1
|
Sámano R, Martínez-Rojano H, Chico-Barba G, Gamboa R, Mendoza-Flores ME, Robles-Alarcón FJ, Pérez-Martínez I, Monroy-Muñoz IE. Gestational Weight Gain: Is the Role of Genetic Variants a Determinant? A Review. Int J Mol Sci 2024; 25:3039. [PMID: 38474283 DOI: 10.3390/ijms25053039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Excessive or insufficient gestational weight gain (GWG) leads to diverse adverse maternal and neonatal outcomes. There is evidence that pregestational body mass index (pBMI) plays a role in GWG, but no genetic cause has been identified. In this review, we aim to analyze genotype variants associated with GWG. Results: We identified seven genotype variants that may be involved in GWG regulation that were analyzed in studies carried out in Brazil, Romania, the USA, Turkey, Ukraine, and Canada. Some genetic variants were only associated with GWG in certain races or depending on the pBMI. In women who were obese or overweight before gestation, some genetic variants were associated with GWG. Environmental and genetic factors together showed a greater association with GWG than genetic factors alone; for example, type of diet was observed to have a significant influence. Conclusions: We found little scientific evidence of an association between genotype variants in countries with a high prevalence of women of reproductive age who are overweight and obese, such as in Latin America. GWG may be more dependent on environmental factors than genetic variants. We suggest a deeper study of genetic variants, cytokines, and their possible association with GWG, always with the respective control of potential cofounding factors, such as pBMI, diet, and race.
Collapse
Affiliation(s)
- Reyna Sámano
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico
- Programa de Posgrado Doctorado en Ciencias Biológicas y de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City 04960, Mexico
| | - Hugo Martínez-Rojano
- Sección de Posgrado e Investigación de la Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Gabriela Chico-Barba
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico
| | - Ricardo Gamboa
- Departamento de Fisiología, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - María Eugenia Mendoza-Flores
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico
| | | | - Itzel Pérez-Martínez
- Facultad de Nutrición, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Mexico
| | - Irma Eloisa Monroy-Muñoz
- Departamento de Investigación Clínica en Salud Reproductiva y Perinatal, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico
| |
Collapse
|
2
|
Macrophage IL-1β promotes arteriogenesis by autocrine STAT3- and NF-κB-mediated transcription of pro-angiogenic VEGF-A. Cell Rep 2022; 38:110309. [PMID: 35108537 PMCID: PMC8865931 DOI: 10.1016/j.celrep.2022.110309] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/20/2021] [Accepted: 01/07/2022] [Indexed: 11/23/2022] Open
Abstract
Peripheral artery disease (PAD) leads to considerable morbidity, yet strategies for therapeutic angiogenesis fall short of being impactful. Inflammatory macrophage subsets play an important role in orchestrating post-developmental angiogenesis, but the underlying mechanisms are unclear. Here, we find that macrophage VEGF-A expression is dependent upon the potent inflammatory cytokine, IL-1β. IL-1β promotes pro-angiogenic VEGF-A165a isoform transcription via activation and promoter binding of STAT3 and NF-κB, as demonstrated by gene-deletion, gain-of-function, inhibition, and chromatin immunoprecipitation assays. Conversely, IL-1β-deletion or inhibition of STAT3 or NF-κB increases anti-angiogenic VEGF-A165b isoform expression, indicating IL-1β signaling may also direct splice variant selection. In an experimental PAD model of acute limb ischemia, macrophage IL-1β expression is required for pro-angiogenic VEGF-A expression and for VEGF-A-induced blood flow recovery via angio- or arteriogenesis. Though further study is needed, macrophage IL-1β-dependent transcription of VEGF-A via STAT3 and NF-κB may have potential to therapeutically promote angiogenesis in the setting of PAD. Mantsounga et al. show inflammatory macrophage IL-1β expression to be required for pro-angiogenic VEGF-A expression and consequent post-developmental angio- or arteriogenesis in an experimental model of peripheral artery disease. Autocrine IL-1β signaling promotes transcription of pro-angiogenic VEGF-A165a isoform expression relative to anti-angiogenic isoform, VEGF-A165b, through activation of STAT3 and NF-κB.
Collapse
|
3
|
Chen F, Chen N, Yu Y, Cui J. Efficacy and Safety of Epidermal Growth Factor Receptor (EGFR) Inhibitors Plus Antiangiogenic Agents as First-Line Treatments for Patients With Advanced EGFR-Mutated Non-small Cell Lung Cancer: A Meta-Analysis. Front Oncol 2020; 10:904. [PMID: 32714857 PMCID: PMC7344312 DOI: 10.3389/fonc.2020.00904] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/11/2020] [Indexed: 01/04/2023] Open
Abstract
Background: Tyrosine kinase inhibitors (TKIs) are standard treatment options for non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutations. Increasing clinical investigations have explored the value of EGFR-TKIs plus antiangiogenic drugs as the first-line treatment for EGFR-mutated NSCLC. Methods: We systematically searched PubMed, Cochrane Library, and EMBASE for randomized controlled trials (RCTs) investigating EGFR-TKIs administered with or without antiangiogenic agents for advanced EGFR-mutated NSCLC. The latest RCT that was presented orally at the 2019 European Society for Medical Oncology Congress was obtained online. The endpoints included progression-free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rates (DCRs), and grade 3 or higher adverse events (AEs). Results: We included seven articles on five trials with 1,226 patients. The interventions for the experimental group were the first-generation EGFR-TKI erlotinib combined with bevacizumab (four studies) or ramucirumab (one study), and erlotinib monotherapy (four studies) or erlotinib plus placebo (one study) for the control group. All studies reached their primary study endpoints (i.e., PFS). Compared to erlotinib monotherapy, erlotinib plus antiangiogenic agents remarkably prolonged PFS [hazard ratio (HR) = 0.59, 95% confidence interval (CI) = 0.51-0.69, P = 0.000]; however, ORR, DCR, and OS were similar between the two groups. The overall grade 3-5 AEs increased in combination group (OR = 5.772, 95% CI = 2.38-13.94, P = 0.000), particularly the incidence of diarrhea (OR = 2.51, 95% CI = 1.21-5.23, P = 0.014), acneiform (OR = 1.815, 95% CI = 1.084-3.037, P = 0.023), hypertension (OR = 6.77, 95% CI = 3.62-12.66, P = 0.000), and proteinuria (OR = 13.48, 95% CI = 4.11-44.22, P = 0.000). Additionally, subgroup analysis demonstrated that Asian patients could significantly benefit from combination therapy (HR = 0.59, 95% CI = 0.50-0.69, P = 0.000). Patients with exon 19 deletions (HR = 0.61, 95% CI = 0.49-0.75, P = 0.000) and 21 Leu858Arg mutations (HR = 0.59, 95% CI = 0.47-0.73, P = 0.000) had almost equivalent PFS benefits when treated with double-blocking therapy. Patients with brain metastases at baseline in the combination group had a trend toward better PFS (HR = 0.55, 95% CI = 0.30-1.01, P = 0.001). Conclusions: Erlotinib plus bevacizumab or ramucirumab in EFGR-mutated NSCLC first-line setting yielded remarkable PFS benefits; however, this was accompanied by higher AEs. Epidermal growth factor receptor-TKI plus antiangiogenic agent therapy may be considered a new option for advanced EGFR-mutated NSCLC patients.
Collapse
Affiliation(s)
| | | | | | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Deng C, Xiong J, Gu X, Chen X, Wu S, Wang Z, Wang D, Tu J, Xie J. Novel recombinant immunotoxin of EGFR specific nanobody fused with cucurmosin, construction and antitumor efficiency in vitro. Oncotarget 2018; 8:38568-38580. [PMID: 28445134 PMCID: PMC5503554 DOI: 10.18632/oncotarget.16930] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/24/2017] [Indexed: 11/25/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) overexpression is related to the increased aggressiveness, metastases, and poor prognosis in various cancers. In this study, we successfully constructed a new EGFR nanobody-based immunotoxin rE/CUS containing cucurmosin (CUS), The immunotoxin was expressed by prokaryotic system and we obtained a yield of 5 mg protein per liter expression medium. The percentage of it's binding ability totumor cell lines A549, HepG2, SW116, which highly expressed EGFR was 55.6%, 79.6% and 97.1%, respectively, but SW620 was only 4.45%. rE/CUS has the ability to bind A549, HepG2, SW116 cells specifically, and the antigen binding capability was not affected because of extra part of CUS component. The rE/CUS significantly inhibited the cell viability against EGFR over expression tumor cell lines in a dose-and time-dependent manner. Moreover, rE/CUS also induced apoptosis of HepG2 and A549 mightily. Our results demonstrate that rE/CUS is a potential therapeutic strategy for treating EGFR-positive solid tumors.
Collapse
Affiliation(s)
- Cuimin Deng
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian, China
| | - Jiani Xiong
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xiaofan Gu
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaoying Chen
- Department of Experimental Teaching Center of Basic Medical Science, Fujian Medical University, Fuzhou, Fujian, China
| | - Shuifa Wu
- Department of Pharmacology, The 180th Hospital of PLA, Quanzhou, Fujian, China
| | - Zhe Wang
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian, China
| | - Duanduan Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jinjin Tu
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian, China
| | - Jieming Xie
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
5
|
Cheung CY. Vascular Endothelial Growth Factor Activation of Intramembranous Absorption: A Critical Pathway for Amniotic Fluid Volume Regulation. ACTA ACUST UNITED AC 2016; 11:63-74. [PMID: 14980307 DOI: 10.1016/j.jsgi.2003.09.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The purpose of this review is to propose a critical role for vascular endothelial growth factor (VEGF) in mediating the transfer of amniotic fluid from the amniotic compartment through the fetal membranes and fetal surface of the placenta into fetal blood. METHODS Experimental findings in humans and animal models on the action of VEGF in mediating fluid transfer are reviewed and interpreted in order to postulate a proposed mechanism for VEGF regulation of amniotic fluid absorption through the fetal membranes and placenta. RESULTS Recent scientific advances suggest that up-regulation of VEGF gene expression in the amnion and chorion is associated with increased transfer of amniotic fluid into fetal blood. The possible mechanisms of action for VEGF appear to involve regulation of intramembranous blood vessel proliferation and membrane transport via passive permeation as well as nonpassive transcytotic vesicular movement of fluid. CONCLUSION Currently evolving concepts suggest that amniotic fluid volume is regulated through modulation of the rate of intramembranous absorption of amniotic fluid by both passive and nonpassive mechanisms. The permeability factor VEGF appears to be a critical regulator of amniotic fluid transport in the fetal membranes.
Collapse
Affiliation(s)
- Cecilia Y Cheung
- Division of Perinatal Medicine, Department of Reproductive Medicine, University of California at San Diego, La Jolla, California 92093-0802, USA.
| |
Collapse
|
6
|
Gridelli C, Rossi A, Ciardiello F, De Marinis F, Crinò L, Morabito A, Morgillo F, Montanino A, Daniele G, Piccirillo MC, Normanno N, Gallo C, Perrone F. BEVERLY: Rationale and Design of a Randomized Open-Label Phase III Trial Comparing Bevacizumab Plus Erlotinib Versus Erlotinib Alone as First-Line Treatment of Patients With EGFR-Mutated Advanced Nonsquamous Non-Small-Cell Lung Cancer. Clin Lung Cancer 2016; 17:461-465. [PMID: 27209164 DOI: 10.1016/j.cllc.2016.04.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 04/12/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND About 20% of advanced non-small-cell lung cancer (NSCLC) cases harbor somatic mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR) gene. In these patients, the standard first-line treatments are the EGFR-tyrosine kinase inhibitors, such as gefitinib, erlotinib, or afatinib. Most of these patients develop resistance and relapse within about 1 year of initiation of an EGFR-tyrosine kinase inhibitor. Consequently, it is important to develop new combination strategies to delay this resistance. Preclinical data have showed that EGFR and vascular endothelial growth factor (VEGF) share a common downstream pathway, suggesting the important role of VEGF in the resistance to EGFR blockade. The combination of erlotinib and bevacizumab, an anti-VEGF agent, showed very interesting clinical results. PATIENTS AND METHODS The bevacizumab plus erlotinib study (BEVERLY) is a randomized, open-label, phase III trial investigating first-line erlotinib plus bevacizumab versus erlotinib in patients with advanced NSCLC harboring activating EGFR mutations. The co-primary endpoints are investigator-assessed progression-free survival (PFS) and blinded, independent centrally reviewed PFS. The secondary endpoints include overall survival, quality of life, objective response rate, and safety. A total of 200 patients will be randomized 1:1 to receive oral erlotinib (150 mg daily) plus bevacizumab (15 mg/kg, intravenously, on day 1 of every 21-day cycle) or erlotinib alone, until objective disease progression or unacceptable toxicity or the patient's or physician's motivated decision to stop the treatment. CONCLUSION If the primary endpoint of PFS is met, the erlotinib plus bevacizumab combination will be confirmed as the best first-line treatment for patients with advanced NSCLC harboring activating EGFR mutations.
Collapse
Affiliation(s)
- Cesare Gridelli
- Division of Medical Oncology, S.G. Moscati Hospital, Avellino, Italy.
| | - Antonio Rossi
- Division of Medical Oncology, S.G. Moscati Hospital, Avellino, Italy
| | - Fortunato Ciardiello
- Division of Medical Oncology and Hematology, Second University of Naples, Naples, Italy
| | - Filippo De Marinis
- Division of Thoracic Oncology, European Institute of Oncology, Milan, Italy
| | - Lucio Crinò
- Division of Medical Oncology, University of Perugia, Perugia, Italy
| | - Alessandro Morabito
- Thoraco-Pulmonary Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"- IRCCS, Naples, Italy
| | - Floriana Morgillo
- Division of Medical Oncology and Hematology, Second University of Naples, Naples, Italy
| | - Agnese Montanino
- Thoraco-Pulmonary Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"- IRCCS, Naples, Italy
| | - Gennaro Daniele
- Clinical Trials Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"- IRCCS, Naples, Italy
| | - Maria Carmela Piccirillo
- Clinical Trials Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"- IRCCS, Naples, Italy
| | - Nicola Normanno
- Cellular Biology and Biotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori Fondazione Giovanni Pascale, IRCCS, Naples, Italy
| | - Ciro Gallo
- Medical Statistics, Second University of Naples, Naples, Italy
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"- IRCCS, Naples, Italy
| |
Collapse
|
7
|
Chen L, Martino V, Dombkowski A, Williams T, West-Mays J, Gage PJ. AP-2β Is a Downstream Effector of PITX2 Required to Specify Endothelium and Establish Angiogenic Privilege During Corneal Development. Invest Ophthalmol Vis Sci 2016; 57:1072-81. [PMID: 26968737 PMCID: PMC4790471 DOI: 10.1167/iovs.15-18103] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 02/03/2016] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The homeodomain transcription factor, PITX2, is at the apex of a genetic pathway required for corneal development, but the critical effector genes regulated by the PITX2 remain unknown. The purpose of this study was to discover and validate PITX2-dependent mechanisms required for specifying cell lineages and establishing angiogenic privilege within the developing cornea. METHODS Microarrays were used to compare gene expression in corneas isolated from temporal Pitx2 knockout embryos and control littermates. Quantitative RT-PCR and immunohistochemistry was used to further validate Tfap2b expression differences in Pitx2 knockout versus control corneas. In situ hybridization and protein immunohistochemistry were used to assay eyes of a Tfap2b allelic series of embryos to identify differentiated cellular lineages in the cornea, blood vessel endothelium, or lymphatic vessel endothelium. RESULTS We show that PITX2 is required for the expression of Tfap2b, encoding the AP-2β transcription factor, in the neural crest during corneal development. Markers of differentiated corneal epithelium and stroma are expressed in the absence of AP-2β. In contrast, markers of differentiated corneal endothelium are not expressed in the absence of AP-2β. Endomucin+ blood vessels are present throughout the developing corneal stroma in the absence of AP-2β, whereas LYVE1+ lymphatic vessels are not found. CONCLUSIONS The AP-2β transcription factor is an important effector of PITX2 function during corneal development, required for differentiation of corneal endothelium and establishment of angiogenic privilege. Unlike PITX2, AP-2β is not required for the early expression of available lineage specific markers for the corneal epithelium and stroma during embryogenesis, nor establishment of lymphangiogenic privilege. Therefore, additional PITX2-dependent factors likely regulate these latter processes during embryonic development. These results extend our understanding of the genetic mechanisms regulating cornea development.
Collapse
Affiliation(s)
- Lisheng Chen
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Vanessa Martino
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alan Dombkowski
- Department of Pediatrics, Wayne State University, Detroit, Michigan, United States
| | - Trevor Williams
- Department of Craniofacial Biology and Department of Cell and Developmental Biology, University of Colorado–Denver, Denver, Colorado, United States
| | - Judith West-Mays
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Philip J. Gage
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
8
|
Botelho MC, Soares R, Alves H. Progesterone in Breast Cancer Angiogenesis. SM JOURNAL OF REPRODUCTIVE HEALTH & INFERTILITY 2015; 1:1001. [PMID: 26640826 PMCID: PMC4667742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The involvement of steroid hormones in breast carcinogenesis is well established. Recent evidence suggests that angiogenesis can be regulated by hormones. Both oestrogen and progesterone have been implicated in the angiogenic process of hormone-dependent cancers, such as breast cancer. Vascular Endothelial Growth Factor (VEGF) is a growth factor involved in angiogenesis in breast cancer that is up-regulated by estrogens. In our study we evaluated the role of progesterone in the expression of this angiogenic growth factor commonly up-regulated in breast cancer. Our findings indicate that progesterone activates an angiogenic pathway involving VEGF stimulation. The elucidation of specific angiogenic pathways promoted by progesterone can raise new therapeutic targets at least in a subset of breast cancers responsive to progesterone.
Collapse
Affiliation(s)
- Monica C Botelho
- Unit of Health Promotion and Chronic Diseases, National Institute of Health (INSA) Dr. Ricardo Jorge, Porto, Portugal
| | - Raquel Soares
- IPATIMUP, Institute of Pathology and Molecular Immunology of the University of Porto, Portugal
| | - Helena Alves
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| |
Collapse
|
9
|
Pabla B, Bissonnette M, Konda VJ. Colon cancer and the epidermal growth factor receptor: Current treatment paradigms, the importance of diet, and the role of chemoprevention. World J Clin Oncol 2015; 6:133-141. [PMID: 26468449 PMCID: PMC4600187 DOI: 10.5306/wjco.v6.i5.133] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/23/2015] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer represents the third most common and the second deadliest type of cancer for both men and women in the United States claiming over 50000 lives in 2014. The 5-year survival rate for patients diagnosed with metastatic colon and rectal cancer is < 15%. Early detection and more effective treatments are urgently needed to reduce morbidity and mortality of patients afflicted with this disease. Here we will review the risk factors and current treatment paradigms for colorectal cancer, with an emphasis on the role of chemoprevention as they relate to epidermal growth factor receptor (EGFR) blockade. We will discuss how various EGFR ligands are upregulated in the presence of Western diets high in saturated and N-6 polyunsaturated fats. We will also outline the various mechanisms of EGFR inhibition that are induced by naturally occurring chemopreventative agents such as ginseng, green tea, and curcumin. Finally, we will discuss the current role of targeted chemotherapy in colon cancer and outline the limitations of our current treatment options, describing mechanisms of resistance and escape.
Collapse
|
10
|
Chen Q, Chen P, Pang X, Hu Y, Zhang Y. Adrenomedullin Up-regulates the Expression of Vascular Endothelial Growth Factor in Epithelial Ovarian Carcinoma Cells via JNK/AP-1 Pathway. Int J Gynecol Cancer 2015; 25:953-60. [PMID: 26098087 PMCID: PMC4485736 DOI: 10.1097/igc.0000000000000465] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Adrenomedullin (AM), a potent vasodilator peptide, presents in various kinds of tumors and promotes angiogenesis. We have previously reported that AM is expressed in epithelial ovarian carcinoma tissue. Here, we investigated the hypothesis that AM might regulate production of vascular endothelial growth factor (VEGF) in epithelial ovarian carcinoma and further promote angiogenic processes. METHODS The messenger RNA expression of VEGF in human epithelial ovarian carcinoma cells (HO-8910) was examined by real-time polymerase chain reaction. Transcriptional control was analyzed by transient transfection assay of VEGF promoter-luciferase hybrid genes and chromatin immunoprecipitation assay. Activation of c-Jun N-terminal kinase (JNK) was detected by Western blotting. The formation of capillarylike structures by EA.hy926 cells cocultured with HO-8910 cells on Matrigel was also studied. RESULTS We found that in HO-8910 cells, AM (10⁻¹⁰ to 10⁻⁷ mol/L) enhanced VEGF messenger RNA expression in a time- and concentration-dependent manner, as well as promoter activity. Furthermore, JNK was activated by AM stimulation. The AM-induced increase in VEGF expression was significantly attenuated by SP600125, a specific JNK inhibitor. Chromatin immunoprecipitation assay and promoter activity analysis showed that VEGF expression induced by AM required the activator protein 1 motif on the VEGF promoter. In an in vitro angiogenesis system for endothelial cells (EA.hy926) cocultured with HO-8910 cells, we observed that the addition of AM stimulated endothelial cell tube formation, which could be abolished by VEGF neutralizing antibody. CONCLUSIONS Our findings suggest that the JNK/Activator protein 1 pathway is involved in AM-induced VEGF expression in HO-8910 cells.
Collapse
MESH Headings
- Adrenomedullin/pharmacology
- Antihypertensive Agents/pharmacology
- Blotting, Western
- Carcinoma, Ovarian Epithelial
- Chromatin Immunoprecipitation
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- JNK Mitogen-Activated Protein Kinases/genetics
- JNK Mitogen-Activated Protein Kinases/metabolism
- Neoplasms, Glandular and Epithelial/drug therapy
- Neoplasms, Glandular and Epithelial/metabolism
- Neoplasms, Glandular and Epithelial/pathology
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Regulatory Elements, Transcriptional/drug effects
- Regulatory Elements, Transcriptional/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Transcription Factor AP-1/genetics
- Transcription Factor AP-1/metabolism
- Transcriptional Activation
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Qingqing Chen
- *Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; and †Department of Gynecology and Obstetrics, Jin hua Municipal Central Hosptial, Jin hua, Zhejiang, China
| | - Pan Chen
- *Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; and †Department of Gynecology and Obstetrics, Jin hua Municipal Central Hosptial, Jin hua, Zhejiang, China
| | - Xiaoyan Pang
- *Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; and †Department of Gynecology and Obstetrics, Jin hua Municipal Central Hosptial, Jin hua, Zhejiang, China
| | - Yanling Hu
- *Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; and †Department of Gynecology and Obstetrics, Jin hua Municipal Central Hosptial, Jin hua, Zhejiang, China
| | - Yi Zhang
- *Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; and †Department of Gynecology and Obstetrics, Jin hua Municipal Central Hosptial, Jin hua, Zhejiang, China
| |
Collapse
|
11
|
Fibromodulin Enhances Angiogenesis during Cutaneous Wound Healing. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2015; 2:e275. [PMID: 25587509 PMCID: PMC4292257 DOI: 10.1097/gox.0000000000000243] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/06/2014] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Fibromodulin (FMOD) plays a critical role in the wound-healing process. Our previous studies revealed that FMOD deficiency led to marked alterations in adult wound healing characterized by delayed dermal cell migration, postponed wound closure, and increased scar formation, all accompanied by impeded angiogenesis. Therefore, the aim of this study was to reveal the effect of FMOD on angiogenesis during the wound-healing process. Methods: In vivo angiogenic effects of FMOD were assessed by a chick embryo chorioallantoic membrane assay, a Matrigel (BD Bioscience, Franklin Lakes, N.J.) plug implant assay, and rodent primary closure wound models. In vitro angiogenic effects of FMOD were recorded by cell invasion and dimensional and topological parameters of human umbilical vein endothelial cells. Results: We provided evidence that FMOD significantly enhanced vascularization: first, FMOD boosted blood vessel formation on the chorioallantoic membrane; second, FMOD markedly stimulated capillary infiltration into Matrigel plugs subcutaneously implanted in adult mice; and finally, FMOD robustly promoted angiogenesis in multiple adult rodent cutaneous wound models. Furthermore, FMOD administration restored the vascularity of fmod−/− mouse wounds. In support of this, FMOD endorsed an angiogenesis-favored microenvironment in adult rodent wounds not only by upregulating angiogenic genes but also by downregulating angiostatic genes. In addition, FMOD significantly enhanced human umbilical vein endothelial cell invasion and tube-like structure formation in vitro. Conclusions: Altogether, we demonstrated that in addition to reducing scar formation, FMOD also promotes angiogenesis. As blood vessels organize and regulate wound healing, its potent angiogenic properties will further expand the clinical application of FMOD for cutaneous healing of poorly vascularized wounds.
Collapse
|
12
|
Bae ON, Noh M, Chun YJ, Jeong TC. Keratinocytic vascular endothelial growth factor as a novel biomarker for pathological skin condition. Biomol Ther (Seoul) 2015; 23:12-8. [PMID: 25593638 PMCID: PMC4286744 DOI: 10.4062/biomolther.2014.102] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/01/2014] [Accepted: 10/07/2014] [Indexed: 12/21/2022] Open
Abstract
Skin is an emerging target tissue in pharmaceutical and cosmetic science. Safety assessment for dermal toxicity is a critical step for development of topically applicable pharmaceutical agents and ingredients in cosmetics. Urgent needs exist to set up toxicity testing methods for dermal safety, and identification of novel biomarkers for pathological cutaneous alteration is highly required. Here we will discuss if vascular endothelial growth factor (VEGF) has a potential as a biomarker for dermal impairment. Experimental and clinical evidences for induction of keratinocytic VEGF under pathological conditions will be reviewed.
Collapse
Affiliation(s)
- Ok-Nam Bae
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791
| | - Minsoo Noh
- Collge of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 151-742
| | - Young-Jin Chun
- College of Pharmacy, Chung-Ang University, Seoul 156-756
| | - Tae Cheon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea
| |
Collapse
|
13
|
Su W, Xia J, Chen X, Xu M, Nie L, Chen N, Gong J, Li X, Zhou Q. Ectopic expression of AP-2α transcription factor suppresses glioma progression. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:8666-8674. [PMID: 25674231 PMCID: PMC4314016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 11/26/2014] [Indexed: 06/04/2023]
Abstract
The transcriptional factor AP-2α is a tumor suppressor gene and is downregulated in various neoplasms including glioma. Although the level of AP-2α is negatively associated with the grade of human glioma, the specific functions of AP-2α in glioma are still unknown. In this study, we experimentally showed that artificial overexpression of AP-2α in glioma T98G and U251 cells significantly downregulated the mRNA levels of Bcl-xl, Bcl-2, c-IAP2 and survivin, together with upregulation of the Hrk mRNA levels. Reintroduction of AP-2α also induced downregulation of the protein levels of survivin and VEGF in glioma cells. In biological assays with T98G and U251 cells, AP-2α reduced tumor cell growth, increased cell death, attenuated cell migration and endothelial tube formation. The AP-2α transcription factor may play an important role in suppressing glioma progression.
Collapse
Affiliation(s)
- Wenjing Su
- Laboratory of Pathology, State Key Laboratory of Biotherapy and Department of Pathology, West China Hospital, West China Medical School, Sichuan UniversityChengdu 610041, China
| | - Juan Xia
- Laboratory of Pathology, State Key Laboratory of Biotherapy and Department of Pathology, West China Hospital, West China Medical School, Sichuan UniversityChengdu 610041, China
- Suining Central HospitalSuining 629000, China
| | - Xueqin Chen
- Laboratory of Pathology, State Key Laboratory of Biotherapy and Department of Pathology, West China Hospital, West China Medical School, Sichuan UniversityChengdu 610041, China
| | - Miao Xu
- Laboratory of Pathology, State Key Laboratory of Biotherapy and Department of Pathology, West China Hospital, West China Medical School, Sichuan UniversityChengdu 610041, China
| | - Ling Nie
- Laboratory of Pathology, State Key Laboratory of Biotherapy and Department of Pathology, West China Hospital, West China Medical School, Sichuan UniversityChengdu 610041, China
| | - Ni Chen
- Laboratory of Pathology, State Key Laboratory of Biotherapy and Department of Pathology, West China Hospital, West China Medical School, Sichuan UniversityChengdu 610041, China
| | - Jing Gong
- Laboratory of Pathology, State Key Laboratory of Biotherapy and Department of Pathology, West China Hospital, West China Medical School, Sichuan UniversityChengdu 610041, China
| | - Xinglan Li
- Laboratory of Pathology, State Key Laboratory of Biotherapy and Department of Pathology, West China Hospital, West China Medical School, Sichuan UniversityChengdu 610041, China
| | - Qiao Zhou
- Laboratory of Pathology, State Key Laboratory of Biotherapy and Department of Pathology, West China Hospital, West China Medical School, Sichuan UniversityChengdu 610041, China
| |
Collapse
|
14
|
Murad S. Toll-like receptor 4 in inflammation and angiogenesis: a double-edged sword. Front Immunol 2014; 5:313. [PMID: 25071774 PMCID: PMC4083339 DOI: 10.3389/fimmu.2014.00313] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/22/2014] [Indexed: 01/04/2023] Open
Affiliation(s)
- Sheeba Murad
- Molecular Immunology Lab, Health Care Biotech Department, Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology , Islamabad , Pakistan
| |
Collapse
|
15
|
Pradhan R, Rajput S, Mandal M, Mitra A, Das S. Frequency dependent impedimetric cytotoxic evaluation of anticancer drug on breast cancer cell. Biosens Bioelectron 2014; 55:44-50. [DOI: 10.1016/j.bios.2013.11.060] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 11/08/2013] [Accepted: 11/20/2013] [Indexed: 11/27/2022]
|
16
|
S0941: a phase 2 SWOG study of sorafenib and erlotinib in patients with advanced gallbladder carcinoma or cholangiocarcinoma. Br J Cancer 2014; 110:882-7. [PMID: 24423918 PMCID: PMC3929880 DOI: 10.1038/bjc.2013.801] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 11/04/2013] [Accepted: 12/02/2013] [Indexed: 12/15/2022] Open
Abstract
Background: Gallbladder cancers and cholangiocarcinomas make up a heterogenous group of tumours with a poor prognosis in advanced stages. On the basis of evidence of dysregulation of the epidermal growth factor receptor, vascular endothelial growth factor and mitogen-activated protein kinase pathways in biliary cancers, we performed a phase 2 trial of sorafenib and erlotinib in patients with advanced biliary cancers. Methods: Eligible patients were previously untreated in the advanced setting with adequate hepatic and bone marrow function. Sorafenib and erlotinib were administered continuously at 400 mg BID and 100 mg daily, respectively. Results: Thirty-four eligible patients were recruited. The study was terminated after the first stage of accrual owing to failure to meet the predetermined number of patients who were alive and progression free at 4 months. There were two unconfirmed partial responses (6%, 95% CI: 1–20%), with a median progression-free survival of 2 months (95% CI: 2–3), and median overall survival of 6 months (95% CI: 3–8 months). Grade 3 and 4 adverse events included hypertension, AST/ALT increase, bilirubin increase, diarrhoea, hypokalaemia, hypophosphatemia and rash. Conclusions: Despite compelling preclinical rationale, the combination of sorafenib and erlotinib does not have promising clinical activity in an unselected population of patients with biliary cancers. Improved patient selection based on tumour biology and molecular markers is critical for future evaluation of targeted therapies in this disease.
Collapse
|
17
|
Vigneault F, Guérin SL. Regulation of gene expression: probing DNA–protein interactionsin vivoandin vitro. Expert Rev Proteomics 2014; 2:705-18. [PMID: 16209650 DOI: 10.1586/14789450.2.5.705] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tremendous efforts have been put together over the last several years to complete the entire sequencing of the human genome. As we enter the proteomic era, when the major aim is understanding which gene encodes which protein, the time has also come to identify their precise function inside the astonishing signaling network required to accomplish all cellular functions. Understanding when, why and how a gene is expressed has now become a necessity toward identifying all the regulatory pathways that mediate cellular processes such as differentiation, migration, replication, DNA repair and apoptosis. Regulation of gene transcription is a process that is primarily under the influence of nuclear-located transcription factors. Consequently, identifying which protein activates or represses a specific gene is a prerequisite for understanding cell fate and function. The current state of, and recent advances in, transcriptional regulation approaches are reviewed here, with special emphasis on new technologies required when probing for DNA-protein interactions. This review explores different strategies aimed at identifying both the regulatory sequences of any given gene and the trans-acting regulatory factors that recognize these elements as their target sites in the nucleus. Ongoing developments in the fields of nanotechnology, RNA silencing and protein modeling toward the investigation of DNA-protein interactions and their relevance in the battle against cancer are discussed.
Collapse
Affiliation(s)
- Francois Vigneault
- Laboratoire d'Endocrinologie Moléculaire et Oncologique, Centre de recherche du CHUL (CHUQ), Sainte-Foy, Québec, G1V 4G2, Canada.
| | | |
Collapse
|
18
|
Ensinger C, Sterlacci W. Implications of EGFR PharmDx™ Kit for cetuximab eligibility. Expert Rev Mol Diagn 2014; 8:141-8. [DOI: 10.1586/14737159.8.2.141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Pradhan R, Rajput S, Mandal M, Mitra A, Das S. Electric cell–substrate impedance sensing technique to monitor cellular behaviours of cancer cells. RSC Adv 2014. [DOI: 10.1039/c3ra45090b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
20
|
MacDonald ST, Bamforth SD, Bragança J, Chen CM, Broadbent C, Schneider JE, Schwartz RJ, Bhattacharya S. A cell-autonomous role of Cited2 in controlling myocardial and coronary vascular development. Eur Heart J 2013; 34:2557-2565. [PMID: 22504313 PMCID: PMC3748368 DOI: 10.1093/eurheartj/ehs056] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 01/30/2012] [Accepted: 02/16/2012] [Indexed: 02/06/2023] Open
Abstract
AIMS Myocardial development is dependent on concomitant growth of cardiomyocytes and a supporting vascular network. The coupling of myocardial and coronary vascular development is partly mediated by vascular endothelial growth factor (VEGFA) signalling and additional unknown mechanisms. We examined the cardiomyocyte specific role of the transcriptional co-activator Cited2 on myocardial microstructure and vessel growth, in relation to Vegfa expression. METHODS AND RESULTS A cardiomyocyte-specific knockout of mouse Cited2 (Cited2(Nkx)) was analysed using magnetic resonance imaging and histology. Ventricular septal defects and significant compact layer thinning (P < 0.02 at right ventricular apex, P < 0.009 at the left ventricular apex in Cited2(Nkx) vs. controls, n = 11 vs. n = 7, respectively) were found. This was associated with a significant decrease in the number of capillaries to larger vessels (ratio 1.56 ± 0.56 vs. 3.25 ± 1.63, P = 2.7 × 10(-6) Cited2(Nkx) vs. controls, n = 11 vs. n = 7, respectively) concomitant with a 1.5-fold reduction in Vegfa expression (P < 0.02, Cited2(Nkx) vs. controls, n = 12 vs. n = 12, respectively). CITED2 was subsequently found at the Vegfa promoter in mouse embryonic hearts using chromatin immunoprecipitation, and moreover found to stimulate human VEGFA promoter activity in cooperation with TFAP2 transcription factors in transient transfection assays. There was no change in the myocardial expression of the left-right patterning gene Pitx2c, a previously known target of CITED2. CONCLUSIONS This study delineates a novel cell-autonomous role of Cited2 in regulating VEGFA transcription and the development of myocardium and coronary vasculature in the mouse. We suggest that coupling of myocardial and coronary growth in the developing heart may occur in part through a Cited2→Vegfa pathway.
Collapse
Affiliation(s)
- Simon T. MacDonald
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Simon D. Bamforth
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - José Bragança
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Chiann-Mun Chen
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Carol Broadbent
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Jürgen E. Schneider
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Robert J. Schwartz
- Institute of Biosciences and Technology, Texas A&M Health Science Centre, Houston, TX 77030-3498, USA
| | - Shoumo Bhattacharya
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| |
Collapse
|
21
|
Pang L, Zhang Y, Yu Y, Zhang S. Resistin promotes the expression of vascular endothelial growth factor in ovary carcinoma cells. Int J Mol Sci 2013; 14:9751-66. [PMID: 23652833 PMCID: PMC3676810 DOI: 10.3390/ijms14059751] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/18/2013] [Accepted: 04/24/2013] [Indexed: 12/16/2022] Open
Abstract
Resistin is a novel hormone that is secreted by human adipocytes and mononuclear cells and is associated with obesity, insulin resistance and inflammation. Recently, resistin has been postulated to play a role in angiogenesis. Here, we investigated the hypothesis that resistin regulates ovary carcinoma production of vascular endothelial growth factor (VEGF) and the angiogenic processes. We found that in human ovarian epithelial carcinoma cells (HO-8910), resistin (10–150 ng/mL) enhanced both VEGF protein and mRNA expression in a time- and concentration-dependent manner, as well as promoter activity. Furthermore, resistin enhanced DNA-binding activity of Sp1 with VEGF promoter in a PI3K/Akt-dependent manner. PI3K/Akt activated by resistin led to increasing interaction with Sp1, triggering a progressive phosphorylation of Sp1 on Thr453 and Thr739, resulting in the upregulation of VEGF expression. In an in vitro angiogenesis system for endothelial cells (EA.hy926) co-cultured with HO-8910 cells, we observed that the addition of resistin stimulated endothelial cell tube formation, which could be abolished by VEGF neutralizing antibody. Our findings suggest that the PI3K/Akt-Sp1 pathway is involved in resistin-induced VEGF expression in HO-8910 cells and indicates that antiangiogenesis therapy may be beneficial treatment against ovarian epithelial carcinoma, especially in obese patients.
Collapse
Affiliation(s)
- Li Pang
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang 110004, Liaoning, China; E-Mails: (L.P.); (Y.Y.)
| | - Yi Zhang
- Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China; E-Mail:
| | - Yu Yu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang 110004, Liaoning, China; E-Mails: (L.P.); (Y.Y.)
| | - Shulan Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang 110004, Liaoning, China; E-Mails: (L.P.); (Y.Y.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel./Fax: +86-24-966-151-41211
| |
Collapse
|
22
|
Terrasi M, Bazan V, Caruso S, Insalaco L, Amodeo V, Fanale D, Corsini LR, Contaldo C, Mercanti A, Fiorio E, Lo Re G, Cicero G, Surmacz E, Russo A. Effects of PPARγ agonists on the expression of leptin and vascular endothelial growth factor in breast cancer cells. J Cell Physiol 2013; 228:1368-74. [DOI: 10.1002/jcp.24295] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 11/27/2012] [Indexed: 11/11/2022]
|
23
|
Schwarzenbach H, Chakrabarti G, Paust HJ, Mukhopadhyay AK. Gonadotropin-Mediated Regulation of the Murine VEGF Expression in MA-10 Leydig Cells. ACTA ACUST UNITED AC 2013; 25:128-39. [PMID: 14662796 DOI: 10.1002/j.1939-4640.2004.tb02768.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Presence of vascular endothelial growth factor (VEGF) is not only limited to cells directly involved in angiogenesis but has also been demonstrated in steroidogenic cells like testicular Leydig cells. Because Leydig cells are subjected to regulation by gonadotropic hormones and produce steroid hormones, we have investigated here the effects of human chorionic gonadotropin (hCG) or steroid hormones on VEGF expression in cultured mouse tumor Leydig cells (MA-10 cells) and have then analyzed the underlying molecular mechanisms. Northern blot analysis and enzyme-linked immunosorbent assays revealed increases in VEGF mRNA and protein levels, respectively, over 3-20 hours in MA-10 cells after stimulation with hCG or 8-Br-cAMP. Although MA-10 cells lack the classical progesterone receptor, progesterone was able to stimulate VEGF expression. Promoter analyses and antibody supershift experiments suggested that the proximal region is able to constitutively bind the transcription factors Sp1 and Sp3. Mutations of 2 potential Sp1 binding sites in the proximal region showed the requirement of these motifs for stimulation of VEGF by hCG and 8-Br-cAMP. The distal cytosine-rich sequence interacts with so far-unidentified faster migrating factors. Following stimulation with hCG or 8-Br-cAMP, the binding of these proteins was increased in the complexes formed in the proximal and distal regions. VEGF expression in Leydig cells is regulated by gonadotropin via a cAMP-dependent mechanism, and the transcription factors Sp1 and Sp3 appear to be involved in the activation of the promoter. Progesterone also appears to play a role in the regulation of VEGF, acting presumably via a nonconventional receptor that remains to be characterized yet.
Collapse
Affiliation(s)
- Heidi Schwarzenbach
- Institute for Hormone and Fertility Research, University of Hamburg, Grandweg 64, D-22529 Hamburg, Germany
| | | | | | | |
Collapse
|
24
|
Transcriptional repression of VEGF by ZNF24: mechanistic studies and vascular consequences in vivo. Blood 2012; 121:707-15. [PMID: 23212515 DOI: 10.1182/blood-2012-05-433045] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
VEGF is a key regulator of normal and pathologic angiogenesis. Although many trans-activating factors of VEGF have been described, the transcriptional repression of VEGF remains much less understood. We have previously reported the identification of a SCAN domain-containing C2H2 zinc finger protein, ZNF24, that represses the transcription of VEGF. In the present study, we identify the mechanism by which ZNF24 represses VEGF transcription. Using reporter gene and electrophoretic mobility shift assays, we identify an 11-bp fragment of the proximal VEGF promoter as the ZNF24-binding site that is essential for ZNF24-mediated repression. We demonstrate in 2 in vivo models the potent inhibitory effect of ZNF24 on the vasculature. Expression of human ZNF24 induced in vivo vascular defects consistent with those induced by VEGF knockdown using a transgenic zebrafish model. These defects could be rescued by VEGF overexpression. Overexpression of ZNF24 in human breast cancer cells also inhibited tumor angiogenesis in an in vivo tumor model. Analyses of human breast cancer tissues showed that ZNF24 and VEGF levels were inversely correlated in malignant compared with normal tissues. These data demonstrate that ZNF24 represses VEGF transcription through direct binding to an 11-bp fragment of the VEGF proximal promoter and that it functions as a negative regulator of tumor growth by inhibiting angiogenesis.
Collapse
|
25
|
Endo T, Arimura Y, Adachi Y, Mita H, Yamashita K, Yamamoto H, Shinomura Y, Ishii Y. A case of Ménétrier's disease without Helicobacter pylori infection. Dig Endosc 2012; 24:275-279. [PMID: 22725115 DOI: 10.1111/j.1443-1661.2012.01242.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ménétrier's disease (MD) is a rare, acquired, premalignant disorder of the stomach characterized by enlarged gastric folds with foveolar hyperplasia, the phenotype of antralization of gastric glands, hypochlorhydria and hypoproteinemia. The etiology of MD is unknown, but both increased signaling by transforming growth factor-α and infection with Helicobacter pylori (H. pylori) have been implicated. Here, a case involving 70-year-old man who lost weight after developing anorexia and diarrhea is reported. He was diagnosed as MD without H. pylori infection, and in spite of intensive care, he died 40 days after admission. An autopsy confirmed MD. Immunohistochemistry revealed overexpression of transforming growth factor-α in the foveolar region of the gastric mucosa. The autopsy also distinguished this H. pylori-negative MD from hyperplastic polyp of the stomach, which is important in clarifying the entity of H. pylori-negative MD.
Collapse
Affiliation(s)
- Takao Endo
- Department of Gastroenterology, Sapporo Shirakaba-dai Hospital, Sapporo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Li Z, Yao L, Li J, Zhang W, Wu X, Liu Y, Lin M, Su W, Li Y, Liang D. Celastrol nanoparticles inhibit corneal neovascularization induced by suturing in rats. Int J Nanomedicine 2012; 7:1163-73. [PMID: 22419865 PMCID: PMC3298384 DOI: 10.2147/ijn.s27860] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Celastrol, a traditional Chinese medicine, is widely used in anti-inflammation and anti-angiogenesis research. However, the poor water solubility of celastrol restricts its further application. This paper aims to study the effect of celastrol nanoparticles (CNPs) on corneal neovascularization (CNV) and determine the possible mechanism. METHODS To improve the hydrophilicity of celastrol, celastrol-loaded poly(ethylene glycol)-block-poly(ɛ-caprolactone) nanopolymeric micelles were developed. The characterization of CNPs was measured by dynamic light scattering and transmission electron microscopy analysis. Celastrol loading content and release were assessed by ultraviolet-visible analysis and high performance liquid chromatography, respectively. In vitro, human umbilical vein endothelial cell proliferation and capillary-like tube formation were assayed. In vivo, suture-induced CNV was chosen to evaluate the effect of CNPs on CNV in rats. Immunohistochemistry for CD68 assessed the macrophage infiltration of the cornea on day 6 after surgery. Real-time quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay were used to evaluate the messenger ribonucleic acid and protein levels, respectively, of vascular endothelial growth factor, matrix metalloproteinase 9, and monocyte chemoattractant protein 1 in the cornea. RESULTS The mean diameter of CNPs with spherical shape was 48 nm. The celastrol loading content was 7.36%. The release behavior of CNPs in buffered solution (pH 7.4) showed a typical two-phase release profile. CNPs inhibited the proliferation of human umbilical vein endothelial cells in a dose-independent manner and suppressed the capillary structure formation. After treatment with CNPs, the length and area of CNV reduced from 1.16 ± 0.18 mm to 0.49 ± 0.12 mm and from 7.71 ± 0.94 mm(2) to 2.29 ± 0.61 mm(2), respectively. Macrophage infiltration decreased significantly in the CNP-treated corneas. CNPs reduced the expression of vascular endothelial growth factor, matrix metalloproteinase 9, and monocyte chemoattractant protein 1 in the cornea on day 6 after suturing. CONCLUSION CNPs significantly inhibited suture-induced CNV by suppressing macrophage infiltration and the expression of vascular endothelial growth factor and matrix metalloproteinase 9 in the rat cornea.
Collapse
Affiliation(s)
- Zhanrong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
EGFRvIII promotes glioma angiogenesis and growth through the NF-κB, interleukin-8 pathway. Oncogene 2011; 31:4054-66. [PMID: 22139077 DOI: 10.1038/onc.2011.563] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Sustaining a high growth rate requires tumors to exploit resources in their microenvironment. One example of this is the extensive angiogenesis that is a typical feature of high-grade gliomas. Here, we show that expression of the constitutively active mutant epidermal growth factor receptor, ΔEGFR (EGFRvIII, EGFR*, de2-7EGFR) is associated with significantly higher expression levels of the pro-angiogenic factor interleukin (IL)-8 in human glioma specimens and glioma stem cells. Furthermore, the ectopic expression of ΔEGFR in different glioma cell lines caused up to 60-fold increases in the secretion of IL-8. Xenografts of these cells exhibit increased neovascularization, which is not elicited by cells overexpressing wild-type (wt)EGFR or ΔEGFR with an additional kinase domain mutation. Analysis of the regulation of IL-8 by site-directed mutagenesis of its promoter showed that ΔEGFR regulates its expression through the transcription factors nuclear factor (NF)-κB, activator protein 1 (AP-1) and CCAAT/enhancer binding protein (C/EBP). Glioma cells overexpressing ΔEGFR showed constitutive activation and DNA binding of NF-κB, overexpression of c-Jun and activation of its upstream kinase c-Jun N-terminal kinase (JNK) and overexpression of C/EBPβ. Selective pharmacological or genetic targeting of the NF-κB or AP-1 pathways efficiently blocked promoter activity and secretion of IL-8. Moreover, RNA interference-mediated knock-down of either IL-8 or the NF-κB subunit p65, in ΔEGFR-expressing cells attenuated their ability to form tumors and to induce angiogenesis when injected subcutaneously into nude mice. On the contrary, the overexpression of IL-8 in glioma cells lacking ΔEGFR potently enhanced their tumorigenicity and produced highly vascularized tumors, suggesting the importance of this cytokine and its transcription regulators in promoting glioma angiogenesis and tumor growth.
Collapse
|
28
|
Reumann MK, Strachna O, Yagerman S, Torrecilla D, Kim J, Doty SB, Lukashova L, Boskey AL, Mayer-Kuckuk P. Loss of transcription factor early growth response gene 1 results in impaired endochondral bone repair. Bone 2011; 49:743-52. [PMID: 21726677 PMCID: PMC3169183 DOI: 10.1016/j.bone.2011.06.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 06/14/2011] [Accepted: 06/17/2011] [Indexed: 01/24/2023]
Abstract
Transcription factors that play a role in ossification during development are expected to participate in postnatal fracture repair since the endochondral bone formation that occurs in embryos is recapitulated during fracture repair. However, inherent differences exist between bone development and fracture repair, including a sudden disruption of tissue integrity followed by an inflammatory response. This raises the possibility that repair-specific transcription factors participate in bone healing. Here, we assessed the consequence of loss of early growth response gene 1 (EGR-1) on endochondral bone healing because this transcription factor has been shown to modulate repair in vascularized tissues. Model fractures were created in ribs of wild type (wt) and EGR-1(-/-) mice. Differences in tissue morphology and composition between these two animal groups were followed over 28 post fracture days (PFDs). In wt mice, bone healing occurred in healing phases characteristic of endochondral bone repair. A similar healing sequence was observed in EGR-1(-/-) mice but was impaired by alterations. A persistent accumulation of fibrin between the disconnected bones was observed on PFD7 and remained pronounced in the callus on PFD14. Additionally, the PFD14 callus was abnormally enlarged and showed increased deposition of mineralized tissue. Cartilage ossification in the callus was associated with hyper-vascularity and -proliferation. Moreover, cell deposits located in proximity to the callus within skeletal muscle were detected on PFD14. Despite these impairments, repair in EGR-1(-/-) callus advanced on PFD28, suggesting EGR-1 is not essential for healing. Together, this study provides genetic evidence that EGR-1 is a pleiotropic regulator of endochondral fracture repair.
Collapse
Affiliation(s)
- Marie K. Reumann
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York
| | - Olga Strachna
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York
| | - Sarah Yagerman
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York
| | - Daniel Torrecilla
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York
| | - Jihye Kim
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York
| | - Steven B. Doty
- Analytical Microscopy Laboratory, Hospital for Special Surgery, New York
| | | | - Adele L. Boskey
- Mineralized Tissue Laboratory, Hospital for Special Surgery, New York
| | - Philipp Mayer-Kuckuk
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York
- Corresponding author: Dr. Philipp Mayer-Kuckuk, Caspary Research Building, Rm. 623, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021, USA, Fax:(212) 774 7877,
| |
Collapse
|
29
|
KHAN MISBAHH, ALAM MURAD, YOO SIMON. Epidermal Growth Factor Receptor Inhibitors in the Treatment of Nonmelanoma Skin Cancers. Dermatol Surg 2011; 37:1199-209. [DOI: 10.1111/j.1524-4725.2011.02038.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
30
|
Ray A, Dhar S, Ray BK. Control of VEGF expression in triple-negative breast carcinoma cells by suppression of SAF-1 transcription factor activity. Mol Cancer Res 2011; 9:1030-41. [PMID: 21665940 DOI: 10.1158/1541-7786.mcr-10-0598] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Angiogenesis plays a significant role in cancer by providing increased blood supply to the affected tissues and thus bringing in growth factors, cytokines, and various nutrients for tumor growth. VEGF is the most prominent angiogenic agent that is markedly induced in cancer. Induction of VEGF has been widely studied but as cancer cells are quite adept at acquiring new alternative processes to circumvent surrounding environmental pressures, our understanding of the molecular mechanisms regulating VEGF expression in cancer, especially in triple-negative breast cancer cells, remains incomplete. Here, we present evidence of a novel mode of VEGF induction in triple-negative MDA-MB-231 breast cancer cells that is regulated by serum amyloid A activating factor 1 (SAF-1) transcription factor. Inhibition of SAF-1 by antisense short hairpin RNA profoundly reduces VEGF expression along with reduction in endothelial cell proliferation and migration. By both in vitro and in vivo molecular studies, we show that the effect of SAF-1 is mediated through its direct interaction with the VEGF promoter. In correlation, DNA-binding activity of SAF-1 is found to be significantly higher in MDA-MB-231 breast cancer cells. Examination of several breast cancer samples further revealed that SAF-1 is overexpressed in clinical breast cancer tissues. Taken together, these findings reveal that SAF-1 is a hitherto unrecognized participant in inducing VEGF expression in triple-negative breast cancer cells, an aggressive form of breast cancer that currently lacks effective treatment options. Suppression of SAF-1 activity in these cells can inhibit VEGF expression, providing a possible new method to control angiogenesis.
Collapse
Affiliation(s)
- Alpana Ray
- Department of Veterinary Pathobiology, University of Missouri, 124 Connaway Hall, Columbia, MO 65211, USA.
| | | | | |
Collapse
|
31
|
Jolma A, Taipale J. Methods for Analysis of Transcription Factor DNA-Binding Specificity In Vitro. Subcell Biochem 2011; 52:155-173. [PMID: 21557082 DOI: 10.1007/978-90-481-9069-0_7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Transcription of genes during development and in response to environmental stimuli is determined by genomic DNA sequence. The DNA sequences regulating transcription are read by sequence-specific transcription factors (TFs) that recognize relatively short sequences, generally between four and twenty base pairs in length. Transcriptional regulation generally requires binding of multiple TFs in close proximity to each other. Mechanistic understanding of transcription in an organism thus requires detailed knowledge of binding affinities of all its TFs to all possible DNA sequences, and the co-operative interactions between the TFs. However, very little is known about such co-operative binding interactions, and even the simple TF-DNA binding information exists only for a very small proportion of all TFs - for example, mammals have approximately 1,300-2,000 TFs [1, 2], yet the largest public databases for TF binding specificity, Jaspar and Uniprobe [3, 4] currently list only approximately 500 moderate to high resolution profiles for human or mouse. This lack of knowledge is in part due to the fact that analysis of TF DNA binding has been laborious and expensive. In this chapter, we review methods that can be used to determine binding specificity of TFs to DNA, mainly focusing on recently developed assays that allow high-resolution analysis of TF binding specificity in relatively high throughput.
Collapse
Affiliation(s)
- Arttu Jolma
- Department of Biosciences and Nutrition, SE-171 77, Stockholm, Sweden,
| | | |
Collapse
|
32
|
Zhou G, Hasina R, Wroblewski K, Mankame TP, Doçi CL, Lingen MW. Dual inhibition of vascular endothelial growth factor receptor and epidermal growth factor receptor is an effective chemopreventive strategy in the mouse 4-NQO model of oral carcinogenesis. Cancer Prev Res (Phila) 2010; 3:1493-502. [PMID: 20978113 DOI: 10.1158/1940-6207.capr-10-0135] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Despite recent therapeutic advances, several factors, including field cancerization, have limited improvements in long-term survival for oral squamous cell carcinoma (OSCC). Therefore, comprehensive treatment plans must include improved chemopreventive strategies. Using the 4-nitroquinoline 1-oxide (4-NQO) mouse model, we tested the hypothesis that ZD6474 (Vandetanib, ZACTIMA) is an effective chemopreventive agent. CBA mice were fed 4-NQO (100 μg/mL) in their drinking water for 8 weeks and then randomized to no treatment or oral ZD6474 (25 mg/kg/d) for 24 weeks. The percentage of animals with OSCC was significantly different between the two groups (71% in control and 12% in the ZD6474 group; P ≤ 0.001). The percentage of mice with dysplasia or OSCC was significantly different (96% in the control and 28% in the ZD6474 group; P ≤ 0.001). Proliferation and microvessel density scores were significantly decreased in the ZD6474 group (P ≤ 0.001 for both). Although proliferation and microvessel density increased with histologic progression in control and treatment cohorts, epidermal growth factor receptor and vascular endothelial growth factor receptor-2 phosphorylation was decreased in the treatment group for each histologic diagnosis, including mice harboring tumors. OSCC from ZD6474-treated mice exhibited features of epithelial to mesenchymal transition, as shown by loss E-cadherin and gain of vimentin protein expression. These data suggest that ZD6474 holds promise as an OSCC chemopreventive agent. They further suggest that acquired resistance to ZD6474 may be mediated by the expression of an epithelial to mesenchymal transition phenotype. Finally, the data suggests that this model is a useful preclinical platform to investigate the mechanisms of acquired resistance in the chemopreventive setting.
Collapse
Affiliation(s)
- Guolin Zhou
- Departments of Pathology, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | |
Collapse
|
33
|
Martinelli E, Troiani T, Morgillo F, Rodolico G, Vitagliano D, Morelli MP, Tuccillo C, Vecchione L, Capasso A, Orditura M, De Vita F, Eckhardt SG, Santoro M, Berrino L, Ciardiello F. Synergistic antitumor activity of sorafenib in combination with epidermal growth factor receptor inhibitors in colorectal and lung cancer cells. Clin Cancer Res 2010; 16:4990-5001. [PMID: 20810384 DOI: 10.1158/1078-0432.ccr-10-0923] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE Cancer cell survival, invasion, and metastasis depend on cancer cell proliferation and on tumor-induced angiogenesis. We evaluated the efficacy of the combination of sorafenib and erlotinib or cetuximab. EXPERIMENTAL DESIGN Sorafenib, erlotinib, and cetuximab, alone or in combination, were tested in vitro in a panel of non-small cell lung cancer (NSCLC) and colorectal cancer cell lines and in vivo in H1299 tumor xenografts. RESULTS Epidermal growth factor receptor (EGFR) ligand mRNAs were expressed in all NSCLC and colorectal cancer cell lines with variable levels ranging from 0.4- to 8.1-fold as compared with GEO colorectal cancer cells. Lung cancer cells had the highest levels of vascular endothelial growth factors (VEGF) A, B, and C, and of VEGF receptors as compared with colorectal cancer cells. Combined treatments of sorafenib with erlotinib or cetuximab produced combination index values between 0.02 and 0.5, suggesting a significant synergistic activity to inhibit soft agar colony formation in all cancer cell lines, which was accompanied by a marked blockade in mitogen-activated protein kinase and AKT signals. The in vitro migration of H1299 cells, which expressed high levels of both VEGF ligands and receptors, was inhibited by treatment with sorafenib, and this effect was significantly increased by the combination with anti-EGFR drugs. In nude mice bearing established human H1299 xenografts, treatment with the combination of sorafenib and erlotinib or cetuximab caused a significant tumor growth delay resulting in 70 to 90 days increase in mice median overall survival as compared with single-agent sorafenib treatment. CONCLUSIONS Combination treatment with sorafenib and erlotinib or cetuximab has synergistic antitumor effects in human colorectal and lung cancer cells.
Collapse
Affiliation(s)
- Erika Martinelli
- Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale F Magrassi e A Lanzara, Seconda Università degli Studi di Napoli, Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Melnikova VO, Dobroff AS, Zigler M, Villares GJ, Braeuer RR, Wang H, Huang L, Bar-Eli M. CREB inhibits AP-2alpha expression to regulate the malignant phenotype of melanoma. PLoS One 2010; 5:e12452. [PMID: 20805990 PMCID: PMC2929203 DOI: 10.1371/journal.pone.0012452] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Accepted: 08/04/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The loss of AP-2alpha and increased activity of cAMP-responsive element binding (CREB) protein are two hallmarks of malignant progression of cutaneous melanoma. However, the molecular mechanism responsible for the loss of AP-2alpha during melanoma progression remains unknown. METHODOLOGY/PRINCIPAL FINDINGS Herein, we demonstrate that both inhibition of PKA-dependent CREB phosphorylation, as well as silencing of CREB expression by shRNA, restored AP-2alpha protein expression in two metastatic melanoma cell lines. Moreover, rescue of CREB expression in CREB-silenced cell lines downregulates expression of AP-2alpha. Loss of AP-2alpha expression in metastatic melanoma occurs via a dual mechanism involving binding of CREB to the AP-2alpha promoter and CREB-induced overexpression of another oncogenic transcription factor, E2F-1. Upregulation of AP-2alpha expression following CREB silencing increases endogenous p21(Waf1) and decreases MCAM/MUC18, both known to be downstream target genes of AP-2alpha involved in melanoma progression. CONCLUSIONS/SIGNIFICANCE Since AP-2alpha regulates several genes associated with the metastatic potential of melanoma including c-KIT, VEGF, PAR-1, MCAM/MUC18, and p21(Waf1), our data identified CREB as a major regulator of the malignant melanoma phenotype.
Collapse
Affiliation(s)
- Vladislava O. Melnikova
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Andrey S. Dobroff
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Maya Zigler
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Gabriel J. Villares
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Russell R. Braeuer
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Hua Wang
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Li Huang
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Menashe Bar-Eli
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
35
|
Li H, Yuan G. Electrospray ionization mass spectrometry probing of formation and recognition of the G-quadruplex in the proximal promoter of the human vascular endothelial growth factor gene. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2010; 24:2030-2034. [PMID: 20552697 DOI: 10.1002/rcm.4613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The formation of the G-quadruplex of the vascular endothelial growth factor (VEGF) gene was probed by electrospray ionization mass spectrometry (ESI-MS). It found that cations (K(+) and NH(4)(+)), CH(3)OH and pH influence significantly the formation of the G-quadruplex structure. Additionally, a perylene derivative (P3) and polydatin (P4) have shown to be potential G-quadruplex binding agents with structurally specific recognition.
Collapse
Affiliation(s)
- Huihui Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | | |
Collapse
|
36
|
Shimoyamada H, Yazawa T, Sato H, Okudela K, Ishii J, Sakaeda M, Kashiwagi K, Suzuki T, Mitsui H, Woo T, Tajiri M, Ohmori T, Ogura T, Masuda M, Oshiro H, Kitamura H. Early growth response-1 induces and enhances vascular endothelial growth factor-A expression in lung cancer cells. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:70-83. [PMID: 20489156 DOI: 10.2353/ajpath.2010.091164] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Vascular endothelial growth factor-A (VEGF-A) is crucial for angiogenesis, vascular permeability, and metastasis during tumor development. We demonstrate here that early growth response-1 (EGR-1), which is induced by the extracellular signal-regulated kinase (ERK) pathway activation, activates VEGF-A in lung cancer cells. Increased EGR-1 expression was found in adenocarcinoma cells carrying mutant K-RAS or EGFR genes. Hypoxic culture, siRNA experiment, luciferase assays, chromatin immunoprecipitation, electrophoretic mobility shift assays, and quantitative RT-PCR using EGR-1-inducible lung cancer cells demonstrated that EGR-1 binds to the proximal region of the VEGF-A promoter, activates VEGF-A expression, and enhances hypoxia inducible factor 1alpha (HIF-1alpha)-mediated VEGF-A expression. The EGR-1 modulator, NAB-2, was rapidly induced by increased levels of EGR-1. Pathology samples of human lung adenocarcinomas revealed correlations between EGR-1/HIF-1alpha and VEGF-A expressions and relative elevation of EGR-1 and VEGF-A expression in mutant K-RAS- or EGFR-carrying adenocarcinomas. Both EGR-1 and VEGF-A expression increased as tumors dedifferentiated, whereas HIF-1alpha expression did not. Although weak correlation was found between EGR-1 and NAB-2 expressions on the whole, NAB-2 expression decreased as tumors dedifferentiated, and inhibition of DNA methyltransferase/histone deacetylase increased NAB-2 expression in lung cancer cells despite no epigenetic alteration in the NAB-2 promoter. These findings suggest that EGR-1 plays important roles on VEGF-A expression in lung cancer cells, and epigenetic silencing of transactivator(s) associated with NAB-2 expression might also contribute to upregulate VEGF-A expression.
Collapse
Affiliation(s)
- Hiroaki Shimoyamada
- Department of Pathology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Leptin upregulates VEGF in breast cancer via canonic and non-canonical signalling pathways and NFkappaB/HIF-1alpha activation. Cell Signal 2010; 22:1350-62. [PMID: 20466060 DOI: 10.1016/j.cellsig.2010.05.003] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 05/05/2010] [Indexed: 01/26/2023]
Abstract
High levels of VEGF and leptin are strongly linked to worse prognosis of breast cancer. Leptin signalling upregulates VEGF in human and mouse mammary tumor cells (MT), but the specific molecular mechanisms are largely unknown. Pharmacologic and genetic approaches were used to dissect the mechanism of leptin regulation of VEGF protein and mRNA in MT (4T1, EMT6 and MMT). A series of VEGF-promoter Luc-reporters (full-length and transcription factor-binding deletions) were transfected into MT to analyze leptin regulation of VEGF transcription. Deletion analysis of VEGF promoter and RNA knockdown shows that HIF-1alpha and NFkappaB are essentials for leptin regulation of VEGF. Leptin activation of HIF-1alpha was mainly linked to canonic (MAPK, PI-3K) and non-canonic (PKC, JNK and p38 MAP) signalling pathways. Leptin non-canonic signalling pathways (JNK, p38 MAP and to less extent PKC) were linked to NFkappaB activation. SP1 was involved in leptin regulation of VEGF in 4T1 cells. AP1 was not involved and AP2 repressed leptin-induced increase of VEGF. Overall, these data suggest that leptin signalling regulates VEGF mainly through HIF-1alpha and NFkappaB. These results delineate a comprehensive mechanism for leptin regulation of VEGF in MT. Disruption of leptin signalling could be used as a novel way to treat breast cancer.
Collapse
|
38
|
Martinelli E, De Palma R, Orditura M, De Vita F, Ciardiello F. Anti-epidermal growth factor receptor monoclonal antibodies in cancer therapy. Clin Exp Immunol 2009; 158:1-9. [PMID: 19737224 PMCID: PMC2759052 DOI: 10.1111/j.1365-2249.2009.03992.x] [Citation(s) in RCA: 231] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2009] [Indexed: 11/30/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a transmembrane tyrosine kinase receptor involved in the proliferation and survival of cancer cells. EGFR is the first molecular target against which monoclonal antibodies (mAb) have been developed for cancer therapy. Here we review the mechanisms underlying the effects of EGFR-specific mAb in cancer therapy. The efficacy of EGFR-specific mAb in cancer occurs thanks to inhibition of EGFR-generated signalling; furthermore, the effects of antibodies on the immune system seem to play an important role in determining the overall anti-tumour response. In this review, attention is focused on cetuximab and panitumumab, two mAb introduced recently into clinical practice for treatment of metastatic colorectal and head and neck cancer which target the external part of EGFR.
Collapse
Affiliation(s)
- E Martinelli
- Dipartimento Medico-Chirurgico di Internistica Clinica, Seconda Università degli Studi di Napoli, Naples, Italy
| | | | | | | | | |
Collapse
|
39
|
Lee HS, Myers A, Kim J. Vascular endothelial growth factor drives autocrine epithelial cell proliferation and survival in chronic rhinosinusitis with nasal polyposis. Am J Respir Crit Care Med 2009; 180:1056-67. [PMID: 19762561 DOI: 10.1164/rccm.200905-0740oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE The pathogenesis of nasal polyps in chronic rhinosinusitis is poorly understood. OBJECTIVES These studies seek to implicate a functional role for vascular endothelial growth factor (VEGF) in perpetuating primary nasal epithelial cell overgrowth, a key feature of hyperplastic polyps. METHODS Comparison of VEGF and receptor expression was assessed by ELISA of nasal lavage, immunohistochemistry of sinus tissue, flow cytometry of nasal epithelial cells, and ELISA of supernatants. VEGF-dependent cell growth and apoptosis were assessed with blocking antibodies to VEGF, their receptors, or small interfering RNA knockdown of neuropilin-1 by cell proliferation assays and flow cytometric binding of annexin V. MEASUREMENTS AND MAIN RESULTS VEGF protein was sevenfold higher in nasal lavage from patients with polyposis compared with control subjects (P < 0.001). We also report elevated expression of VEGF (P < 0.012), receptors VEGFR2 and phospho-VEGFR2 (both P < 0.04), and identification of VEGF coreceptor neuropilin-1 in these tissues. Nasal epithelial cells from patients with polyps demonstrated faster growth rates (P < 0.005). Exposure of cells to blocking antibodies against VEGF resulted in inhibition of cell growth (P < 0.05). VEGF receptor blockade required blockade of neuropilin-1 (P < 0.05) and resulted in increased apoptosis (P < 0.001) and inhibition of autocrine epithelial VEGF production (P < 0.05). CONCLUSIONS These data demonstrate that VEGF is a novel biomarker for chronic rhinosinusitis with hyperplastic sinonasal polyposis that functions in an autocrine feed-forward manner to promote nasal epithelial cell growth and to inhibit apoptosis. These findings implicate a previously unrecognized and novel role of VEGF functioning through neuropilin-1 on nonneoplastic primary human airway epithelial cells, to amplify cell growth, contributing to exuberant hyperplastic polyposis.
Collapse
Affiliation(s)
- Hyun Sil Lee
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Room 3B65A, Baltimore, MD 21224, USA
| | | | | |
Collapse
|
40
|
Abstract
The NADPH oxidase (Nox) family of enzymes generates reactive oxygen species (ROS). At low ROS concentration, intracellular signaling is initiated, whereas at high ROS concentration, oxidative stress is induced. The extensive studies over the years have shed light on the mediating roles of the Nox enzymes in a variety of normal physiological processes ranging from bactericidal activity to remodeling of the extracellular matrix. Consequently, imbalance of Nox activities could be the potential cause of acute or chronic diseases. With regard to functional relationships between Nox isoforms and pathogenesis, it is of particular interest to study whether they are involved in carcinogenesis, because overproduction of ROS has long been implicated as a risk factor in cancer development. We see one remarkable example of the causal relationship between Nox1 and cancer in Ras oncogene-induced cell transformation. Other studies also indicate that the Nox family of genes appears to be required for survival and growth of a subset of human cancer cells. Thus, the Nox family will be a focus of attention in cancer biology and etiology over the next couple years.
Collapse
Affiliation(s)
- Tohru Kamata
- Department of Molecular Biology and Biochemistry, Shinshu University Graduate School of Medicine, Matsumoto, Nagano, Japan.
| |
Collapse
|
41
|
Milas L, Mason KA, Ang KK. Epidermal growth factor receptor and its inhibition in radiotherapy:in vivofindings. Int J Radiat Biol 2009; 79:539-45. [PMID: 14530163 DOI: 10.1080/0955300031000114747] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Increasing evidence shows that dysregulated epidermal growth factor receptor (EGFR) signalling plays an important part in neoplasia. When over expressed or mutated, EGFR is frequently associated with more aggressive tumour growth, poor patient prognosis and resistance of tumours to cytotoxic agents, including radiation. The present studies with murine carcinomas showed that there is an inverse correlation between the level of EGFR and tumour radiocurability. Likewise, the present clinical study in patients with head and neck cancer shows that EGFR over expression correlates with poorer tumour response to radiotherapy. Adding EGFR to tumour cells in vitro protected cells against the cytotoxic action of radiation, whereas blocking EGFR with anti-EGFR antibodies enhanced cell radiosensitivity. A casual relationship between EGFR and increased cellular resistance to radiation was established by transferring the EGFR gene into low EGFR-expressing radiosensitive tumour cells, which then become radioresistant. Radiation activated EGFR and its downstream signalling pathways in radioresistant but not in radiosensitive tumours, and this effect was associated with increased resistance to radiation, and enhanced repopulation in irradiated tumours. Increasing evidence shows that blockage of EGFR or interference with any of the steps in its signal transduction cascade can counteract negative outcomes of EGFR signalling, which has recently been explored as a therapeutic strategy in cancer treatment. The present findings demonstrate that treatment of human tumour xenografts with C225, an anti-EGFR monoclonal antibody, dramatically enhanced tumour response to radiation. Overall, the findings show that over expression of EGFR may serve as a predictor of tumour treatment outcome by radiotherapy and as a therapeutic target to enhance the efficacy of radiotherapy.
Collapse
Affiliation(s)
- L Milas
- The University of Texas M. D. Anderson Cancer Center, Department of Experimental Radiation Oncology, Houston, TX 77030-4009, USA
| | | | | |
Collapse
|
42
|
Grépin R, Pagès G. Le Vascular Endothelial Growth Factor (VEGF) : un modèle de régulation d'expression génique et un marqueur d'agressivité tumorale. Une cible thérapeutique évidente ? ACTA ACUST UNITED AC 2009; 203:181-92. [DOI: 10.1051/jbio/2009022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
43
|
Transforming growth factor alpha induces angiogenesis and neurogenesis following stroke. Neuroscience 2009; 163:233-43. [PMID: 19481589 DOI: 10.1016/j.neuroscience.2009.05.050] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Revised: 05/21/2009] [Accepted: 05/22/2009] [Indexed: 01/01/2023]
Abstract
The cytokine transforming growth factor alpha (TGF alpha) has proangiogenic and proneurogenic effects and can potentially reduce infarct volumes. Therefore, we administered TGF alpha or vehicle directly into the area surrounding the infarct in female mice that received gender-mismatched bone marrow transplants from green fluorescent protein (GFP)-expressing males prior to undergoing permanent middle cerebral artery occlusion. Newborn cells were tracked with bromodeoxyuridine (BrdU) labeling and immunohistochemistry at 90 days after stroke onset. We also studied the ingress of bone marrow-derived cells into the ischemic brain to determine whether such cells contribute to angiogenesis or neurogenesis. Infarct volumes were measured at 90 days poststroke. The results show that TGF alpha led to significant increments in the number of newborn neurons and glia in the ischemic hemisphere. TGF alpha also led to significant increments in the number of bone marrow-derived cells entering into the ischemic hemisphere. Most of these cells did not label with BrdU and represented endothelial cells that incorporated into blood vessels in the infarct border zone. Our results also show that infarct size was significantly reduced in animals treated with TGF alpha compared with controls. These results suggest that TGF alpha can induce angiogenesis, neurogenesis and neuroprotection after stroke. At least part of the pro-angiogenic effect appears to be secondary to the incorporation of bone marrow-derived endothelial cells into blood vessels in the infarct border zone.
Collapse
|
44
|
Katakowski M, Jiang F, Zheng X, Gutierrez JA, Szalad A, Chopp M. Tumorigenicity of cortical astrocyte cell line induced by the protease ADAM17. Cancer Sci 2009; 100:1597-604. [PMID: 19515085 DOI: 10.1111/j.1349-7006.2009.01221.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The metalloprotease ADAM17 (a.k.a. TACE) plays a pivotal role in the cleavage and activation of membrane-anchored receptor ligands. More recently, it has been revealed that ADAM17 is a potent sheddase of the epidermal growth factor (EGF) family of ligands and regulates epidermal growth factor receptor (EGFR) activity in a variety of tumors. EGFR is a key component of autonomous growth signaling in several tumors, and correlates with the malignancy grade of astrocytoma. In this study, we tested the hypothesis that over-expression of ADAM17 in cortical astrocytes derived from normal brain would induce a progression towards a malignant phenotype. Over-expression of human ADAM17 (hADAM17) in the CTX-TNA2 cortical astrocyte cell line resulted in non-adherent growth, increased proliferation, invasiveness, production of angiogenic factors, and expression of genes associated with immature and/or neoplastic cells. hADAM17 up-regulated EGFR and AKT phosphorylation, and increased proliferation and cell invasion were significantly dependent upon EGFR activity. When implanted in the nude mouse brain, CTX-TNA2 cells induced low histological grade, benign intraventricular gliomas. In contrast, the same astrocytes with hADAM17 formed large malignant gliomas. Taken together, these findings suggest that unregulated ADAM17 activity induces functional changes in astrocytes that significantly advance the malignant phenotype.
Collapse
Affiliation(s)
- Mark Katakowski
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | | | | | | | | | | |
Collapse
|
45
|
Mongerard-Coulanges M, Migianu-Griffoni E, Lecouvey M, Jolles B. Impact of alendronate and VEGF-antisense combined treatment on highly VEGF-expressing A431 cells. Biochem Pharmacol 2009; 77:1580-5. [PMID: 19426694 DOI: 10.1016/j.bcp.2009.02.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 02/18/2009] [Accepted: 02/19/2009] [Indexed: 01/01/2023]
Abstract
Bisphosphonates, and more specially nitrogen-containing bisphosphonates, which are in current use for the treatment of bone diseases, demonstrate proapoptotic, antiproliferative, antiangiogenic and anti-invasive properties on tumor cells. The amino-bisphosphonate alendronate is considered as a potential anticancer drug. In the case of A431 cells, which express high levels of VEGF, it had a two-step effect. At 24h, the antitumor properties of alendronate were counterbalanced by a survival process, which consisted of an enhancement of VEGF expression (mRNA and protein secretion) and TGF alpha secretion. It was only at 48 h that alendronate displayed the expected antiproliferative and antiangiogenic properties. The first step, in which the PI3K pathway was engaged, could be prevented by the use of a VEGF-antisense oligonucleotide. The combination of such an antisense with small concentrations of alendronate (approximately 2 microM), which is of the order of clinically used concentrations, was shown to have an antiangiogenic effect as soon as 12h.
Collapse
|
46
|
Gaudreault M, Gingras ME, Lessard M, Leclerc S, Guérin SL. Electrophoretic mobility shift assays for the analysis of DNA-protein interactions. Methods Mol Biol 2009; 543:15-35. [PMID: 19378156 DOI: 10.1007/978-1-60327-015-1_2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Electromobility shift assay is a simple, efficient, and rapid method for the study of specific DNA-protein interactions. It relies on the reduction in the electrophoretic mobility conferred to a DNA fragment by an interacting protein. The technique is suitable to qualitative, quantitative, and kinetic analyses. It can also be used to analyze conformational changes.
Collapse
Affiliation(s)
- Manon Gaudreault
- Oncology and Molecular Endocrinology Research Center, CHUL, Centre Hospitalier Universitaire de Québec and Laval University, 2705 Laurier Blvd, Québec, QC, Canada, G1V 4G2
| | | | | | | | | |
Collapse
|
47
|
A requirement for thioredoxin in redox-sensitive modulation of T-cadherin expression in endothelial cells. Biochem J 2008; 416:271-80. [PMID: 18627351 DOI: 10.1042/bj20080765] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
T-cad (T-cadherin), a glycosylphosphatidylinositol-anchored cadherin superfamily member, is expressed widely in the brain and cardiovascular system, and absent, decreased, or even increased, in cancers. Mechanisms controlling T-cad expression are poorly understood. The present study investigated transcriptional regulation of T-cad in ECs (endothelial cells). Conditions of oxidative stress (serum-deprivation or presence of H(2)O(2)) elevate T-cad mRNA and protein levels in ECs. Reporter gene analysis, using serially deleted T-cad promoter stretches ranging from -99 to -2304 bp, located the minimal promoter region of T-cad within -285 bp from the translation start site. Reporter activity in ECs transfected with the -285 bp construct increased under conditions of oxidative stress, and this was normalized by antioxidant N-acetylcysteine. An electrophoretic-mobility-shift assay revealed a specific nucleoprotein complex unique to -156 to -203 bp, which increased when nuclear extracts from oxidatively stressed ECs were used, suggesting the presence of redox-sensitive binding element(s). MS analysis of the nucleoprotein complex unique to -156 to -203 bp after streptavidin-agarose pull-down detected the presence of the redox-active protein thioredoxin. The presence of thioredoxin-1 in a nuclear extract from oxidatively stressed ECs was demonstrated after immunoprecipitation and immunoblotting. Transfection of ECs with thioredoxin-1 small interfering RNA abrogated oxidative-stress-induced up-regulation of T-cad transcripts and protein. We conclude that thioredoxin-1 is an important determinant of redox-sensitive transcriptional up-regulation of T-cad in ECs.
Collapse
|
48
|
Clifford RL, Deacon K, Knox AJ. Novel regulation of vascular endothelial growth factor-A (VEGF-A) by transforming growth factor (beta)1: requirement for Smads, (beta)-CATENIN, AND GSK3(beta). J Biol Chem 2008; 283:35337-53. [PMID: 18952601 DOI: 10.1074/jbc.m803342200] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is a vital angiogenic effector, regulating key angiogenic processes. Vascular development relies on numerous signaling pathways, of which those induced by transforming growth factor-beta (TGFbeta) are critical. The Wnt/beta-catenin signaling pathway is emerging as necessary for vascular development. Although VEGF, TGFbeta, and Wnt signal transductions are well studied individually, it has not been demonstrated previously that all three can interact or be dependent on each other. We show that regulation of VEGF by TGFbeta(1), in human pulmonary artery smooth muscle cells (PASMCs), depends on a direct interaction between TGFbeta signaling proteins, Smads, and members of the Wnt/beta-catenin signaling family. VEGF promoter reporter constructs identified a region of the VEGF promoter containing two T cell factor (TCF)-binding sites as necessary for TGFbeta(1)-induced VEGF transcription. Mutation of TCF sites and expression of dominant negative TCF4 abolished TGFbeta(1)-induced VEGF promoter activity. Studies in Smad2 and Smad3 knock-out mouse embryonic fibroblasts demonstrated that one or both are required for VEGF regulation by TGFbeta(1), with transfection of dominant negative Smad2 or Smad3 into PASMCs confirming this. Chromatin immunoprecipitation assays showed in cell interactions of Smad2 and Smad3 with TCF4 and beta-catenin at the VEGF promoter, whereas co-immunoprecipitation showed a direct physical interaction between Smad2 and beta-catenin in the nucleus of PASMCs. Finally, we demonstrate that TGFbeta(1) regulates TCF by modifying beta-catenin phosphorylation via regulation of glycogen synthase kinase 3beta. These results provide new insight into the molecular regulation of VEGF by two interacting pathways necessary for vascular development, maintenance, and disease.
Collapse
Affiliation(s)
- Rachel L Clifford
- Centre for Respiratory Research, Clinical Sciences Building, University of Nottingham, Nottingham NG5 1PB, United Kingdom
| | | | | |
Collapse
|
49
|
Bianco R, Rosa R, Damiano V, Daniele G, Gelardi T, Garofalo S, Tarallo V, De Falco S, Melisi D, Benelli R, Albini A, Ryan A, Ciardiello F, Tortora G. Vascular endothelial growth factor receptor-1 contributes to resistance to anti-epidermal growth factor receptor drugs in human cancer cells. Clin Cancer Res 2008; 14:5069-80. [PMID: 18694994 DOI: 10.1158/1078-0432.ccr-07-4905] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The resistance to selective EGFR inhibitors involves the activation of alternative signaling pathways, and Akt activation and VEGF induction have been described in EGFR inhibitor-resistant tumors. Combined inhibition of EGFR and other signaling proteins has become a successful therapeutic approach, stimulating the search for further determinants of resistance as basis for novel therapeutic strategies. EXPERIMENTAL DESIGN We established human cancer cell lines with various degrees of EGFR expression and sensitivity to EGFR inhibitors and analyzed signal transducers under the control of EGFR-dependent and EGFR-independent pathways. RESULTS Multitargeted inhibitor vandetanib (ZD6474) inhibited the growth and the phosphorylation of Akt and its effector p70S6 kinase in both wild-type and EGFR inhibitor-resistant human colon, prostate, and breast cancer cells. We found that the resistant cell lines exhibit, as common feature, VEGFR-1/Flt-1 overexpression, increased secretion of VEGF and placental growth factor, and augmented migration capabilities and that vandetanib is able to antagonize them. Accordingly, a new kinase assay revealed that in addition to VEGF receptor (VEGFR)-2, RET, and EGFR, vandetanib efficiently inhibits also VEGFR-1. The contribution of VEGFR-1 to the resistant phenotype was further supported by the demonstration that VEGFR-1 silencing in resistant cells restored sensitivity to anti-EGFR drugs and impaired migration capabilities, whereas exogenous VEGFR-1 overexpression in wild-type cells conferred resistance to these agents. CONCLUSIONS This study shows that VEGFR-1 contributes to anti-EGFR drug resistance in different human cancer cells. Moreover, vandetanib inhibits VEGFR-1 activation, cell proliferation, and migration, suggesting its potential utility in patients resistant to EGFR inhibitors.
Collapse
Affiliation(s)
- Roberto Bianco
- Cattedra di Oncologia Medica, Dipartimento di Endocrinologia e Oncologia Molecolare e Clinica, Università di Napoli Federico II, Via S. Pansini 5, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Magné N, Chargari C, Castadot P, Ghalibafian M, Soria JC, Haie-Meder C, Bourhis J, Deutsch E. The efficacy and toxicity of EGFR in the settings of radiotherapy: Focus on published clinical trials. Eur J Cancer 2008; 44:2133-43. [PMID: 18692389 DOI: 10.1016/j.ejca.2008.06.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 06/13/2008] [Accepted: 06/20/2008] [Indexed: 10/21/2022]
Abstract
Basic research in solid malignant tumours has led to a wealth of knowledge about this disease process and about novel ways to more effectively target our therapies. Laboratory research continues to identify novel therapeutic targets and moreover, clinical research is identifying effective new treatment regimens. Many preclinical studies in this area have targeted the epidermal growth factor receptor (EGFR) signalling pathway to increase radiosensitivity. The in vitro rationale for targeting EGFR and concurrent ionising radiation is well established, but to date, rare clinical data could provide proof-of-principle. Here we report all the different published clinical trials focusing on efficacy and toxicity in order to clarify and to summarise the present state-of-the-art of this particularly promising combination in solid tumour management.
Collapse
Affiliation(s)
- Nicolas Magné
- Department of Radiotherapy, Institut Gustave Roussy, 39 rue Camille Desmoulins, 94 805 Villejuif, France.
| | | | | | | | | | | | | | | |
Collapse
|