibitors of both tubulin polymerization and the Akt pathway and good candidates for further study. More importantly, the strategy of microtubule and Akt dual inhibitors might be a promising direction for developing novel drugs for cancer.
- Published
- 2011
31. Epidermal growth factor receptor revisited: prognostic marker or target for therapy?
- Author
-
Rosalyn B. Irby
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Colorectal cancer ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Epidermal growth factor receptor ,Molecular Targeted Therapy ,Neoplasm Metastasis ,Pharmacology ,biology ,business.industry ,Antibodies, Monoclonal ,medicine.disease ,Prognosis ,Egfr expression ,ErbB Receptors ,Mutation ,biology.protein ,Molecular Medicine ,business ,Colorectal Neoplasms - Abstract
Commentary to:EGFR expression variance in paired colorectal cancer primary and metastatic tumorsNirit Yarom, Celia Marginean, Terence Moyana, Ivan Gorn-Hondermann , Chaim Birnboim, Horia Marginean, Rebecca Auer, Micheal Vickers, Timothy Asmis, Jean Maroun and Derek Jonker
- Published
- 2010
32. Voltage-gated Na+ channel SCN5A is a key regulator of a gene transcriptional network that controls colon cancer invasion
- Author
-
Bryan Frank, Tim G. Hales, Vincent Obias, Narine Sarvazyan, Norman H. Lee, Carrie D. House, Truong V. Luu, Rosalyn B. Irby, Robert L. Strausberg, Joshua M. Stuart, Charles J. Vaske, and Arnold M. Schwartz
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Transcription, Genetic ,Colorectal cancer ,Muscle Proteins ,Biology ,Sodium Channels ,Article ,Metastasis ,NAV1.5 Voltage-Gated Sodium Channel ,Steroid metabolic process ,Cell Movement ,medicine ,Humans ,Gene Regulatory Networks ,Neoplasm Invasiveness ,Regulation of gene expression ,Gene knockdown ,Microarray analysis techniques ,Reverse Transcriptase Polymerase Chain Reaction ,Wnt signaling pathway ,Cancer ,medicine.disease ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Oncology ,Colonic Neoplasms ,Cancer research ,Caco-2 Cells ,HT29 Cells - Abstract
Voltage-gated Na+ channels (VGSC) have been implicated in the metastatic potential of human breast, prostate, and lung cancer cells. Specifically, the SCN5A gene encoding the VGSC isotype Nav1.5 has been defined as a key driver of human cancer cell invasion. In this study, we examined the expression and function of VGSCs in a panel of colon cancer cell lines by electrophysiologic recordings. Na+ channel activity and invasive potential were inhibited pharmacologically by tetrodotoxin or genetically by small interfering RNAs (siRNA) specifically targeting SCN5A. Clinical relevance was established by immunohistochemistry of patient biopsies, with strong Nav1.5 protein staining found in colon cancer specimens but little to no staining in matched-paired normal colon tissues. We explored the mechanism of VGSC-mediated invasive potential on the basis of reported links between VGSC activity and gene expression in excitable cells. Probabilistic modeling of loss-of-function screens and microarray data established an unequivocal role of VGSC SCN5A as a high level regulator of a colon cancer invasion network, involving genes that encompass Wnt signaling, cell migration, ectoderm development, response to biotic stimulus, steroid metabolic process, and cell cycle control. siRNA-mediated knockdown of predicted downstream network components caused a loss of invasive behavior, demonstrating network connectivity and its function in driving colon cancer invasion. Cancer Res; 70(17); 6957–67. ©2010 AACR.
- Published
- 2010
33. Prostate apoptosis response protein 4 sensitizes human colon cancer cells to chemotherapeutic 5-FU through mediation of an NFκB and microRNA network
- Author
-
Norman H. Lee, Arun Sharma, Joshua M. Stuart, Danielle M. Pastor, Bi-Dar Wang, Truong V. Luu, Gavin P. Robertson, Steven R. Patierno, Rosalyn B. Irby, Thomas L. Olson, Bryan Frank, Christina Leah B. Kline, and Matthew T. Weirauch
- Subjects
Male ,Ribonuclease III ,Chromatin Immunoprecipitation ,Cancer Research ,Colorectal cancer ,Blotting, Western ,Gene Expression ,Antineoplastic Agents ,Apoptosis ,Biology ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,microRNA ,medicine ,Humans ,Oligonucleotide Array Sequence Analysis ,030304 developmental biology ,0303 health sciences ,Microscopy, Confocal ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Research ,NF-kappa B ,Prostatic Neoplasms ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,NFKB1 ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,MicroRNAs ,Oncology ,Fluorouracil ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Signal transduction ,Apoptosis Regulatory Proteins ,HT29 Cells ,Proto-Oncogene Proteins c-akt ,Signal Transduction ,medicine.drug - Abstract
BackgroundDiminished expression or activity of prostate apoptosis response protein 4 (Par-4) has been demonstrated in a number of cancers, although reports on Par-4 expression during colon cancer progression are lacking. An understanding of the molecular events in conjunction with the genetic networks affected by Par-4 is warranted.ResultsColon cancer specimens derived from patients have significantly diminished expression of Par-4 mRNA relative to paired normal colon. Hence, the functional consequences of reintroducing Par-4 into HT29 colon cancer cells were assessed. Overexpression augmented the interaction of Par-4 with NFκB in the cytosol but not nucleus, and facilitated apoptosis in the presence of 5-fluorouracil (5-FU). Analogous findings were obtained when AKT1 pro-survival signaling was inhibited. Transcriptome profiling identified ~700 genes differentially regulated by Par-4 overexpression in HT29 cells. Nearly all Par-4-regulated genes were shown by promoter analysis to containcis-binding sequences for NFκB, and meta-analysis of patient expression data revealed that one-third of these genes exist as a recurrent co-regulated network in colon cancer specimens. Sets of genes involved in programmed cell death, cell cycle regulation and interestingly the microRNA pathway were found overrepresented in the network. Noteworthy, Par-4 overexpression decreased NFκB occupancy at the promoter of one particular network geneDROSHA, encoding a microRNA processing enzyme. The resulting down-regulation ofDROSHAwas associated with expression changes in a cohort of microRNAs. Many of these microRNAs are predicted to target mRNAs encoding proteins with apoptosis-related functions. Western and functional analyses were employed to validate several predictions. For instance, miR-34a up-regulation corresponded with a down-regulation of BCL2 protein. Treating Par-4-overexpressing HT29 cells with a miR-34a antagomir functionally reversed both BCL2 down-regulation and apoptosis by 5-FU. Conversely, bypassing Par-4 overexpression by direct knockdown ofDROSHAexpression in native HT29 cells increased miR-34a expression and 5-FU sensitivity.ConclusionOur findings suggest that the initiation of apoptotic sensitivity in colon cancer cells can be mediated by Par-4 binding to NFκB in the cytoplasm with consequential changes in the expression of microRNA pathway components.
- Published
- 2010
- Full Text
- View/download PDF
34. Primary cell lines: false representation or model system? a comparison of four human colorectal tumors and their coordinately established cell lines
- Author
-
Danielle M, Pastor, Lisa S, Poritz, Thomas L, Olson, Christina L, Kline, Leonard R, Harris, Walter A, Koltun, Vernon M, Chinchilli, and Rosalyn B, Irby
- Subjects
Original Article - Abstract
Cultured cell lines have played an integral role in the study of tumor biology since the early 1900's. The purpose of this study is to evaluate colorectal cancer (CRC) cell lines with respect to progenitor tumors and assess whether these cells accurately and reliably represent the cancers from which they are derived. Primary cancer cell lines were derived from fresh CRC tissue. Tumorigenicity of cell lines was assessed by subcutaneous injection of cells into athymic mice and calculation of tumor volume after 3 weeks. DNA ploidy was evaluated by flow cytometry. Invasive ability of the lines was tested with the MATRIGEL™ invasion assay at 24 or 48 hours. Cells were assessed for the presence of Kirsten-Ras (K-Ras), p-53, deleted in colon cancer (DCC), and Src. Protein profiling of cells and tissue was performed by surface enhanced laser desorption/ionization-time of flight/mass spectroscopy. microRNA expression in cell and tumor tissue samples was evaluated by FlexmiR™ MicroRNA Assays. Four cell lines were generated from tumor tissue from patients with CRC and confirmed to be tumorigenic (mean tumor volume 158.46 mm3). Two cell lines were noted to be diploid; two were aneuploid. All cell lines invaded the MATRIGEL™ starting as early as 24 hours. K-Ras, p53, DCC, and Src expression were markedly different between cell lines and corresponding tissue. Protein profiling yielded weak-to-moderate correlations between cell samples and respective tissues of origin. Weak-to-moderate tau correlations for levels of expression of human microRNAs were found between cells and respective tissue samples for each of the four pairings. Although our primary CRC cell lines vary in their expression of several tumor markers and known microRNAs from their respective progenitor tumor tissue, they do not statistically differ in overall profiles. Characteristics are retained that deem these cell lines appropriate models of disease; however, data acquired through the use of cell culture may not always be a completely reliable representation of tumor activity in vivo.
- Published
- 2010
35. Effects of copper and tributyltin on stress protein abundance in the rotifer Brachionus plicatilis
- Author
-
Bruce J. Cochrane, Terry W. Snell, and Rosalyn B. Irby
- Subjects
Hot Temperature ,Sodium arsenite ,Blotting, Western ,Immunology ,Rotifera ,Gene Expression ,Rotifer ,Chloride ,Antibodies ,Toxicology ,chemistry.chemical_compound ,Heat shock protein ,medicine ,Animals ,Sodium dodecyl sulfate ,Heat-Shock Proteins ,Pharmacology ,biology ,Proteins ,Brachionus ,biology.organism_classification ,Molecular Weight ,Biochemistry ,chemistry ,Metals ,Protein Biosynthesis ,Tributyltin ,Sodium azide ,Trialkyltin Compounds ,Copper ,medicine.drug - Abstract
1. Exposure of the rotifer Brachionus plicatilis to elevated temperature resulted in the synthesis of a number of proteins, including a prominent one of 58,000 Da (SP58). 2. This protein is immunologically crossreactive with the 65,000 Da heat shock protein of the moth Hehothis virescens, which is a member of a highly conserved family of mitochondrial proteins. 3. Exposure of rotifers to sublethal doses ofCuSO4 leads to a 4–5-fold increase in abundance of SP58, with maximum increase occurring at a dose that is approximately 5% of the lc 50 for that compound. 4. A similar response was seen with tributyl tin (TBT). Kinetics of induction were sigmoidal, with induction occurring in the range of 20–30 μg/l. 5. No response was observed when rotifers were exposed to aluminum chloride, mercury chloride, pentachlorophenol, sodium arsenite, sodium azide, sodium dodecyl sulfate, or zinc chloride. 6. These results indicate that changes in stress protein abundance may prove useful as a biomarker of exposure to particular toxicants.
- Published
- 1991
- Full Text
- View/download PDF
36. Src kinase induces tumor formation in the c-SRC C57BL/6 mouse
- Author
-
Robert W. Engelman, Christina Leah B. Kline, Rosalind J. Jackson, Warren Jack Pledger, Rosalyn B. Irby, and Timothy J. Yeatman
- Subjects
Genetically modified mouse ,Cancer Research ,Tumor suppressor gene ,Transgene ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Metastasis ,Mice ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,DNA Primers ,Oncogene ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Cancer ,Neoplasms, Experimental ,medicine.disease ,Mice, Inbred C57BL ,src-Family Kinases ,Oncology ,Cancer research ,Metallothionein ,Carcinogenesis ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Src kinase has been linked as a causative agent in the progression of a number of cancers including colon, breast, lung and melanoma. Src protein and activity levels are increased in colorectal cancer and liver metastases arising secondary to colon cancer. However, although Src protein is increased in colon cancer as early as the adenomatous polyp stage, a role for Src in carcinogenesis has not been established. We developed the c-SRC transgenic mouse in the C57BL/6 strain to address the issue of carcinogenesis in cells with high levels of Src expression. The transgene was constructed with the human c-SRC gene downstream of the mouse metallothionein promoter to create zinc inducible gene expression. In these C57BL/6 mice, Src protein was increased in a number of tissues both with and without zinc induction. No additional carcinogenic agent was administered. After 20 months, mice were assessed for tumor development in the liver and GI tract, as well as other organs. Of the mice with the transgene, 15% developed tumors in the liver while no tumors were detected in wild type C57BL/6 mice. A further study was conducted by crossing c-SRC C57BL/6 mice with p21 nullizygous mice to determine the effect of oncogene expression combined with inactivation of the tumor suppressor gene, p21. Addition of the c-SRC transgene to the p21−/− background increased tumor formation almost 3-fold, while it increased metastasis 6-fold. The data from our study show, for the first time, that Src kinase may play a role in carcinogenesis. © 2008 Wiley-Liss, Inc.
- Published
- 2008
37. Iterative microarray and RNA interference-based interrogation of the SRC-induced invasive phenotype
- Author
-
Greg Bloom, Rosalyn B. Irby, Bryan C. Frank, Timothy J. Yeatman, Noah E. Letwin, Kathleen Verratti, Norman H. Lee, Jennifer Tsai, and Renae L. Malek
- Subjects
Cancer Research ,Biology ,RNA interference ,Gene expression ,Biomarkers, Tumor ,Cell Adhesion ,Tumor Cells, Cultured ,Humans ,Neoplasm Invasiveness ,Gene Silencing ,Gene ,Oligonucleotide Array Sequence Analysis ,Genetics ,Gene knockdown ,Gene Expression Profiling ,Phenotype ,Cell biology ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Genes, src ,Oncology ,Colonic Neoplasms ,RNA Interference ,DNA microarray ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Src kinase has long been recognized as a factor in the progression of colorectal cancer and seems to play a specific role in the development of the metastatic phenotype. In spite of numerous studies conducted to elucidate the exact role of Src in cancer progression, downstream targets of Src remain poorly understood. Gene expression profiling has permitted the identification of large sets of genes that may be functionally interrelated but it is often unclear as to which molecular pathways they belong. Here we have developed an iterative approach to experimentally reconstruct a network of gene activity regulated by Src and contributing to the invasive phenotype. Our strategy uses a combination of phenotypic anchoring of gene expression profiles and loss-of-function screening by way of RNA-mediated interference. Using a panel of human colon cancer cell lines exhibiting differential Src-specific activity and invasivity, we identify the first two levels of gene transcription responsible for the invasive phenotype, where first-tier genes are controlled by Src activity and the second-tier genes are under the influence of the first tier. Specifically, perturbation of first-tier gene activity by either pharmacologic inhibition of Src or RNA-mediated interference–directed knockdown leads to a loss of invasivity and decline of second-tier gene activity. The targeting of first-tier genes may be bypassed altogether because knockdown of second-tier genes led to a similar loss of invasive potential. In this manner, numerous members of a “transcriptional cascade” pathway for metastatic activity have been identified and functionally validated.
- Published
- 2005
38. Osteopontin identified as colon cancer tumor progression marker
- Author
-
Timothy J. Yeatman, Domenico Coppola, Alan B. Cantor, John Quackenbush, Ann F. Chambers, Marianna Szabo, Tingan Chen, Deepak Agrawal, and Rosalyn B. Irby
- Subjects
Microarray ,Colorectal cancer ,Sialoglycoproteins ,Integrin ,Biology ,General Biochemistry, Genetics and Molecular Biology ,chemistry.chemical_compound ,Molecular marker ,medicine ,Biomarkers, Tumor ,Humans ,Osteopontin ,chemistry.chemical_classification ,General Immunology and Microbiology ,CD44 ,General Medicine ,medicine.disease ,chemistry ,Tumor progression ,Immunology ,Colonic Neoplasms ,Cancer research ,biology.protein ,Disease Progression ,RNA ,General Agricultural and Biological Sciences ,Glycoprotein - Abstract
Identifying molecular markers for colon cancer is a top priority. Using a pooled sample approach with Affymetrix GeneChip technology, we assayed colon cancers derived from a series of clinical stages to identify molecular markers of potential prognostic value. Of 12000 genes assessed, osteopontin emerged as the leading candidate tumor progression marker. Osteopontin is a secreted glycoprotein known to bind integrins and CD44. Its actual molecular function remains elusive but its increased expression correlates strongly with tumor progression.
- Published
- 2004
39. Increased Src activity disrupts cadherin/catenin-mediated homotypic adhesion in human colon cancer and transformed rodent cells
- Author
-
Rosalyn B, Irby and Timothy J, Yeatman
- Subjects
Protein-Tyrosine Kinases ,Cadherins ,Transfection ,Rats ,Proto-Oncogene Proteins p21(ras) ,Cytoskeletal Proteins ,src-Family Kinases ,Focal Adhesion Kinase 1 ,Focal Adhesion Protein-Tyrosine Kinases ,Colonic Neoplasms ,Cell Adhesion ,Trans-Activators ,Tumor Cells, Cultured ,Animals ,Humans ,Phosphorylation ,beta Catenin ,Cell Line, Transformed - Abstract
Src has been implicated in the development and progression of human colon cancer. Because the capacity for tumor cells to dissociate from the primary tumor is a critical step in the development of metastases, the effect of a naturally occurring, activated Src-531 on intercellular adhesion was examined. Homotypic adhesion was assessed using dissociation assays on Src-transformed rat fibroblasts and human colon cancer cell lines. The data indicate that both rodent and human cells expressing the mutant Src protein display up to 7-fold less homotypic adhesion than do wild-type cells (P0.01). Experiments demonstrated that cadherin was phosphorylated in cells transfected with activated Src and that cadherin/catenin complexes were disrupted as a result. Experiments using dominant negative (DN) Src or an Src-specific inhibitor (PD 180970), demonstrated that adhesion was restored when Src activity was inhibited in Src-531 transfectants, confirming that Src is a causal factor in the decreased homotypic adhesion observed. In addition, DN Ras, DN focal adhesion kinase (FAK), but not Stat3beta, restored intercellular adhesion, which suggested that Ras and FAK may be downstream effectors of Src-mediated homotypic adhesion. Collectively, these data support a role for Src, Ras, and FAK in the regulation of intercellular adhesion, which may in turn regulate metastatic potential of human colon cancer cells.
- Published
- 2002
40. Identification of Src transformation fingerprint in human colon cancer
- Author
-
Renae L. Malek, Jennifer Tsai, Qingbin M. Guo, Timothy J. Yeatman, Mei He, Edison T. Liu, Richard Jove, Sylvia Wong, Norman H. Lee, Rosalyn B. Irby, John Quackenbush, Bryan C. Frank, and Kerry Lee
- Subjects
Cancer Research ,Pyridones ,Biology ,medicine.disease_cause ,Gene expression ,Genetics ,medicine ,Animals ,Cluster Analysis ,Humans ,Molecular Biology ,Gene ,Transcription factor ,Cell Line, Transformed ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Cancer ,medicine.disease ,Phenotype ,Cell biology ,Rats ,Up-Regulation ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Genes, src ,Cell Transformation, Neoplastic ,Pyrimidines ,Colonic Neoplasms ,Carcinogenesis ,Proto-oncogene tyrosine-protein kinase Src - Abstract
We used a classical rodent model of transformation to understand the transcriptional processes, and hence the molecular and cellular events a given cell undergoes when progressing from a normal to a transformed phenotype. Src activation is evident in 80% of human colon cancer, yet the myriad of cellular processes effected at the level of gene expression has yet to be fully documented. We identified a Src 'transformation fingerprint' within the gene expression profiles of Src-transformed rat 3Y1 fibroblasts demonstrating a progression in transformation characteristics. To evaluate the role of this gene set in human cancer development and progression, we extracted the orthologous genes present on the Affymetrix Hu95A GeneChip (12k named genes) and compared expression profiles between the Src-induced rodent cell line model of transformation and staged colon tumors where Src is known to be activated. A similar gene expression pattern between the cell line model and staged colon tumors for components of the cell cycle, cytoskeletal associated proteins, transcription factors and lysosomal proteins suggests the need for co-regulation of several cellular processes in the progression of cancer. Genes not previously implicated in tumorigenesis were detected, as well as a set of 14 novel, highly conserved genes with here-to-fore unknown function. These studies define a set of transformation associated genes whose up-regulation has implications for understanding Src mediated transformation and strengthens the role of Src in the development and progression of human colon cancer. Supportive Supplemental Data can be viewed at http://pga.tigr.org/PGApubs.shtml.
- Published
- 2002
41. Stat3-mediated Myc expression is required for Src transformation and PDGF-induced mitogenesis
- Author
-
Timothy J. Yeatman, John M. Sedivy, Sara A. Courtneidge, Tammy Bowman, Richard Jove, W. J. Pledger, Rosalyn B. Irby, Martin A. Broome, Dominic Sinibaldi, and Walker Wharton
- Subjects
STAT3 Transcription Factor ,Platelet-derived growth factor ,Genes, myc ,Models, Biological ,Proto-Oncogene Proteins c-myc ,chemistry.chemical_compound ,Mice ,Animals ,STAT3 ,Cell Line, Transformed ,Platelet-Derived Growth Factor ,Multidisciplinary ,biology ,3T3 Cells ,Biological Sciences ,Recombinant Proteins ,DNA-Binding Proteins ,Genes, src ,Cell Transformation, Neoplastic ,SU6656 ,chemistry ,Gene Expression Regulation ,biology.protein ,STAT protein ,Cancer research ,Trans-Activators ,Ectopic expression ,Signal transduction ,Platelet-derived growth factor receptor ,Cell Division ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
Signal transducer and activator of transcription (STAT) proteins perform key roles in mediating signaling by cytokines and growth factors, including platelet-derived growth factor (PDGF). In addition, Src family kinases activate STAT signaling and are required for PDGF-induced mitogenesis in normal cells. One STAT family member, Stat3, has been shown to have an essential role in cell transformation by the Src oncoprotein. However, the mechanisms by which STAT-signaling pathways contribute to mitogenesis and transformation are not fully defined. We show here that disruption of Stat3 signaling by using dominant-negative Stat3β protein in NIH 3T3 fibroblasts suppresses c-Myc expression concomitant with inhibition of v-Src-induced transformation. Ectopic expression of c-Myc is able to partially reverse this inhibition, suggesting that c-Myc is a downstream effector of Stat3 signaling in v-Src transformation. Furthermore, c -myc gene knockout fibroblasts are refractory to transformation by v-Src, consistent with a requirement for c-Myc protein in v-Src transformation. In normal NIH 3T3 cells, disruption of Stat3 signaling with dominant-negative Stat3β protein inhibits PDGF-induced mitogenesis in a manner that is reversed by ectopic c-Myc expression. Moreover, inhibition of Src family kinases with the pharmacologic agent, SU6656, blocks Stat3 activation by PDGF. These findings, combined together, delineate the signaling pathway, PDGF → Src → Stat3 → Myc, that is important in normal PDGF-induced mitogenesis and subverted in Src transformation.
- Published
- 2001
42. Role of Src expression and activation in human cancer
- Author
-
Rosalyn B. Irby and Timothy J. Yeatman
- Subjects
Regulation of gene expression ,Genetics ,Cancer Research ,biology ,Drug discovery ,Cancer ,Antineoplastic Agents ,biology.organism_classification ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Genes, src ,Retrovirus ,src-Family Kinases ,Neoplasms ,Gene expression ,Cancer cell ,Cancer research ,medicine ,Humans ,Signal transduction ,Molecular Biology ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Since the original identification of a transmissible agent responsible for the development of tumors in chickens, now known to be a retrovirus encoding the v-src gene, significant progress has been made in defining the potential functions of its human homolog, SRC. The product of the human SRC gene, c-Src, is found to be over-expressed and highly activated in a wide variety of human cancers. The relationship between Src activation and cancer progression appears to be significant. Moreover, Src may have an influence on the development of the metastatic phenotype. This review discusses the data supporting a role for c-Src as a critical component of the signal transduction pathways that control cancer cell development and growth, and provides the rationale for targeting Src in drug discovery efforts.
- Published
- 2000
43. Activation of c-Src by receptor tyrosine kinases in human colon cancer cells with high metastatic potential
- Author
-
Marek Wloch, Timothy J. Yeatman, Rosalyn B. Irby, Hua Yu, Richard Jove, Joel G. Turner, Weiguang Mao, Ling Fu, Roy Garcia, and Domenico Coppola
- Subjects
Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,Colonic Polyps ,Receptor tyrosine kinase ,Epidermal growth factor ,Internal medicine ,Genetics ,medicine ,Tumor Cells, Cultured ,Humans ,Epidermal growth factor receptor ,Neoplasm Metastasis ,Phosphorylation ,Molecular Biology ,biology ,Epidermal Growth Factor ,Receptor Protein-Tyrosine Kinases ,Neoplasm Proteins ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Genes, src ,Endocrinology ,Phenotype ,Hepatocyte Growth Factor Receptor ,ROR1 ,Colonic Neoplasms ,Cancer research ,biology.protein ,Signal transduction ,Tyrosine kinase ,Protein Kinases ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Recent data suggest that signal transduction may have a critical role in the development and regulation of the metastatic phenotype. Here, we investigated the role of c-Src activation in the process of human colon cancer metastasis to the liver. Our data, derived from two different sets of human colon cancer cell line metastatic variants, suggest that not only do highly-metastatic cells display constitutively elevated c-Src protein kinase activity when compared to poorly metastatic cells, but also that receptor tyrosine kinases participate in the ligand-activation of c-Src above basal levels. Specifically, the epidermal growth factor receptor (EGFR), p185HER2/Neu and the hepatocyte growth factor receptor (c-Met) appear to be linked to the process because they preferentially activate c-Src in highly-metastatic cells. EGFR was found to associate with c-Src in colon cancer cells and specific inhibitors of the EGFR resulted in a reduction of c-Src activity to basal levels. In addition, c-Src transfectants displayed partially-activated EGFRs, suggesting a feedback role for c-Src in the regulation of the EGFR. p185HER2/Neu was also identified in immunocomplexes of c-Src following ligand activation of the EGFR, but only in highly-metastatic cells. Collectively, these observations suggest a paradigm whereby c-Src interacts with multiple cell-surface growth factors in a catalytic fashion for the development of tumor cells with metastatic potential.
- Published
- 1998
44. The Akt Inhibitor ISC-4 Synergizes with Cetuximab in 5-FU-Resistant Colon Cancer
- Author
-
David T. Dicker, Rosalyn B. Irby, Wafik S. El-Deiry, Arun Sharma, Shantu Amin, Joshua E. Allen, and Jean-Nicolas Gallant
- Subjects
Mouse ,Colorectal cancer ,Cancer Treatment ,lcsh:Medicine ,Cetuximab ,Apoptosis ,Pharmacology ,medicine.disease_cause ,Mice ,0302 clinical medicine ,Organoselenium Compounds ,Molecular Cell Biology ,Pathology ,lcsh:Science ,0303 health sciences ,Multidisciplinary ,Melanoma ,Cell Cycle ,Drug Synergism ,Animal Models ,Signaling Cascades ,3. Good health ,Gene Expression Regulation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Medicine ,Female ,Oncology Agents ,Fluorouracil ,KRAS ,Research Article ,Signal Transduction ,medicine.drug ,Combination therapy ,Cell Survival ,Mice, Nude ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Model Organisms ,Diagnostic Medicine ,In vivo ,Cell Line, Tumor ,Gastrointestinal Tumors ,Akt Signaling Cascade ,medicine ,Animals ,Humans ,Biology ,neoplasms ,Cell Proliferation ,030304 developmental biology ,business.industry ,lcsh:R ,Cancers and Neoplasms ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,Drug Resistance, Neoplasm ,Cell culture ,Cancer research ,lcsh:Q ,business ,Proto-Oncogene Proteins c-akt ,Biomarkers ,General Pathology - Abstract
Phenylbutyl isoselenocyanate (ISC-4) is an Akt inhibitor with demonstrated preclinical efficacy against melanoma and colon cancer. In this study, we sought to improve the clinical utility of ISC-4 by identifying a synergistic combination with FDA-approved anti-cancer therapies, a relevant and appropriate disease setting for testing, and biomarkers of response. We tested the activity of ISC-4 and 19 FDA-approved anticancer agents, alone or in combination, against the SW480 and RKO human colon cancer cell lines. A synergistic interaction with cetuximab was identified and validated in a panel of additional colon cancer cell lines, as well as the kinetics of synergy. ISC-4 in combination with cetuximab synergistically reduced the viability of human colon cancer cells with wild-type but not mutant KRAS genes. Further analysis revealed that the combination therapy cooperatively decreased cell cycle progression, increased caspase-dependent apoptosis, and decreased phospho-Akt in responsive tumor cells. The synergism between ISC-4 and cetuximab was retained independently of acquired resistance to 5-FU in human colon cancer cells. The combination demonstrated synergistic anti-tumor effects in vivo without toxicity and in the face of resistance to 5-FU. These results suggest that combining ISC-4 and cetuximab should be explored in patients with 5-FU-resistant colon cancer harboring wild-type KRAS.
- Published
- 2013
- Full Text
- View/download PDF
45. Abstract 1418: Voltage-gated Na+ channel activation leads to increased invasion potential through persistent MAPK signaling involving PKA, Src, and Rap1
- Author
-
Norman H. Lee, Carrie D. House, Rosalyn B. Irby, and May Samaan
- Subjects
MAPK/ERK pathway ,Cancer Research ,Voltage-gated ion channel ,Biology ,medicine.disease_cause ,Cell biology ,chemistry.chemical_compound ,Oncology ,chemistry ,Cancer cell ,Immunology ,medicine ,Signal transduction ,Veratridine ,Carcinogenesis ,Transcription factor ,Proto-oncogene tyrosine-protein kinase Src - Abstract
An interesting new field in cancer research is the role of voltage-gated Na+ channels (VGSCs) in cancer cell invasion. Expression of these ion channels is normally restricted to excitable cells, such as neurons, however functional expression has been demonstrated in multiple cancer cell types where channel activity leads to changes in invasion potential in vitro. We have recently demonstrated functional expression of Nav1.5, a protein encoded by SCN5A, in a variety of colon cancer cell lines. We showed that pharmacological inhibition of channel activity leads to a decrease in invasion potential, whereas pharmacological activation of the channel leads to an increase in invasion potential. Moreover, upon examination of patient samples we found that protein expression is restricted to malignant tissue. What has remained largely uncharacterized is the mechanism by which these channels promote oncogenic behavior. We have recently established that VGSC isoform SCN5A participates in an invasion gene network for colon cancer, regulating expression of several genes important for invasion. Our current study focuses on determining what non-genomic signaling pathway(s) are activated in response to VGSC activity that might lead gene expression changes. The MAPK pathway is deregulated in many cancers and VGSC activation in neurons leads to MAPK activation during neurogenesis. Coincidentally, many genes regulated by SCN5A have binding sites for transcription factors phosphorylated by ERK1/2. We hypothesized that VGSC activation promotes MAPK activation in colon cancer cells to provide an oncogenic advantage. Using pharmacological inhibition of MAPK, we demonstrate the requirement of this molecule for colon cancer invasion in vitro. Furthermore, we show through Western blot analysis of phosphorylated ERK1/2, that MAPK activity is diminished when colon cancer cells are treated with VGSC inhibitor, lidocaine, and persistently enhanced for 24-48 hours in the presence of the VGSC activator, veratridine. This persistent activation and the increase in invasion potential are partially lost when veratridine is used in the presence of a PKA inhibitor. Similarly, veratridine treatment in the presence of a siRNA directed against Rap1 leads to a decrease in both invasion potential and MAPK activity compared to veratridine alone. Lastly, we demonstrate increased Src activity after treatment with veratridine. Taken together, our data suggest that VGSC activation leads to an increase in MAPK activity through activation of PKA, Rap1 and Src in colon cancer cells. We further show that Rap1 activation is biphasic with early persistent signaling likely through PKA and delayed persistent signaling through Src. The proposed signaling pathway illustrates for the first time a potential mechanism by which VGSC activity promotes oncogenesis in colon cancer cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1418. doi:10.1158/1538-7445.AM2011-1418
- Published
- 2011
- Full Text
- View/download PDF
46. HTLV-1 Epitope (BA21) Reactivity in Rare Bone Marrow Failure Diseases
- Author
-
Lubomir Sokol, Kendall Thomas Baab, Ronald Paquette, Nancy Ruth Jarbadan, Pearlie K. Epling-Burnette, David Cuthbertson, Jason Liao, Thomas P. Loughran, Daniel Krissinger, Rosalyn B. Irby, Michael J. Clemente, Susan B. Nyland, Jaroslaw P. Maciejewski, and Eric Schaefer
- Subjects
education.field_of_study ,biology ,Myelodysplastic syndromes ,Immunology ,Population ,chemical and pharmacologic phenomena ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Virology ,Epitope ,CTL ,Leukemia ,medicine.anatomical_structure ,Antigen ,medicine ,biology.protein ,Bone marrow ,Antibody ,education - Abstract
Abstract 1724 LGL leukemia is thought to be an antigen-driven disease. Extensive data support the antigen activated nature of leukemic LGL cells. Leukemic LGL constitutively express perforin and other markers of activated killer cells. T-LGL leukemia has been characterized as an accumulation of apoptosis resistant effector memory cytotoxic T lymphocytes (CTL). The actual target recognized by these activated CTL is still not characterized. LGL leukemia is at one end of a spectrum of hematologic disorders characterized by aberrant CTL responses. Conditions at the other end of this spectrum also feature CTL attacks on bone marrow, leading to cytopenia. These conditions are known as bone marrow failure (BMF) diseases and include pure red cell aplasia (PRCA), myelodysplastic syndromes (MDS), aplastic anemia (AA), and paroxysmal nocturnal hemoglobinuria (PNH). LGL leukemia is characterized by an easily detectable dominant clone whereas the CTL clones in these marrow failure diseases are less obvious. It has been postulated that exposure to infectious agents contributes to the onset of the full spectrum of LGL-driven diseases. One approach to determine exposure to infectious agents is to study the antibody patterns of infected and non-infected populations. In clinical screening, sera are usually assayed by ELISA to check for reactivity against recombinant viral proteins, followed by Western blot and PCR. In the case of human T cell leukemia viruses 1 and 2 (HTLV-1/2), less than 1% of the general U.S. population demonstrates cross-reactive antibodies. Conversely, at least 21% of LGL leukemia patients exhibit cross-reactive serology to HTLV1/2. The pattern of reactivity is very different between LGL leukemia patients and other cross-reactive groups. Comprehensive testing reveals that most LGL leukemia patients are not infected with HTLV-1/2, or with HTLV-3/4. We have demonstrated that HTLV Env reactivity in LGL leukemia was directed at the transmembrane-associated BA21 region/p21e. The BA21 epitope overlaps the immunogenic p21e region of the HTLV-1 envelope. Small scale studies indicated that at least 25% of sera from LGL leukemia patients were reactive to BA21, and that reactivity coincided with elevated overall HTLV-1 serum antibody expression. Since the presumptive antigen involved in the pathogenesis of LGL leukemia is not known, the identification of disease-specific antigens that are not cross-reactive with normal specimens is critical. To achieve this goal, a different way to look at epitope specificity is needed. We addressed this need by performing a B-cell epitope analysis for the BA21 sequence using multiple prediction databases. Antigenicity was predicted for three regions of BA21, namely the amino terminus containing EQCR, and two regions near the carboxyl terminus (PPLE, WGLN). Of these, PPLE-containing sequences were predicted to be the most immunogenic. We then used array-adapted alanine screening followed by ELISA to select disease-specific BA21 epitopes. Overall, 80 sequences were tested for specificity against alanine substitutions, using up to 10 mcL of serum per patient. The results of the array-adapted alanine screen clearly demonstrated that PPLE-containing sequences were universally recognized by LGL leukemia and normal serum groups. Conversely, peptides that included the amino terminus with sequence EQCR were better recognized against their alanine substituted sequences. ELISA testing was therefore performed for peptides from these regions. The resulting BA21 epitope was recognized by at least 40% of LGL leukemia sera but not by normal donor sera. Since a common pathogenesis is postulated for other CTL-mediated hematologic diseases, the levels of BMF-BA21 IgG were determined for participants in the Bone Marrow Failure Diseases Consortium of the Rare Diseases Clinical Research Network. We found that a substantial number of sera from participants with MDS, AA, PNH and PRCA also recognized the epitope. Reactivity with the BMF-BA21 epitope was independently associated with CTL-driven BMF diseases. These data provide further support for the hypothesis that a variety of hematologic diseases characterized by antigen-activated CTL result from a common pathogenetic mechanism. This project was supported by NIH Grant Number U54RR019397. Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the NIH. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2010
- Full Text
- View/download PDF
47. Abstract 1209: Par-4 as therapy for colon cancer
- Author
-
Christina Leah B. Kline, Rosalyn B. Irby, and Arun Sharma
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,Colorectal cancer ,Wild type ,Cancer ,Transfection ,medicine.disease ,HT29 Cells ,Oncology ,Apoptosis ,Cell culture ,Cancer stem cell ,Cancer research ,Medicine ,business - Abstract
Par-4 (Prostate apoptosis response-4) is a tumor suppressor that induces apoptosis in tumor cells in response to apoptotic stimuli. It has been reported to be reduced in a number of human cancers. We examined the expression levels of Par-4 in human colon cancers and explored the possibility of using Par-4 as a treatment for colon cancer. We found that expression of Par-4 is reduced in human colon cancers 2.65-fold compared to paired normal colon. To assess the role of Par-4 in colon cancer growth, the human colon cancer cell line, HT29, with low native Par-4 expression, was transfected to overexpress Par-4. It has been shown that apoptosis was higher in the transfected cells than in the mock transfected cells. It was also shown that tumors growing in nude mice were smaller when the cells were transfected to overexpress Par-4. Importantly for therapeutic considerations, it has been reported that Par-4 expressing cells can have a bystander effect on tumor cells that are not transfected, also inducing apoptosis in the untransfected cells. We hypothesized that HT29 colon cancer cells producing Par-4 in vivo would secrete Par-4 into the animal system and result in lower tumor volume of tumors created with wild type cells growing distally. Methods: To show proof of concept, nude mice were injected subcutaneously with wild type HT29 cells. Half of the mice were also injected with HT29 cells overexpressing Par-4 in the opposite flank. Mice were treated with 5-fluorouracil to test the sensitivity of tumors to the chemotherapy. Results: At the end of the first week, the wild type tumors in the mice with Par-4 tumors also were 1.8-fold smaller (p=0) than those tumors in mice who received only wild type tumor cell injections. Over the course of three weeks, the wild type tumors growing in mice without Par-4 tumors increased in volume 12-fold more than those wild type tumors growing in mice with Par-4 tumors. In addition, in the event that Par-4 tumors resolved completely (5% of the cases) the wild type tumors in those mice showed increased growth rates. This data suggests that overexpression of Par-4 in tumor cells in vivo not only sensitized those cells to chemotherapy, but had a bystander effect on tumor cells growing distally to the Par-4 overexpressing cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1209.
- Published
- 2010
- Full Text
- View/download PDF
48. Osteopontin induces multiple changes in gene expression that reflect the six hallmarks of cancer in a model of breast cancer progression.
- Author
-
Amy C. Cook, Alan B. Tuck, Susan McCarthy, Joel G. Turner, Rosalyn B. Irby, Gregory C. Bloom, Timothy J. Yeatman, and Ann F. Chambers
- 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.