1
|
Batan S, Kuppuswamy S, Wood M, Reddy M, Annex B, Ganta V. Inhibiting anti-angiogenic VEGF165b activates a miR-17-20a-Calcipressin-3 pathway that revascularizes ischemic muscle in peripheral artery disease. COMMUNICATIONS MEDICINE 2024; 4:3. [PMID: 38182796 PMCID: PMC10770062 DOI: 10.1038/s43856-023-00431-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/19/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND VEGF165a increases the expression of the microRNA-17-92 cluster, promoting developmental, retinal, and tumor angiogenesis. We have previously shown that VEGF165b, an alternatively spliced anti-angiogenic VEGF-A isoform, inhibits the VEGFR-STAT3 pathway in ischemic endothelial cells (ECs) to decrease their angiogenic capacity. In ischemic macrophages (Møs), VEGF165b inhibits VEGFR1 to induce S100A8/A9 expression, which drives M1-like polarization. Our current study aims to determine whether VEGF165b inhibition promotes perfusion recovery by regulating the microRNA(miR)-17-92 cluster in preclinical PAD. METHODS Femoral artery ligation and resection was used as a preclinical PAD model. Hypoxia serum starvation (HSS) was used as an in vitro PAD model. VEGF165b was inhibited/neutralized by an isoform-specific VEGF165b antibody. RESULTS Here, we show that VEGF165b-inhibition induces the expression of miR-17-20a (within miR-17-92 (miR-17-18a-19a-19b-20a-92) cluster) in HSS-ECs and HSS-Møs vs. respective normal and/or isotype-matched IgG controls to enhance perfusion recovery. Consistent with the bioinformatics analysis that revealed RCAN3 as a common target of miR-17 and miR-20a, Argonaute-2 pull-down assays showed decreased miR-17-20a expression and higher RCAN3 expression in the RNA-induced silencing complex of HSS-ECs and HSS-Møs vs. respective controls. Inhibiting miR-17-20a induced RCAN3 levels to decrease ischemic angiogenesis and promoted M1-like polarization to impair perfusion recovery. Finally, using STAT3 inhibitors, S100A8/A9 silencers, and VEGFR1-deficient ECs and Møs, we show that VEGF165b-inhibition activates the miR-17-20a-RCAN3 pathway independent of VEGFR1-STAT3 or VEGFR1-S100A8/A9 in ischemic-ECs and ischemic-Møs respectively. CONCLUSIONS Our data revealed a hereunto unrecognized therapeutic 'miR-17-20a-RCAN3' pathway in the ischemic vasculature that is VEGFR1-STAT3/S100A8/A9 independent and is activated only upon VEGF165b-inhibition in PAD.
Collapse
Affiliation(s)
- Sonia Batan
- Vascular Biology Center, Department of Medicine, Augusta University, Augusta, GA, 30912, USA
| | - Sivaraman Kuppuswamy
- Vascular Biology Center, Department of Medicine, Augusta University, Augusta, GA, 30912, USA
| | - Madison Wood
- Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Meghana Reddy
- Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Brian Annex
- Vascular Biology Center, Department of Medicine, Augusta University, Augusta, GA, 30912, USA
| | - Vijay Ganta
- Vascular Biology Center, Department of Medicine, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
2
|
Maduabuchi WO, Tansi FL, Heller R, Hilger I. Hyperthermia Influences the Secretion Signature of Tumor Cells and Affects Endothelial Cell Sprouting. Biomedicines 2023; 11:2256. [PMID: 37626752 PMCID: PMC10452125 DOI: 10.3390/biomedicines11082256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Tumors are a highly heterogeneous mass of tissue showing distinct therapy responses. In particular, the therapeutic outcome of tumor hyperthermia treatments has been inconsistent, presumably due to tumor versus endothelial cell cross-talks related to the treatment temperature and the tumor tissue environment. Here, we investigated the impact of the average or strong hyperthermic treatment (43 °C or 47 °C for 1 h) of the human pancreatic adenocarcinoma cell line (PANC-1 and BxPC-3) on endothelial cells (HUVECs) under post-treatment normoxic or hypoxic conditions. Immediately after the hyperthermia treatment, the distinct repression of secreted pro-angiogenic factors (e.g., VEGF, PDGF-AA, PDGF-BB, M-CSF), intracellular HIF-1α and the enhanced phosphorylation of ERK1/2 in tumor cells were detectable (particularly for strong hyperthermia, 2D cell monolayers). Notably, there was a significant increase in endothelial sprouting when 3D self-organized pancreatic cancer cells were treated with strong hyperthermia and the post-treatment conditions were hypoxic. Interestingly, for the used treatment temperatures, the intracellular HIF-1α accumulation in tumor cells seems to play a role in MAPK/ERK activation and mediator secretion (e.g., VEGF, PDGF-AA, Angiopoietin-2), as shown by inhibition experiments. Taken together, the hyperthermia of pancreatic adenocarcinoma cells in vitro impacts endothelial cells under defined environmental conditions (cell-to-cell contact, oxygen status, treatment temperature), whereby HIF-1α and VEGF secretion play a role in a complex context. Our observations could be exploited for the hyperthermic treatment of pancreatic cancer in the future.
Collapse
Affiliation(s)
- Wisdom O. Maduabuchi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital—Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (W.O.M.); (F.L.T.)
| | - Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital—Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (W.O.M.); (F.L.T.)
| | - Regine Heller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Hans-Knöll-Str. 2, D-07745 Jena, Germany;
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital—Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (W.O.M.); (F.L.T.)
| |
Collapse
|
3
|
Genetic Mechanism Study of Auditory Phoenix Spheres and Transcription Factors Prediction for Direct Reprogramming by Bioinformatics. Int J Mol Sci 2022; 23:ijms231810287. [PMID: 36142199 PMCID: PMC9499413 DOI: 10.3390/ijms231810287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Hearing loss is the most common irreversible sensory disorder. By delivering regenerative cells into the cochlea, cell-based therapy provides a novel strategy for hearing restoration. Recently, newly-identified phoenix cells have drawn attention due to their nearly unlimited self-renewal and neural differentiation capabilities. They are a promising cell source for cell therapy and a potential substitute for induced pluripotent stem cells (iPSCs) in many in vitro applications. However, the underlying genomic mechanism of their self-renewal capabilities is largely unknown. The aim of this study was to identify hub genes and potential molecular mechanisms between differentiated and undifferentiated phoenix cells and predict transcription factors (TFs) for direct reprogramming. Material and Methods: The datasets were downloaded from the ArrayExpress database. Samples of differentiated and undifferentiated phoenix cells with three biological replicates were utilised for bioinformatic analysis. Differentially expressed genes (DEGs) were screened and the Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were investigated. The gene set enrichment analysis (GSEA) was conducted to verify the enrichment of four self-defined gene set collections, followed by protein-protein interaction (PPI) network construction and subcluster analysis. The prediction of TFs for direct reprogramming was performed based on the TRANSFAC database. Results: Ten hub genes were identified to be the key candidates for self-renewal. Ten TFs were predicted as the direct reprogramming factors. This study provides a theoretical foundation for understanding phoenix cells and clues for direct reprogramming, which would stimulate further experiments and clinical applications in hearing research and treatment.
Collapse
|
4
|
Campello E, Bosh F, Simion C, Spiezia L, Simioni P. Mechanisms of thrombosis in pancreatic ductal adenocarcinoma. Best Pract Res Clin Haematol 2022; 35:101346. [DOI: 10.1016/j.beha.2022.101346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022]
|
5
|
López Albors O, Olsson F, Llinares A, Gutiérrez H, Latorre R, Candanosa E, Guillén-Martínez A, Izquierdo-Rico M. Expression of the vascular endothelial growth factor system (VEGF) in the porcine oviduct during the estrous cycle. Theriogenology 2017; 93:46-54. [DOI: 10.1016/j.theriogenology.2017.01.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 01/15/2017] [Accepted: 01/15/2017] [Indexed: 11/25/2022]
|
6
|
Iacopetta D, Carocci A, Sinicropi MS, Catalano A, Lentini G, Ceramella J, Curcio R, Caroleo MC. Old Drug Scaffold, New Activity: Thalidomide-Correlated Compounds Exert Different Effects on Breast Cancer Cell Growth and Progression. ChemMedChem 2017; 12:381-389. [PMID: 28099781 DOI: 10.1002/cmdc.201600629] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/16/2017] [Indexed: 12/17/2022]
Abstract
Thalidomide was first used for relief of morning sickness in pregnant women and then withdrawn from the market because of its dramatic effects on normal fetal development. Over the last decades, it has been used successfully for the treatment of several pathologies, including cancer. Many analogues with improved activity have been synthesized and tested. Herein we report some effects on the growth and progression of MCF-7 and MDA-MB-231 breast cancer cells by a small series of thalidomide-correlated compounds, which are very effective at inducing cancer cell death by triggering TNFα-mediated apoptosis. The most active compounds are able to drastically reduce the migration of breast cancer cells by regulation of the two major proteins involved in epithelial-mesenchymal transition (EMT): vimentin and E-cadherin. Moreover, these compounds diminish the intracellular biosynthesis of vascular endothelial growth factor (VEGF), which is primarily involved in the promotion of angiogenesis, sustaining tumor progression. The multiple features of these compounds that act on various key points of the tumorigenesis process make them good candidates for preclinical studies.
Collapse
Affiliation(s)
- Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Alessia Carocci
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70126, Bari, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70126, Bari, Italy
| | - Giovanni Lentini
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70126, Bari, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Rosita Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Maria Cristina Caroleo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| |
Collapse
|
7
|
Szabó A, Mayor R. Modelling collective cell migration of neural crest. Curr Opin Cell Biol 2016; 42:22-28. [PMID: 27085004 PMCID: PMC5017515 DOI: 10.1016/j.ceb.2016.03.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 03/24/2016] [Accepted: 03/29/2016] [Indexed: 02/04/2023]
Abstract
Collective cell migration has emerged in the recent decade as an important phenomenon in cell and developmental biology and can be defined as the coordinated and cooperative movement of groups of cells. Most studies concentrate on tightly connected epithelial tissues, even though collective migration does not require a constant physical contact. Movement of mesenchymal cells is more independent, making their emergent collective behaviour less intuitive and therefore lending importance to computational modelling. Here we focus on such modelling efforts that aim to understand the collective migration of neural crest cells, a mesenchymal embryonic population that migrates large distances as a group during early vertebrate development. By comparing different models of neural crest migration, we emphasize the similarity and complementary nature of these approaches and suggest a future direction for the field. The principles derived from neural crest modelling could aid understanding the collective migration of other mesenchymal cell types.
Collapse
Affiliation(s)
- András Szabó
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
8
|
Sağsöz H, Saruhan BG, Erdoğan S. Functional roles of angiogenic factors and receptors on non-endothelial cells in the oropharyngeal cavity of the chukar partridge (Alectoris chukar). ACTA ZOOL-STOCKHOLM 2015. [DOI: 10.1111/azo.12149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hakan Sağsöz
- Department of Histology and Embryology; Faculty of Veterinary Medicine; Dicle University; 21280 Diyarbakir Turkey
| | - Berna G. Saruhan
- Department of Histology and Embryology; Faculty of Veterinary Medicine; Dicle University; 21280 Diyarbakir Turkey
| | - Serkan Erdoğan
- Department of Anatomy; Faculty of Veterinary Medicine; Namık Kemal University; 59030 Tekirdağ Turkey
| |
Collapse
|
9
|
Xiang L, Varshney R, Rashdan NA, Shaw JH, Lloyd PG. Placenta growth factor and vascular endothelial growth factor a have differential, cell-type specific patterns of expression in vascular cells. Microcirculation 2015; 21:368-79. [PMID: 24410720 DOI: 10.1111/micc.12113] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 01/07/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE PLGF, a VEGF-A related protein, mediates collateral enlargement via monocytes but plays little role in capillary proliferation. In contrast, VEGF-A mediates both collateral enlargement and capillary proliferation. PLGF has been less thoroughly studied than VEGF-A, and questions remain regarding its regulation and function. Therefore, our goal was to characterize the expression of PLGF by vascular cells. We hypothesized that vascular SMC would express more PLGF than EC, since VEGF-A is primarily expressed by non-EC. METHODS We compared PLGF and VEGF-A across eight EC and SMC lines, then knocked down PLGF and evaluated cell function. We also assessed the effect of hypoxia on PLGF expression and promoter activity. RESULTS PLGF was most highly expressed in EC, whereas VEGF-A was most highly expressed in SMC. PLGF knockdown did not affect EC number, migration, or tube formation, but reduced monocyte migration toward EC. Monocyte migration was rescued by exogenous PLGF. Hypoxia increased PLGF protein without activating PLGF gene transcription. CONCLUSIONS PLGF and VEGF-A have distinct patterns of expression in vascular cells. EC derived PLGF may function primarily in communication between EC and circulating cells. Hypoxia increases EC PLGF expression posttranscriptionally.
Collapse
Affiliation(s)
- Lingjin Xiang
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma, USA
| | | | | | | | | |
Collapse
|
10
|
Stewart MD, Ramani VC, Sanderson RD. Shed syndecan-1 translocates to the nucleus of cells delivering growth factors and inhibiting histone acetylation: a novel mechanism of tumor-host cross-talk. J Biol Chem 2014; 290:941-9. [PMID: 25404732 DOI: 10.1074/jbc.m114.608455] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The heparan sulfate proteoglycan syndecan-1 is proteolytically shed from the surface of multiple myeloma cells and is abundant in the bone marrow microenvironment where it promotes tumor growth, angiogenesis, and metastasis. In this study, we demonstrate for the first time that shed syndecan-1 present in the medium conditioned by tumor cells is taken up by bone marrow-derived stromal cells and transported to the nucleus. Translocation of shed syndecan-1 (sSDC1) to the nucleus was blocked by addition of exogenous heparin or heparan sulfate, pretreatment of conditioned medium with heparinase III, or growth of cells in sodium chlorate, indicating that sulfated heparan sulfate chains are required for nuclear translocation. Interestingly, cargo bound to sSDC1 heparan sulfate chains (i.e. hepatocyte growth factor) was transported to the nucleus along with sSDC1, and removal of heparan sulfate-bound cargo from sSDC1 abolished its translocation to the nucleus. Once in the nucleus, sSDC1 binds to the histone acetyltransferase enzyme p300, and histone acetyltransferase activity and histone acetylation are diminished. These findings reveal a novel function for shed syndecan-1 in mediating tumor-host cross-talk by shuttling growth factors to the nucleus and by altering histone acetylation in host cells. In addition, this work has broad implications beyond myeloma because shed syndecan-1 is present in high levels in many tumor types as well as in other disease states.
Collapse
Affiliation(s)
| | | | - Ralph D Sanderson
- From the Department of Pathology, UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
11
|
Postnatal hyperoxia exposure differentially affects hepatocytes and liver haemopoietic cells in newborn rats. PLoS One 2014; 9:e105005. [PMID: 25115881 PMCID: PMC4130630 DOI: 10.1371/journal.pone.0105005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/17/2014] [Indexed: 11/29/2022] Open
Abstract
Premature newborns are frequently exposed to hyperoxic conditions and experimental data indicate modulation of liver metabolism by hyperoxia in the first postnatal period. Conversely, nothing is known about possible modulation of growth factors and signaling molecules involved in other hyperoxic responses and no data are available about the effects of hyperoxia in postnatal liver haematopoiesis. The aim of the study was to analyse the effects of hyperoxia in the liver tissue (hepatocytes and haemopoietic cells) and to investigate possible changes in the expression of Vascular Endothelial Growth Factor (VEGF), Matrix Metalloproteinase 9 (MMP-9), Hypoxia-Inducible Factor-1α (HIF-1α), endothelial Nitric Oxide Synthase (eNOS), and Nuclear Factor-kB (NF-kB). Experimental design of the study involved exposure of newborn rats to room air (controls), 60% O2 (moderate hyperoxia), or 95% O2 (severe hyperoxia) for the first two postnatal weeks. Immunohistochemical and Western blot analyses were performed. Severe hyperoxia increased hepatocyte apoptosis and MMP-9 expression and decreased VEGF expression. Reduced content in reticular fibers was found in moderate and severe hyperoxia. Some other changes were specifically produced in hepatocytes by moderate hyperoxia, i.e., upregulation of HIF-1α and downregulation of eNOS and NF-kB. Postnatal severe hyperoxia exposure increased liver haemopoiesis and upregulated the expression of VEGF (both moderate and severe hyperoxia) and eNOS (severe hyperoxia) in haemopoietic cells. In conclusion, our study showed different effects of hyperoxia on hepatocytes and haemopoietic cells and differential involvement of the above factors. The involvement of VEGF and eNOS in the liver haemopoietic response to hyperoxia may be hypothesized.
Collapse
|
12
|
Clara CA, Marie SKN, de Almeida JRW, Wakamatsu A, Oba-Shinjo SM, Uno M, Neville M, Rosemberg S. Angiogenesis and expression of PDGF-C, VEGF, CD105 and HIF-1α in human glioblastoma. Neuropathology 2014; 34:343-52. [PMID: 24612214 DOI: 10.1111/neup.12111] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 01/24/2014] [Accepted: 01/25/2014] [Indexed: 12/29/2022]
Abstract
Glioblastoma (GBM), the most frequent and aggressive brain tumor, is characterized by marked angiogenesis directly related to invasiveness and poor prognosis. Hypoxia is considered to be an important stimulus for angiogenesis by inducing hypoxia-inducible factor 1-alpha (HIF-1α) overexpression that activates platelet-derived growth factor (PDGF) and VEGF. The aim of this study is to analyze the expression of PDGF-C, VEGF in endothelial and tumor cells of GBM and their relation to HIF-1α expression. Two hundred and eight GBM cases were studied by tissue microarray immunohistochemical preparation. Expression of HIF-1α, VEGF and PDGF-C was observed in 184 (88.5%), 131 (63%) and 160 (76.9%) tumor cases, respectively. The numbers of vessels were quantified by CD34, PDGF-C, VEGF and CD105 staining, and were in median 20, 16, 5 and 6, respectively. The GBMs that showed positive or negative expression for HIF-1α showed a median vascular density of 30 and 14, respectively, for CD34 (P < 0.015). Positive expression for HIF-1α was correlated with VEGF and PDGF-C expression in tumors (P < 0.001). There was a significant correlation between VEGF and PDGF-C expression in the cytoplasm of GBM tumor cells (P < 0.0001). We showed that VEGF expression in tumor cells was correlated with its expression in blood vessels (P < 0.0001). Endothelial cells with PDGF-C and VEGF positive expression were also positive for CD105 and their nuclei for Ki-67, confirming the neoangiogenic and proliferative influence of VEGF and PDGF-C. VEGF nuclear staining in tumor cells (P = 0.002) as well as nuclear staining for HIF-1α and VEGF (P = 0.005) correlated with survival. In summary, our present findings of the concomitant upregulation of PDGF-C with VEGF in GBM tumor cells and vessels further reinforce the benefit of using combined anti-angiogenic approaches to potentially improve the therapeutic response for GBM.
Collapse
|
13
|
Guzmán-Hernández ML, Potter G, Egervári K, Kiss JZ, Balla T. Secretion of VEGF-165 has unique characteristics, including shedding from the plasma membrane. Mol Biol Cell 2014; 25:1061-72. [PMID: 24501421 PMCID: PMC3967971 DOI: 10.1091/mbc.e13-07-0418] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
VEGF secretion is studied using VEGF165-GFP chimera. Efficient secretion requires Sar1- and Arf1-dependent steps and glycosylation in the Golgi. VEGF is retained in the outer surface of the plasma membrane, and shedding with other membrane components is an important step in the secretion process. Vascular endothelial growth factor (VEGF) is a critical regulator of endothelial cell differentiation and vasculogenesis during both development and tumor vascularization. VEGF-165 is a major form that is secreted from the cells via a poorly characterized pathway. Here we use green fluorescent protein– and epitope-tagged VEGF-165 and find that its early trafficking between the endoplasmic reticulum and the Golgi requires the small GTP-binding proteins Sar1 and Arf1 and that its glycosylation in the Golgi compartment is necessary for efficient post-Golgi transport and secretion from the cells. The relative temperature insensitivity of VEGF secretion and its Sar1 and Arf1 inhibitory profiles distinguish it from other cargoes using the “constitutive” secretory pathway. Prominent features of VEGF secretion are the retention of the protein on the outer surface of the plasma membrane and the stimulation of its secretion by Ca2+ and protein kinase C. Of importance, shedding of VEGF-165 from the cell surface together with other membrane components appears to be a unique feature by which some VEGF is delivered to the surroundings to exert its known biological actions. Understanding VEGF trafficking can reveal additional means by which tumor vascularization can be inhibited by pharmacological interventions.
Collapse
Affiliation(s)
- Maria Luisa Guzmán-Hernández
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892 Department of Neurosciences, University of Geneva, 1211 Geneva 4, Switzerland
| | | | | | | | | |
Collapse
|
14
|
Berendsen AD, Olsen BR. How vascular endothelial growth factor-A (VEGF) regulates differentiation of mesenchymal stem cells. J Histochem Cytochem 2013; 62:103-8. [PMID: 24309509 PMCID: PMC3902099 DOI: 10.1369/0022155413516347] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF), a key factor in angiogenesis, plays an essential role in skeletal development and postnatal homeostasis. VEGF serves as a survival factor for chondrocytes and couples the resorption of cartilage with bone formation during endochondral ossification. Recently, it has also been found to regulate the balance between osteoblast and adipocyte differentiation in bone marrow mesenchymal stem cells. Surprisingly, this regulatory function of VEGF is not based on paracrine signaling involving cell surface receptor activation. Instead, the mechanism appears to utilize intracellular VEGF, which is functionally linked to the nuclear envelope protein lamin A. Lamin A and VEGF control osteoblast and adipocyte differentiation by regulating the levels of the osteoblast and adipocyte transcription factors Runx2 and PPARγ, respectively. These data raise the intriguing possibility that loss of bone mass during aging may be manipulated by controlling the levels and activity of intracellular VEGF in bone marrow mesenchymal stem cells.
Collapse
Affiliation(s)
- Agnes D Berendsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | | |
Collapse
|
15
|
Raikwar NS, Liu KZ, Thomas CP. Protein kinase C regulates FLT1 abundance and stimulates its cleavage in vascular endothelial cells with the release of a soluble PlGF/VEGF antagonist. Exp Cell Res 2013; 319:2578-87. [PMID: 23911939 DOI: 10.1016/j.yexcr.2013.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 06/25/2013] [Accepted: 07/09/2013] [Indexed: 01/18/2023]
Abstract
FLT1 and its soluble form (sFLT1) arise as alternate transcripts from the same gene and sFLT1 can antagonize the effect of vascular endothelial growth factor (VEGF) on its cognate receptors. We investigated the effect of VEGF and protein kinase C (PKC) activation on sFLT1 abundance. We demonstrated that VEGF stimulates sFLT1 and FLT1 mRNA and protein levels in vascular endothelial cells via VEGFR2 and PKC. Using an FLT1 expression vector with N and C-terminal epitope tags, we show that PKC activation increases the cleavage of FLT1 into an N-terminal extracellular fragment and a C-terminal intracellular fragment with the cleavage occurring adjacent to the transmembrane domain. The trafficking and glycosylation inhibitors brefeldin, monensin and tunicamycin substantially reduced cleavage and release of the N-terminal ectodomain of FLT1 and inhibited secretion of the isoforms of sFLT1. The shed FLT1 ectodomain can bind VEGF and PlGF and inhibit VEGF-induced vascular tube formation thus confirming that it is functionally equivalent to the alternately spliced and secreted sFLT1 isoforms.
Collapse
Affiliation(s)
- Nandita S Raikwar
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA, USA
| | | | | |
Collapse
|
16
|
The effect of bevacizumab on human malignant melanoma cells with functional VEGF/VEGFR2 autocrine and intracrine signaling loops. Neoplasia 2013; 14:612-23. [PMID: 22904678 DOI: 10.1593/neo.11948] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 06/21/2012] [Accepted: 06/25/2012] [Indexed: 11/18/2022] Open
Abstract
Receptors for the angiogenic factor VEGF are expressed by tumor cancer cells including melanoma, although their functionality remains unclear. Paired human melanoma cell lines WM115 and WM239 were used to investigate differences in expression and functionality of VEGF and VEGFR2 in vitro and in vivo with the anti-VEGF antibody bevacizumab. Both WM115 and WM239 cells expressed VEGF and VEGFR2, the levels of which were modulated by hypoxia. Detection of native and phosphorylated VEGFR2 in subcellular fractions under serum-free conditions showed the presence of a functional autocrine as well as intracrine VEGF/VEGFR2 signaling loops. Interestingly, treatment of WM115 and WM239 cells with increasing doses of bevacizumab (0-300 µg/ml) in vitro did not show any significant inhibition of VEGFR2 phosphorylation. Small-molecule tyrosine kinase inhibitor, sunitinib, caused an inhibition of VEGFR2 phosphorylation in WM239 but not in WM115 cells. An increase in cell proliferation was observed in WM115 cells treated with bevacizumab, whereas sunitinib inhibited proliferation. When xenografted to immune-deficient mice, we found bevacizumab to be an effective antiangiogenic but not antitumorigenic agent for both cell lines. Because bevacizumab is unable to neutralize murine VEGF, this supports a paracrine angiogenic response. We propose that the failure of bevacizumab to generate an antitumorigenic effect may be related to its generation of enhanced autocrine/intracrine signaling in the cancer cells themselves. Collectively, these results suggest that, for cancers with intracrine VEGF/ VEGFR2 signaling loops, small-molecule inhibitors of VEGFR2 may be more effective than neutralizing antibodies at disease control.
Collapse
|
17
|
Liu Y, Berendsen AD, Jia S, Lotinun S, Baron R, Ferrara N, Olsen BR. Intracellular VEGF regulates the balance between osteoblast and adipocyte differentiation. J Clin Invest 2012; 122:3101-13. [PMID: 22886301 DOI: 10.1172/jci61209] [Citation(s) in RCA: 277] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 07/05/2012] [Indexed: 02/06/2023] Open
Abstract
Osteoporotic bones have reduced spongy bone mass, altered bone architecture, and increased marrow fat. Bone marrow stem cells from osteoporotic patients are more likely to differentiate into adipocytes than control cells, suggesting that adipocyte differentiation may play a role in osteoporosis. VEGF is highly expressed in osteoblastic precursor cells and is known to stimulate bone formation. Here we tested the hypothesis that VEGF is also an important regulator of cell fate, determining whether differentiation gives rise to osteoblasts or adipocytes. Mice with conditional VEGF deficiency in osteoblastic precursor cells exhibited an osteoporosis-like phenotype characterized by reduced bone mass and increased bone marrow fat. In addition, reduced VEGF expression in mesenchymal stem cells resulted in reduced osteoblast and increased adipocyte differentiation. Osteoblast differentiation was reduced when VEGF receptor 1 or 2 was knocked down but was unaffected by treatment with recombinant VEGF or neutralizing antibodies against VEGF. Our results suggested that VEGF controls differentiation in mesenchymal stem cells by regulating the transcription factors RUNX2 and PPARγ2 as well as through a reciprocal interaction with nuclear envelope proteins lamin A/C. Importantly, our data support a model whereby VEGF regulates differentiation through an intracrine mechanism that is distinct from the role of secreted VEGF and its receptors.
Collapse
Affiliation(s)
- Yanqiu Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts, USA.
| | | | | | | | | | | | | |
Collapse
|
18
|
Vieira JM, Ruhrberg C, Schwarz Q. VEGF receptor signaling in vertebrate development. Organogenesis 2012; 6:97-106. [PMID: 20885856 DOI: 10.4161/org.6.2.11686] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 04/10/2008] [Indexed: 01/27/2023] Open
Abstract
The secreted glycoprotein vascular endothelial growth factor A (VEGF or VEGFA) affects many different cell types and modifies a wide spectrum of cellular behaviors in tissue culture models, including proliferation, migration, differentiation and survival. The versatility of VEGF signaling is reflected in the complex composition of its cell surface receptors and their ability to activate a variety of different downstream signaling molecules. A major challenge for VEGF research is to determine which of the specific signaling pathways identified in vitro control development and homeostasis of tissues containing VEGF-responsive cell types in vivo.
Collapse
|
19
|
Berger S, Dyugovskaya L, Polyakov A, Lavie L. Short-term fibronectin treatment induces endothelial-like and angiogenic properties in monocyte-derived immature dendritic cells: Involvement of intracellular VEGF and MAPK regulation. Eur J Cell Biol 2012; 91:640-53. [DOI: 10.1016/j.ejcb.2012.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 02/21/2012] [Accepted: 02/21/2012] [Indexed: 12/16/2022] Open
|
20
|
Diaz-Padilla I, Siu LL, San Pedro-Salcedo M, Razak ARA, Colevas AD, Shepherd FA, Leighl NB, Neal JW, Thibault A, Liu L, Lisano J, Gao B, Lawson EB, Wakelee HA. A phase I dose-escalation study of aflibercept administered in combination with pemetrexed and cisplatin in patients with advanced solid tumours. Br J Cancer 2012; 107:604-11. [PMID: 22805331 PMCID: PMC3419963 DOI: 10.1038/bjc.2012.319] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: To evaluate the safety, pharmacokinetics (PKs), and pharmacodynamics of aflibercept, and to identify the recommended phase II dose (RP2D) of aflibercept in combination with pemetrexed and cisplatin. Methods: Aflibercept was administered at escalating doses of 2, 4, or 6 mg kg−1 in combination with fixed doses of pemetrexed (500 mg m−2) plus cisplatin (75 mg m−2) every 3 weeks. Blood samples were collected for PK analyses. Serum antiaflibercept antibodies were quantified to assess their impact on systemic aflibercept concentrations. Results: Eighteen patients were enrolled. One patient dosed at 4 mg kg−1 experienced grade 3 hypophosphatemia (dose-limiting toxicity; DLT), which prompted a cohort expansion. No further DLTs were observed in the 4 mg kg−1 cohort or the 6 mg kg−1 dose cohort. Most common adverse events (AEs) of all grades included (%): fatigue (89), anaemia (89), nausea (83), hyponatremia (78), and neutropenia (72). Grade ⩾3 AEs consistent with anti-vascular endothelial growth factor therapy included (%): hypertension (22), pulmonary embolism (11), and deep vein thrombosis (6). Five patients (28%) experienced mild neurocognitive disturbance. No episodes of reversible posterior leukoencephalopathy syndrome (RPLS) were noted. Conclusion: The results of this phase I study allowed further evaluation of the combination of aflibercept with pemetrexed and cisplatin in a phase II study. The RP2D of aflibercept was 6 mg kg−1, to be administered intravenously every 3 weeks in combination with pemetrexed and cisplatin.
Collapse
Affiliation(s)
- I Diaz-Padilla
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Hospital, University Health Network, University of Toronto, 610 University Avenue, 5-700, Toronto, Ontario M5G 2M9, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
McLennan R, Dyson L, Prather KW, Morrison JA, Baker RE, Maini PK, Kulesa PM. Multiscale mechanisms of cell migration during development: theory and experiment. Development 2012; 139:2935-44. [PMID: 22764050 DOI: 10.1242/dev.081471] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Long-distance cell migration is an important feature of embryonic development, adult morphogenesis and cancer, yet the mechanisms that drive subpopulations of cells to distinct targets are poorly understood. Here, we use the embryonic neural crest (NC) in tandem with theoretical studies to evaluate model mechanisms of long-distance cell migration. We find that a simple chemotaxis model is insufficient to explain our experimental data. Instead, model simulations predict that NC cell migration requires leading cells to respond to long-range guidance signals and trailing cells to short-range cues in order to maintain a directed, multicellular stream. Experiments confirm differences in leading versus trailing NC cell subpopulations, manifested in unique cell orientation and gene expression patterns that respond to non-linear tissue growth of the migratory domain. Ablation experiments that delete the trailing NC cell subpopulation reveal that leading NC cells distribute all along the migratory pathway and develop a leading/trailing cellular orientation and gene expression profile that is predicted by model simulations. Transplantation experiments and model predictions that move trailing NC cells to the migratory front, or vice versa, reveal that cells adopt a gene expression profile and cell behaviors corresponding to the new position within the migratory stream. These results offer a mechanistic model in which leading cells create and respond to a cell-induced chemotactic gradient and transmit guidance information to trailing cells that use short-range signals to move in a directional manner.
Collapse
Affiliation(s)
- Rebecca McLennan
- Stowers Institute for Medical Research, 1000 East 50th St, Kansas City, MO 64110, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Tadevosyan A, Vaniotis G, Allen BG, Hébert TE, Nattel S. G protein-coupled receptor signalling in the cardiac nuclear membrane: evidence and possible roles in physiological and pathophysiological function. J Physiol 2011; 590:1313-30. [PMID: 22183719 DOI: 10.1113/jphysiol.2011.222794] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptors (GPCRs) play key physiological roles in numerous tissues, including the heart, and their dysfunction influences a wide range of cardiovascular diseases. Recently, the notion of nuclear localization and action of GPCRs has become more widely accepted. Nuclear-localized receptors may regulate distinct signalling pathways, suggesting that the biological responses mediated by GPCRs are not solely initiated at the cell surface but may result from the integration of extracellular and intracellular signalling pathways. Many of the observed nuclear effects are not prevented by classical inhibitors that exclusively target cell surface receptors, presumably because of their structures, lipophilic properties, or affinity for nuclear receptors. In this topical review, we discuss specifically how angiotensin-II, endothelin, β-adrenergic and opioid receptors located on the nuclear envelope activate signalling pathways, which convert intracrine stimuli into acute responses such as generation of second messengers and direct genomic effects, and thereby participate in the development of cardiovascular disorders.
Collapse
Affiliation(s)
- Artavazd Tadevosyan
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | | | | | | | | |
Collapse
|
23
|
Is coagulation factor VIII a useful marker for colorectal carcinoma? Int J Biol Markers 2011; 27:20-6. [PMID: 22139641 DOI: 10.5301/jbm.2011.8832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2011] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND OBJECTIVE Increased thromboembolic events are well known in patients suffering from malignant diseases. In the following pilot study, we investigated the usefulness of coagulation factor VIII (FVIII) as a possible prognostic marker in patients with colorectal carcinoma (CRC). METHODS Plasma FVIII levels were measured in 79 patients with CRC, correlated with tumor characteristics, and compared with normal ranges of blood group (BG) 0 and BG A/AB/B and with 19 control patients. RESULTS In CRC patients mean FVIII levels were elevated compared with controls (BG 0: p=0.283, BG A/AB/B: p=0.001) and normal ranges. Interestingly, mean FVIII levels varied significantly in different blood groups (p=0.002). UICC stage I CRC patients presented with mean FVIII plasma levels within normal ranges, whereas UICC stage II-IV CRC patients presented with elevated FVIII plasma levels. In BG A/AB/B a significantly elevated FVIII level was found in G2 compared with G1 tumors (p< 0.001). Patients with elevated carcinoembryonic antigen also showed significantly elevated FVIII levels (p=0.050). FVIII levels at time of surgery did not correlate with survival within the first 2 years following surgery. CONCLUSION In this pilot study, we demonstrated that FVIII plasma levels are elevated in patients with CRC and affected by T-stage and differentiation of the tumor. Whether FVIII is a clinical useful marker needs to be tested in a larger cohort.
Collapse
|
24
|
Domingues I, Rino J, Demmers JAA, de Lanerolle P, Santos SCR. VEGFR2 translocates to the nucleus to regulate its own transcription. PLoS One 2011; 6:e25668. [PMID: 21980525 PMCID: PMC3182252 DOI: 10.1371/journal.pone.0025668] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 09/09/2011] [Indexed: 01/01/2023] Open
Abstract
Vascular Endothelial Growth Factor Receptor-2 (VEGFR2) is the major mediator of the angiogenic effects of VEGF. In addition to its well known role as a membrane receptor that activates multiple signaling pathways, VEGFR2 also has a nuclear localization. However, what VEGFR2 does in the nucleus is still unknown. In the present report we show that, in endothelial cells, nuclear VEGFR2 interacts with several nuclear proteins, including the Sp1, a transcription factor that has been implicated in the regulation of genes needed for angiogenesis. By in vivo chromatin immunoprecipitation (ChIP) assays, we found that VEGFR2 binds to the Sp1-responsive region of the VEGFR2 proximal promoter. These results were confirmed by EMSA assays, using the same region of the VEGFR2 promoter. Importantly, we show that the VEGFR2 DNA binding is directly linked to the transcriptional activation of the VEGFR2 promoter. By reporter assays, we found that the region between -300/-116 relative to the transcription start site is essential to confer VEGFR2-dependent transcriptional activity. It was previously described that nuclear translocation of the VEGFR2 is dependent on its activation by VEGF. In agreement, we observed that the binding of VEGFR2 to DNA requires VEGF activation, being blocked by Bevacizumab and Sunitinib, two anti-angiogenic agents that inhibit VEGFR2 activation. Our findings demonstrate a new mechanism by which VEGFR2 activates its own promoter that could be involved in amplifying the angiogenic response.
Collapse
Affiliation(s)
- Inês Domingues
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - José Rino
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Jeroen A. A. Demmers
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Primal de Lanerolle
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | | |
Collapse
|
25
|
Souza CA, Ocarino NM, Silva JF, Boeloni JN, Nascimento EF, Silva IJ, Castro RD, Moreira LP, Almeida FRCL, Chiarini-Garcia H, Serakides R. Administration of thyroxine affects the morphometric parameters and VEGF expression in the uterus and placenta and the uterine vascularization but does not affect reproductive parameters in gilts during early gestation. Reprod Domest Anim 2011; 46:e7-16. [PMID: 20403130 DOI: 10.1111/j.1439-0531.2010.01615.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The aim of this study was to evaluate the effects of thyroxine administration on morphometric parameters, expression of vascular endothelial growth factor (VEGF) and vascularization in the uterus and placenta and reproductive parameters in gilts at 70 days of gestation. At 150 days of age, i.e., before first heat, 20 gilts were randomly divided into two experimental groups: treated (n=10) and control (n=10). The treated group received a daily dose of 400 μg of L-thyroxine (T(4)) in their diet until slaughter and the control group received only typical meals. Before artificial insemination, blood was collected to determine plasma total T(4). The gilts were inseminated in the second oestrus and slaughtered at 70 days of gestation. The foetal thyroid follicular epithelium height, number, size and weight of foetuses; foetal myogenesis, corpora lutea number, embryonic mortality rate, uterine weight, placental weight and placental fluid volume were measured. Histomorphometric and immunohistochemical analysis of uterus and placenta were determined. The averages of all variables were compared by the Student's t-test. The gilts treated with thyroxine showed significant increase of plasma total T(4). At 70 days of gestation, the heights of the trophoblastic epithelium, endometrial epithelium and endometrial gland epithelium were significantly higher in the group treated with T(4). The expression of cytoplasmatic and nuclear VEGF in trophoblastic cells and the number of blood vessels per field in endometrial stroma were significantly higher in the gilts treated with T(4). No other significant differences between groups were obtained with respect to other parameters (p>0.05). We conclude that oral administration of T(4) up to 70 days of pregnancy in gilts affects the morphometric parameters, the expression of placental VEGF and the uterine vascularization but does not affect reproductive parameters in gilts during early gestation.
Collapse
Affiliation(s)
- C A Souza
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária da Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Pawson EJ, Duran-Jimenez B, Surosky R, Brooke HE, Spratt SK, Tomlinson DR, Gardiner NJ. Engineered zinc finger protein-mediated VEGF-a activation restores deficient VEGF-a in sensory neurons in experimental diabetes. Diabetes 2010; 59:509-18. [PMID: 19934008 PMCID: PMC2809974 DOI: 10.2337/db08-1526] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The objectives of the study were to evaluate retrograde axonal transport of vascular endothelial growth factor A (VEGF-A) protein to sensory neurons after intramuscular administration of an engineered zinc finger protein activator of endogenous VEGF-A (VZ+434) in an experimental model of diabetes, and to characterize the VEGF-A target neurons. RESEARCH DESIGN AND METHODS We compared the expression of VEGF-A in lumbar (L)4/5 dorsal root ganglia (DRG) of control rats and VZ+434-treated and untreated streptozotocin (STZ)-induced diabetic rats. In addition, axonal transport of VEGF-A, activation of signal transduction pathways in the DRG, and mechanical sensitivity were assessed. RESULTS VEGF-A immunoreactivity (IR) was detected in small- to medium-diameter neurons in DRG of control rats. Fewer VEGF-A-IR neurons were observed in DRG from STZ-induced diabetic rats; this decrease was confirmed and quantified by Western blotting. VZ+434 administration resulted in a significant increase in VEGF-A protein expression in ipsilateral DRG, 24 h after injection. VEGF-A was axonally transported to the DRG via the sciatic nerve. VZ+434 administration resulted in significant activation of AKT in the ipsilateral DRG by 48 h that was sustained for 1 week after injection. VZ+434 protected against mechanical allodynia 8 weeks after STZ injection. CONCLUSIONS Intramuscular administration of VZ+434 increases VEGF-A protein levels in L4/5 DRG, correcting the deficit observed after induction of diabetes, and protects against mechanical allodynia. Elevated VEGF-A levels result from retrograde axonal transport and are associated with altered signal transduction, via the phosphatidylinositol 3'-kinase pathway. These data support a neuroprotective role for VEGF-A in the therapeutic actions of VZ+434 and suggest a mechanism by which VEGF-A exerts this activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Natalie J. Gardiner
- Faculty of Life Sciences, University of Manchester, Manchester, U.K
- Corresponding author: Natalie Jane Gardiner,
| |
Collapse
|
27
|
Vormittag R, Simanek R, Ay C, Dunkler D, Quehenberger P, Marosi C, Zielinski C, Pabinger I. High factor VIII levels independently predict venous thromboembolism in cancer patients: the cancer and thrombosis study. Arterioscler Thromb Vasc Biol 2009; 29:2176-81. [PMID: 19778945 DOI: 10.1161/atvbaha.109.190827] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Patients with cancer are at an increased risk for venous thromboembolism (VTE). Clotting factor VIII activity (FVIII) has been established as risk factor of primary and recurrent VTE. We investigated FVIII as predictive parameter of VTE in cancer patients. METHODS AND RESULTS The prospective observational Cancer and Thrombosis Study (CATS) includes patients with newly diagnosed cancer or disease progression, study end point is symptomatic VTE. FVIII was measured on a Sysmex CA 7000 analyzer. Data on 840 patients (median age: 62 years, 25th to 75th percentile 53 to 68, 378 women) were available for analyses, of these 111 patients had hematologic malignancies and 729 solid cancer. During a median observation time of 495 days 62 events occurred. Cumulative probability of VTE after 6 months was 14% in patients with elevated FVIII-levels and 4% in those with normal levels (P=0.001). The association was strongest in younger patients: whereas in 40-year-old patients a 2-fold VTE risk per factor VIII increase of 20% was observed (HR=2.0 [95% CI: 1.5 to 2.7], P<0.0001), this association was still present but attenuated in older patients. CONCLUSIONS FVIII is independently associated with an increased risk of VTE in cancer patients. The association between FVIII and VTE risk declines with increasing age.
Collapse
Affiliation(s)
- R Vormittag
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
This paper reviews the current concepts of soft-tissue injury in orthopaedic trauma. Six topics are described in this Injury Supplement, including influencing factors and mechanisms, co-morbidities, biological responses, diagnosis and treatment of closed soft-tissue injury, compartment syndrome and gunshot wounds. Since one of the current AO principles emphasises respect for soft tissue when performing open reduction and internal fixation, this article further discusses the pathophysiology of soft-tissue injury and the specific concerns in treating compartment syndrome and gunshot injury. Understanding the basic and updated principles of soft-tissue management will be beneficial for the clinical practice of orthopaedic trauma surgeons.
Collapse
Affiliation(s)
- Yuan-Kun Tu
- Orthopaedic Department, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|
29
|
Shen YH, Shoichet MS, Radisic M. Vascular endothelial growth factor immobilized in collagen scaffold promotes penetration and proliferation of endothelial cells. Acta Biomater 2008; 4:477-89. [PMID: 18328795 DOI: 10.1016/j.actbio.2007.12.011] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 11/20/2007] [Accepted: 12/18/2007] [Indexed: 11/19/2022]
Abstract
A key challenge in engineering functional tissues in vitro is the limited transport capacity of oxygen and nutrients into the tissue. Inducing vascularization within engineered tissues is a key strategy to improving their survival in vitro and in vivo. The presence of vascular endothelial growth factor (VEGF) in a three-dimensional porous collagen scaffold may provide a useful strategy to promote vascularization of the engineered tissue in a controlled manner. To this end, we investigated whether immobilized VEGF could promote the invasion and assembly of endothelial cells (ECs) into the collagen scaffolds. We conjugated VEGF onto collagen scaffolds using N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride chemistry, and measured the concentrations of immobilized VEGF in collagen scaffolds by direct VEGF enzyme-linked immunosorbent assay. We demonstrated that immobilized VEGF (relative to soluble VEGF) promoted the penetration and proliferation of ECs in the collagen scaffold, based on results of cell density analysis in histological sections, immunohistochemistry, XTT proliferation assay, glucose consumption and lactate production. Furthermore, we observed increased viability of ECs cultured in scaffolds with immobilized VEGF relative to soluble VEGF. This research demonstrates that immobilization of VEGF is a useful strategy to promote the invasion and proliferation of ECs into a scaffold, which may in turn lead to a vascularized scaffold.
Collapse
Affiliation(s)
- Yi Hao Shen
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Rm. 407, Toronto, Ontario, Canada M5S 3G9
| | | | | |
Collapse
|
30
|
Oh Y, Herbst RS, Burris H, Cleverly A, Musib L, Lahn M, Bepler G. Enzastaurin, an oral serine/threonine kinase inhibitor, as second- or third-line therapy of non-small-cell lung cancer. J Clin Oncol 2008; 26:1135-41. [PMID: 18309949 DOI: 10.1200/jco.2007.14.3685] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
PURPOSE Enzastaurin, an oral serine/threonine kinase inhibitor, suppresses protein kinase C (PKC) and protein kinase B/AK transforming (AKT) signaling, induces tumor cell apoptosis, and inhibits proliferation and angiogenesis. Increased PKC and AKT activity is associated with poor prognosis in non-small-cell lung cancer (NSCLC). This phase II trial of enzastaurin was conducted to determine the 6-month progression-free survival (PFS) rate in advanced, metastatic NSCLC. PATIENTS AND METHODS Patients with metastatic (stage IV and wet IIIB) NSCLC, Eastern Cooperative Oncology Group performance status <or= 2, and <or= two prior systemic regimens (including one or more platinum-based chemotherapy regimens) received 500 mg of enzastaurin administered once daily. RESULTS Fifty-five patients were enrolled (55% male patients, 45% female patients; median age, 63 years; range, 44 to 82 years; 78% of patients having stage IV disease). Adenocarcinoma was the most common diagnosis (65%). Prior therapies included radiotherapy (73%) and epidermal growth factor inhibitors (29%). Median PFS was 1.8 months (95% CI, 1.7 to 1.9). Six-month PFS rate was 13% (95% CI, 3.9% to 21.5%). Median overall survival (OS) was 8.4 months (95% CI, 6.0 to 13.6 months). The 12-month OS rate was 44% (95% CI, 30.5% to 57.3%). Nineteen patients (35%) had stable disease. No objective responses were observed. Seven patients (13%) had PFS >or= 6 months, three of whom continued for more than 10 months. The most common toxicity was fatigue (grade <or= 3; n = 17). Grade 3 or worse toxicities were fatigue (n = 2), thromboembolism (n = 1), ataxia (n = 1), and anemia (n = 1). Two patients discontinued treatment because of drug-related fatigue and dizziness. Five patients died while enrolled in the study (non drug-related). CONCLUSION Although the primary end point of a 20% PFS rate was not achieved, 13% of the patients had PFS for >or= 6 months. Given the tolerability and survival data, evaluation of enzastaurin in combination with cytotoxic drugs is warranted in NSCLC.
Collapse
Affiliation(s)
- Yun Oh
- Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Calvani M, Trisciuoglio D, Bergamaschi C, Shoemaker RH, Melillo G. Differential involvement of vascular endothelial growth factor in the survival of hypoxic colon cancer cells. Cancer Res 2008; 68:285-91. [PMID: 18172321 DOI: 10.1158/0008-5472.can-07-5564] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The recent approval of bevacizumab (Avastin), a humanized anti-vascular endothelial growth factor (VEGF) monoclonal antibody, in combination with chemotherapy for the treatment of patients with metastatic colorectal cancer, has provided proof of principle of the efficacy of antiangiogenic strategies for cancer therapy. The activity of bevacizumab is primarily attributed to its ability to inhibit endothelial cell survival. Whether anti-VEGF strategies may also have a direct effect on cancer cell survival is poorly understood. We show that serum-starved colon cancer cells differentially respond to autocrine production of VEGF with the induction of hypoxia inducible factor-1 alpha (HIF-1 alpha) and survival under hypoxic conditions. Inhibition of VEGF or VEGF receptor 2 (VEGFR2)/KDR, but not VEGFR1/Flt-1, was sufficient to abrogate VEGF-mediated induction of HIF-1 alpha and survival in sensitive HCT116, but not in resistant HT29, colon cancer cells. These results provide evidence that a VEGF/KDR/HIF-1 alpha autocrine loop differentially mediates survival of hypoxic colon cancer cells, and they suggest that colon cancer cells may be intrinsically sensitive or resistant to anti-VEGF strategies, which may determine the therapeutic efficacy of bevacizumab.
Collapse
Affiliation(s)
- Maura Calvani
- Developmental Therapeutics Program, National Cancer Institute-Frederick, Frederick, Maryland 21702, USA
| | | | | | | | | |
Collapse
|
32
|
Thanos CG, Schneider PA, Bintz BE, Jensen R, Bryant B, Bell WJ, Hudak J, Emerich DF. TheIn VitroExpression and Secretion of Vascular Endothelial Growth Factor from Free and Alginate-Polyornithine Encapsulated Choroid Plexus Epithelium. ACTA ACUST UNITED AC 2007; 13:747-56. [PMID: 17432950 DOI: 10.1089/ten.2006.0294] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The choroid plexus (CP) is a transplantable cell source secreting tropic and trophic factors for the treatment of brain and peripheral trauma characterized by cellular loss or dysfunction. Here we characterize the expression and secretion of vascular endothelial growth factor (VEGF) from neonatal porcine CP. Light and electron microscopy revealed that enzymatic digestion of the CP produced a preparation consisting primarily of epithelial cells without notable contaminating cells. Microarray analysis, quantitative polymerase chain reaction, and enzyme-linked immunosorbent assay were used to quantify the nuclear, cytoplasmic, and secretory compartmentalization of VEGF. In vitro, the kinetics of VEGF release were orderly, with stepwise increases in secretion over time. The secretory profile of VEGF from CP grown in configurations ranging from a simple monolayer to free-floating 3-dimensional clusters to clusters encapsulated within alginate-polyornithine microcapsules was similar. VEGF output was not affected notably when the cells were maintained in 90% stress medium or in other maintenance media devoid of serum proteins. Secreted VEGF was bioactive, as confirmed by demonstrating its continued ability to proliferate co-cultured human umbilical vascular endothelial cells. The robust ability of these cells to continue to secrete VEGF (and presumably other bioactive proteins) across a variety of dimensional configurations and medium types has implications for their use in clinical indications requiring novel and imaginative use of engineered ectopic transplant sites.
Collapse
|
33
|
Arnoys EJ, Wang JL. Dual localization: proteins in extracellular and intracellular compartments. Acta Histochem 2007; 109:89-110. [PMID: 17257660 DOI: 10.1016/j.acthis.2006.10.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2006] [Revised: 10/01/2006] [Accepted: 10/09/2006] [Indexed: 12/24/2022]
Abstract
The goal of this article is to provide a comprehensive catalog of those proteins documented to exhibit dual localization, being found in both the extracellular compartment (cell surface and extracellular medium) as well as the intracellular compartment (cytosol and nucleus). A large subset of these proteins that show dual localization is found both in the nucleus and outside of cells. Proteins destined to be secreted out of the cell or to be expressed at the cell surface usually enter the endomembrane pathway on the basis of a signal sequence that targets them into the endoplasmic reticulum. Proteins destined for import into the nucleus, on the other hand, usually carry a nuclear localization signal. We have organized our catalog in terms of the presence and absence of these trafficking signals: (a) proteins that contain a signal sequence but no nuclear localization signal; (b) proteins that contain both a signal sequence as well as a nuclear localization signal; (c) proteins that contain a nuclear localization signal but lack a signal sequence; and (d) proteins containing neither a signal sequence nor a nuclear localization signal. Novel insights regarding the activities of several classes of proteins exhibiting dual localization can be derived when one targeting signal is experimentally abrogated. For example, the mitogenic activity of both fibroblasts growth factor-1 and schwannoma-derived growth factor clearly requires nuclear localization, independent of the activation of the receptor tyrosine kinase signaling pathway. In addition, there is a growing list of integral membrane receptors that undergo translocation to the nucleus, with bona fide nuclear localization signals and transcription activation activity. The information provided in this descriptive catalog will, hopefully, stimulate investigations into the pathways and mechanisms of transport between these compartments and the physiological significance of dual localization.
Collapse
Affiliation(s)
- Eric J Arnoys
- Department of Chemistry and Biochemistry, Calvin College, Grand Rapids, MI 49546, USA
| | | |
Collapse
|
34
|
Backer MV, Patel V, Jehning BT, Claffey KP, Backer JM. Surface immobilization of active vascular endothelial growth factor via a cysteine-containing tag. Biomaterials 2006; 27:5452-8. [PMID: 16843524 DOI: 10.1016/j.biomaterials.2006.06.025] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2006] [Accepted: 06/29/2006] [Indexed: 02/04/2023]
Abstract
Developing tissue engineering scaffolds with immobilized growth factors requires facile and reliable methods for the covalent attachment of functionally active proteins. We describe here a new approach to immobilize recombinant proteins based on expression of the protein of interest with a 15-aa long fusion tag (Cys-tag), which avails a free sulfhydryl group for site-specific conjugation. To validate this approach, we conjugated a single-chain vascular endothelial growth factor expressed with an N-terminal Cys-tag (scVEGF) to fibronectin (FN) using a common thiol-directed bi-functional cross-linking agent. We found that the FN-scVEGF conjugate retains VEGF activity similar to that of free scVEGF when used as a soluble ligand. Cells expressing VEGF receptor VEGFR-2 grown on plates coated with FN-scVEGF displayed morphological phenotypes similar to those observed for cells grown on FN in the presence of equivalent amounts of free scVEGF. In addition, 293/KDR cell growth stimulation was observed in the same concentration range with either immobilized or free scVEGF. The effects of immobilized scVEGF, and soluble scVEGF were blocked by NVP-AAD777-NX, a VEGF receptor tyrosine kinase inhibitor. These data indicate that site-specific immobilization via Cys-tag provides a facile and reliable method for permanent deposition of functionally active growth factors on synthetic or protein scaffolds with applications for advanced tissue engineering.
Collapse
|
35
|
Planque N. Nuclear trafficking of secreted factors and cell-surface receptors: new pathways to regulate cell proliferation and differentiation, and involvement in cancers. Cell Commun Signal 2006; 4:7. [PMID: 17049074 PMCID: PMC1626074 DOI: 10.1186/1478-811x-4-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Accepted: 10/18/2006] [Indexed: 12/14/2022] Open
Abstract
Secreted factors and cell surface receptors can be internalized by endocytosis and translocated to the cytoplasm. Instead of being recycled or proteolysed, they sometimes translocate to the nucleus. Nuclear import generally involves a nuclear localization signal contained either in the secreted factor or its transmembrane receptor, that is recognized by the importins machinery. In the nucleus, these molecules regulate transcription of specific target genes by direct binding to transcription factors or general coregulators. In addition to the transcription regulation, nuclear secreted proteins and receptors seem to be involved in other important processes for cell life and cellular integrity such as DNA replication, DNA repair and RNA metabolism. Nuclear secreted proteins and transmembrane receptors now appear to induce new signaling pathways to regulate cell proliferation and differentiation. Their nuclear localization is often transient, appearing only during certain phases of the cell cycle. Nuclear secreted and transmembrane molecules regulate the proliferation and differentiation of a large panel of cell types during embryogenesis and adulthood and are also potentially involved in wound healing. Secreted factors such as CCN proteins, EGF, FGFs and their receptors are often detected in the nucleus of cancer cells. Nuclear localization of these molecules has been correlated with tumor progression and poor prognosis for patient survival. Nuclear growth factors and receptors may be responsible for resistance to radiotherapy.
Collapse
Affiliation(s)
- Nathalie Planque
- Laboratoire d'Oncologie Virale et Moléculaire, Université Paris7-Denis Diderot, UFR de Biochimie, 2 place Jussieu, 75005 Paris, France.
| |
Collapse
|
36
|
Dardik R, Inbal A. Complex formation between tissue transglutaminase II (tTG) and vascular endothelial growth factor receptor 2 (VEGFR-2): Proposed mechanism for modulation of endothelial cell response to VEGF. Exp Cell Res 2006; 312:2973-82. [PMID: 16914140 DOI: 10.1016/j.yexcr.2006.05.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2005] [Revised: 04/25/2006] [Accepted: 05/22/2006] [Indexed: 10/24/2022]
Abstract
We have recently demonstrated that thrombin-activated FXIII (FXIIIA-subunit), a plasma transglutaminase, activates VEGFR-2 by crosslinking it with the alpha(v)beta(3) integrin on the surface of endothelial cells (EC), thereby stimulating angiogenesis. Tissue transglutaminase (tTG), which is functionally and structurally related to FXIIIA, is expressed by numerous cell types, among them EC. However, its role in EC function has not been fully characterized. In the present study, we investigated the potential involvement of tTG in angiogenesis. Using co-immunoprecipitation and immunofluorescent staining experiments, we observed that tTG forms a complex with VEGFR-2 on the cell surface and within the cytoplasm of EC. Stimulation of EC with VEGF resulted in translocation of the tTG-VEGFR-2 complex from the cytoplasm to the nucleus. In VEGF-treated cells, tTG-VEGFR-2 interaction resulted in incorporation of VEGFR-2 into high molecular weight crosslinked complex (es), as revealed by an antibody against gamma-glutamyl-epsilon-lysine isopeptide bond. tTG -VEGFR-2 association was inhibited by a specific VEGFR-2 protein tyrosine kinase inhibitor (PTKI ), as well as by cystamine, inhibitor of the transglutaminase activity of tTG, but not by bacitracin which inhibits the protein-disulfide isomerase (PDI) activity of tTG. Furthermore, cystamine completely abolished the VEGF-induced nuclear translocation of the tTG-VEGFR-2 complex. Blockade of the crosslinking activity of tTG by cystamine enhanced VEGF-induced migration of EC in Boyden chamber by 31% (P < 0.02), and prolonged VEGF-induced signaling response, as demonstrated by sustained activation of the MAP kinase ERK. Taken together, our findings suggest that endothelial cell tTG might be involved in modulation of the cellular response to VEGF by forming an intracellular complex with VEGFR-2, and mediating its translocation into the nucleus upon VEGF stimulation.
Collapse
Affiliation(s)
- Rima Dardik
- Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer 52621, Israel
| | | |
Collapse
|
37
|
Abstract
Angiogenesis, the generation of new blood vessels from pre-existing vessels, is an integral component of wound healing, responses to inflammation and other physiologic processes. It is also an essential part of tumor growth; in the absence of new vessel formation, tumors cannot expand beyond a small volume. Although much is known about angiogenesis and its regulation, there is no overall theory that describes or explains this process. It is here suggested that the intracrine hypothesis, which ascribes to certain extracellular signaling peptides (whether hormones, growth factors, DNA-binding proteins or enzymes) a role in both intracellular biology and extracellular signaling, can contribute to a more general understanding of angiogenesis. Intracrine factors participate in angiogenesis in the following ways: (1) they can act within the cells that synthesized them (type I intracrine action), (2) they can be secreted and then taken up by their cell of synthesis to act intracellularly (type II intracrine action ), or (3) they can be secreted and internalized by a distant target cell (type III intracrine action). The parallels between the intracrine growth factor mechanisms cancer cells employ in stimulating their own growth and the mechanisms operative in endothelial cell proliferation during angiogenesis ("intracrine reciprocity") are discussed. Collectively, these explorations lead to testable hypotheses regarding the regulation of normal and pathological angiogenesis, and point to similarities between tumor-induced angiogenesis and tissue differentiation.
Collapse
Affiliation(s)
- Richard N Re
- Research Division, Ochsner Clinic Foundation, New Orleans, LA 70121, USA.
| | | |
Collapse
|
38
|
Blazquez C, Cook N, Micklem K, Harris AL, Gatter KC, Pezzella F. Phosphorylated KDR can be located in the nucleus of neoplastic cells. Cell Res 2006; 16:93-8. [PMID: 16467880 DOI: 10.1038/sj.cr.7310012] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
KDR (kinase insert domain receptor) phosphorylation induces several effects which lead eventually to cell proliferation and survival. The precise mechanisms by which KDR, once it is activated, communicates with the nucleus are starting to be understood but have not yet been completely unravelled. Two in vitro studies on animal cell lines reported in the literature have demonstrated that, following stimulation with VEGF, KDR is actually translocated within the nucleus. Our aim was to investigate whether this translocation occurs in human cells both in vitro and in vivo. Using laser scanning confocal microscopy, a variable nuclear localization of phosphorylated and total KDR in cell lines and tumour samples was found. In human neoplastic cell lines, hypoxic stimulation greatly increased the nuclear amount of total KDR but less so that of the phosphorylated form. Only after hypoxia and VEGF stimulation there was a comparably increased expression of phosphorylated and total KDR observed in the nuclei of these cells. We conclude that neoplastic cells show a variable expression of total and phosphorylated KDR in the nucleus. The precise functional meaning of nuclear location remains to be established.
Collapse
Affiliation(s)
- Cristina Blazquez
- Departamento de Bioquimica y Biologia Molecular I, Facultad de Biologia, Universidad Complutense de Madrid, Ciudad Universitaria, Madrid 28040, Espania, Spain
| | | | | | | | | | | |
Collapse
|
39
|
Mukdsi JH, De Paul AL, Gutiérrez S, Roth FD, Aoki A, Torres AI. Subcellular localisation of VEGF in different pituitary cells. Changes of its expression in oestrogen induced prolactinomas. J Mol Histol 2006; 36:447-54. [PMID: 16733790 DOI: 10.1007/s10735-005-9012-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2005] [Accepted: 11/23/2005] [Indexed: 01/06/2023]
Abstract
Vascular endothelial growth factor (VEGF) is an important angiogenic factor in the pituitary gland. The objective of this study was to unveil the VEGF subcellular localisation in different pituitary cell types and to evaluate changes in its expression at different time intervals after oestrogen stimulation. A relevant feature demonstrated was the identification of this cytokine in the nucleus and cytoplasm of lactotrophs, somatotrophs and gonadotrophs, as well as in follicle-stellate cells of male rats. Oestrogen treatment increased the number of VEGF immunopositive cells and its expression detected differentially by western blot in both nucleus and cytoplasm of pituitary cells when compared to the control. At ultrastructural level VEGF appeared associated with nucleolus and euchromatin involving a possible internal autocrine loop. In lactotrophs, the predominant cell of the tumour, VEGF was immunodetected in RER, Golgi complex, and vesicular organelles, supporting further the association with an auto-paracrine effect exerted by VEGF. The nucleus/cytoplasm ratio of VEGF revealed a prevalent accumulation of VEGF in the cytoplasm. The presence of VEGF in the nucleus may probably be associated with a translocation to this cell compartment. This study demonstrated a cytoplasmic and nuclear immunolocalisation of VEGF in normal and tumoural adenohypophyseal cells. In the course of prolactinoma development, the oestrogen stimulated VEGF expression in tumoural cells, promoting a vascular adaptation which contributes to growth and progression of the tumour.
Collapse
Affiliation(s)
- Jorge Humberto Mukdsi
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | | | | | | | | |
Collapse
|
40
|
Mitola S, Brenchio B, Piccinini M, Tertoolen L, Zammataro L, Breier G, Rinaudo MT, den Hertog J, Arese M, Bussolino F. Type I collagen limits VEGFR-2 signaling by a SHP2 protein-tyrosine phosphatase-dependent mechanism 1. Circ Res 2005; 98:45-54. [PMID: 16339483 DOI: 10.1161/01.res.0000199355.32422.7b] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
During angiogenesis, a combined action between newly secreted extracellular matrix proteins and the repertoire of integrins expressed by endothelial cells contributes in the regulation of their biological functions. Extracellular matrix-engaged integrins influence tyrosine kinase receptors, thus promoting a regulatory cross-talk between adhesive and soluble stimuli. For instance, vitronectin has been reported to positively regulate VEGFR-2. Here, we show that collagen I downregulates VEGF-A-mediated VEGFR-2 activation. This activity requires the tyrosine phosphatase SHP2, which is recruited to the activated VEGFR-2 when cells are plated on collagen I, but not on vitronectin. Constitutive expression of SHP2(C459S) mutant inhibits the negative role of collagen I on VEGFR-2 phosphorylation. VEGFR-2 undergoes internalisation, which is associated with dynamin II phosphorylation. Expression of SHP2(C459S) impairs receptor internalisation suggesting that SHP2-dependent dephosphorylation regulates this process. These findings demonstrate that collagen I in provisional extracellular matrix surrounding nascent capillaries triggers a signaling pathway that negatively regulates angiogenesis.
Collapse
Affiliation(s)
- Stefania Mitola
- Institute for Cancer Research and Treatment, Department of Oncological Sciences, University of Torino, Turin, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Crombez M, Chevallier P, -Gaudreault RC, Petitclerc E, Mantovani D, Laroche G. Improving arterial prosthesis neo-endothelialization: application of a proactive VEGF construct onto PTFE surfaces. Biomaterials 2005; 26:7402-9. [PMID: 16005960 DOI: 10.1016/j.biomaterials.2005.05.051] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The formation of a confluent endothelium on expanded polytetrafluoroethylene (PTFE) vascular prostheses has never been observed. This lack of endothelialization is known to be one of the main reasons leading to the development of thromboses and/or intimal hyperplasia. In this context, several efforts were put forward to promote endothelial cell coverage on the internal surface of synthetic vascular prostheses. The goal of the present study was to immobilize the vascular endothelial growth factor (VEGF) onto Teflon PTFE surfaces to generate a proactive polymer construct favoring interaction with endothelial cells. An ammonia plasma treatment was first used to graft amino groups on PTFE films. Subsequent reactions were performed to covalently bind human serum albumin (HSA) on the polymer surface and to load this protein with negative charges, which allows adsorbtion of VEGF onto HSA via strong electrostatic interactions. X-ray photoelectron spectroscopy (XPS) experiments along with surface derivatization strategies were performed between each synthesis step to ascertain the occurrence of the various molecules surface immobilization. Finally, the electrostatic binding of VEGF to the negatively charged HSA matrix was performed and validated by ELISA. Endothelial cell adhesion and migration experiments were carried out to validate the potential of this VEGF-containing biological construct to act as a proactive media toward the development of endothelial cells.
Collapse
Affiliation(s)
- M Crombez
- Unité de Biotechnologie et de Bioingénierie, Centre de Recherche de l'Hôpital Saint-François d'Assise, C.H.U.Q., Qué., Canada G1L 3L5
| | | | | | | | | | | |
Collapse
|
42
|
Cooney MM, Tserng KY, Makar V, McPeak RJ, Ingalls ST, Dowlati A, Overmoyer B, McCrae K, Ksenich P, Lavertu P, Ivy P, Hoppel CL, Remick S. A phase IB clinical and pharmacokinetic study of the angiogenesis inhibitor SU5416 and paclitaxel in recurrent or metastatic carcinoma of the head and neck. Cancer Chemother Pharmacol 2004; 55:295-300. [PMID: 15538570 DOI: 10.1007/s00280-004-0871-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Accepted: 06/17/2004] [Indexed: 11/24/2022]
Abstract
PURPOSE SU5416 is a novel small organic molecule that non-competitively inhibits the phosphorylation of the VEGF tyrosine kinase receptor, Flk-1. This phase IB study was performed to determine the safety, pharmacokinetics, and preliminary efficacy of the combination of SU5416 and paclitaxel in recurrent or metastatic carcinoma of the head and neck. METHODS Enrolled in the study were 12 patients with biopsy-proven recurrent or metastatic carcinoma of the head and neck. Six patients received intravenous SU5416 110 mg/m2 on days 1, 15, 18, 22 and 25, and paclitaxel 70 mg/m2 on days 8, 15 and 22. Since two patients experienced a dose-limiting toxicity (DLT) in cohort 1, the next six patients received identical treatment as above except the paclitaxel dose was reduced to 55 mg/m2 per week. RESULTS A total of 42 cycles at two different dose levels were given. In cohort 1 there were two deep venous thromboses that were DLTs. In the second cohort there was a DLT consisting of a transient ischemic attack after receiving SU5416. Most of the other toxicities seen were grade 1 or 2 in nature and consisted of headache, facial flushing, and fatigue. Two patients developed extensive ulcerative cavities at sites of prior radiation. There were no significant changes in the pharmacokinetic parameters of SU5416 given with paclitaxel. Four patients had prolonged freedom from progression of 18, 28, 42, and 60 weeks duration. CONCLUSIONS The combination of SU5416 with paclitaxel had a higher than expected incidence of thromboembolic events and prophylactic anticoagulation should be considered for future trials that combine an angiogenesis inhibitor with cytotoxic chemotherapy. Although the future development of SU5416 as a chemotherapeutic agent is unclear, there was a clinical benefit seen with this combination in 36% of the patients. This trial supports the use of developing antiangiogenic combinations, using molecular targeted agents, in head and neck carcinoma.
Collapse
Affiliation(s)
- Matthew M Cooney
- Developmental Therapeutics Program, CASE Comprehensive Cancer Center, University Hospitals of Cleveland, 211100 Euclid Avenue, Cleveland, OH, 44106, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Islamov RR, Chintalgattu V, Pak ES, Katwa LC, Murashov AK. Induction of VEGF and its Flt-1 receptor after sciatic nerve crush injury. Neuroreport 2004; 15:2117-21. [PMID: 15486493 DOI: 10.1097/00001756-200409150-00024] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Expression of vascular endothelial growth factor and its receptors Flt-1 and Flk-1 was studied in lumbar spinal cord after sciatic nerve crush injury. Immunohistochemical staining revealed strikingly different distribution of VEGF, Flt-1, and Flk-1 in lumbar motor neurons. VEGF was observed both in the nuclei and perikarya, while Flk-1 had cytoplasmic and Flt-1 perinuclear localization. Real-time RT-PCR showed a significant increase in the expression of VEGF and Flt-1 on the injured side of the lumbar spinal cord. The increased level of VEGF was also detected by immunoblot. Here we show that lumbar motor neurons increase the expression of VEGF and Flt-1 in response to injury. We propose that VEGF/Flt-1 signaling may be involved in regeneration of the spinal motor neurons.
Collapse
Affiliation(s)
- Rustem R Islamov
- Department of Physiology, The Brody School of Medicine, East Carolina University, 600 Moye Blvd, Greenville, NC 27834, USA
| | | | | | | | | |
Collapse
|
44
|
Crombez M, Mantovani D. Progresses in synthetic vascular prostheses: toward the endothelialization. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 534:165-77. [PMID: 12903719 DOI: 10.1007/978-1-4615-0063-6_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Affiliation(s)
- Mathilde Crombez
- Bioengineering and Biotechnology Unit, St-François d'Assise Hospital Research Centre and Laval University, Department of Materials Engineering, Laboratory for Biomaterials and Bioengineering, Quebec City, G1K 7P4, Canada
| | | |
Collapse
|
45
|
Zisch AH, Lutolf MP, Ehrbar M, Raeber GP, Rizzi SC, Davies N, Schmökel H, Bezuidenhout D, Djonov V, Zilla P, Hubbell JA. Cell‐demanded release of VEGF from synthetic, biointeractive cell‐ingrowth matrices for vascularized tissue growth. FASEB J 2003; 17:2260-2. [PMID: 14563693 DOI: 10.1096/fj.02-1041fje] [Citation(s) in RCA: 438] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Local, controlled induction of angiogenesis remains a challenge that limits tissue engineering approaches to replace or restore diseased tissues. We present a new class of bioactive synthetic hydrogel matrices based on poly(ethylene glycol) (PEG) and synthetic peptides that exploits the activity of vascular endothelial growth factor (VEGF) alongside the base matrix functionality for cellular ingrowth, that is, induction of cell adhesion by pendant RGD-containing peptides and provision of cell-mediated remodeling by cross-linking matrix metalloproteinase substrate peptides. By using a Michael-type addition reaction, we incorporated variants of VEGF121 and VEGF165 covalently within the matrix, available for cells as they invade and locally remodel the material. The functionality of the matrix-conjugated VEGF was preserved and was critical for in vitro endothelial cell survival and migration within the matrix environment. Consistent with a scheme of locally restricted availability of VEGF, grafting of these VEGF-modified hydrogel matrices atop the chick chorioallontoic membrane evoked strong new blood vessel formation precisely at the area of graft-membrane contact. When implanted subcutaneously in rats, these VEGF-containing matrices were completely remodeled into native, vascularized tissue. This type of synthetic, biointeractive matrix with integrated angiogenic growth factor activity, presented and released only upon local cellular demand, could become highly useful in a number of clinical healing applications of local therapeutic angiogenesis.
Collapse
Affiliation(s)
- Andreas H Zisch
- Department of Materials Science and Institute for Biomedical Engineering, Swiss Federal Institute of Technology Zurich and University of Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ilan N, Tucker A, Madri JA. Vascular endothelial growth factor expression, beta-catenin tyrosine phosphorylation, and endothelial proliferative behavior: a pathway for transformation? J Transl Med 2003; 83:1105-15. [PMID: 12920240 DOI: 10.1097/01.lab.0000083531.84403.8b] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The hypothesis that tumor growth is angiogenesis dependent has been documented by a considerable body of direct and indirect experimental data and has generated intense basic and pharmaceutical-related interest. In contrast, the study of endothelial cell tumors has been modest by comparison. Hemangioma is the most common tumor of any kind seen in infancy and also, perhaps, the least understood. We compared a mouse hemangioma-derived cell line (EOMA) and primary human endothelial cells (HUVEC) for their proliferative behavior and molecular alterations. EOMA cells intrinsically expressed vascular endothelial growth factor (VEGF), which acts in an autocrine manner, resulting in an increase in CD1 expression and cell proliferation, both of which were inhibited by anti-VEGF neutralizing antibodies. Such an autocrine loop is supported by constitutive VEGF receptor (Flk-1) tyrosine phosphorylation, Flk-1 and Flt-1 nuclear localization, and mitogen-activated protein kinase activation. beta-catenin was also found to exhibit significant nuclear localization and constitutively associate with Flk-1 and Flt-1 in EOMA cells but much less so in HUVEC, and immunoprecipitated Flk-1 was able to phosphorylate purified beta-catenin in an immune complex kinase assay. EOMA cells were also noted to express reduced levels of N-cadherin and gamma-catenin compared with HUVEC. Interestingly, sequestration of endogenous VEGF in EOMA cultures resulted in a dramatic decrease in nuclear beta-catenin and a reduction in CD1 levels, whereas addition of exogenous VEGF elicited increased nuclear beta-catenin localization and increased CD1 levels in HUVEC. The possible contributions of VEGF signaling pathways, cell junction component expression levels, and phosphorylation states to endothelial cell transformation and proliferation are discussed.
Collapse
Affiliation(s)
- Neta Ilan
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | |
Collapse
|
47
|
Ito M, Tanaka S, Kim S, Kuwai T, Matsutani N, Kamada T, Kitadai Y, Sumii M, Yoshihara M, Haruma K, Chayama K. The specific expression of hypoxia inducible factor-1alpha in human gastric mucosa induced by nonsteroidal anti-inflammatory drugs. Aliment Pharmacol Ther 2003; 18 Suppl 1:90-98. [PMID: 12925145 DOI: 10.1046/j.1365-2036.18.s1.10.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Hypoxia is a cause of gastric mucosal damage induced by nonsteroidal anti-inflammatory drugs (NSAIDs). The expression of hypoxia inducible factor-1alpha (HIF-1alpha) reflects the status of tissue ischaemia. AIM To investigate the effect of NSAID administration on the expression of HIF-1alpha in human gastric mucosa. METHODS We employed 71 patients including 14 with NSAID administration. The HIF-1alpha expression was estimated by immunohistochemistry using monoclonal antibody (H1alpha67) and raised antiserum (HI-3). Vascular endothelial growth factor expression was also examined by immunohistochemistry. HI-3 recognized hypoxia-induced protein in HeLa cells. RESULTS In human gastric mucosa, HIF-1alpha was mainly expressed in the nuclei of the surface epithelial cells and in the neck zone both by use of HI-3 and of H1alpha67. The expression of vascular endothelial growth factor correlated well with that of HIF-1alpha. The level of HIF-1alpha in the surface epithelium was significantly higher in patients with administration of NSAIDs than those without NSAID use (P < 0.001) both in the gastric corpus and antrum. Helicobacter pylori infection did not affected the levels of HIF-1alpha. Long-term administration of rebamipide reduced the level of HIF-1alpha. CONCLUSION HIF-1alpha expression is a new biological marker of ischaemia especially in NSAID-related gastric lesions.
Collapse
Affiliation(s)
- M Ito
- Department of Medicine and Molecular Science, Graduate School of Biomedical Science, Hiroshima University, Hiroshima, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Li W, Keller GA. An antibody to VEGF upregulates factor VIII via interleukin-1 in activated adrenal cortex-derived capillary endothelial cells. Int Immunopharmacol 2003; 3:493-512. [PMID: 12689655 DOI: 10.1016/s1567-5769(03)00002-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We have previously shown in an in vitro wounding system of cultured endothelial cells (EC) that vascular endothelial growth factor (VEGF(165)) treatment upregulated the level of factor VIII (FVIII) in the cells facing an experimental wound. The FVIII upregulation induced by VEGF(165) could be abolished by rhuMab VEGF, a humanized antibody that blocks VEGF functions and inhibits tumorigenesis. Because the thrombotic system is actively involved in angiogenesis, we further investigated the effects of rhuMab VEGF on the regulation of FVIII. Although non-disturbed cells distant from a wound were not affected by rhuMab VEGF treatment, FVIII was also significantly upregulated in the cells along the wound in cultures treated with the antibody alone. RhuMab VEGF stimulation of FVIII could be blocked by VEGF(165). When cells were treated with rhuMab VEGF and VEGF(165) combined together, the stimulation or inhibition of FVIII expression was dose-related and dependent on the amount of excess free rhuMab VEGF and rhuMab VEGF:VEGF(165) immune complexes. Thus, both VEGF(165) and rhuMab VEGF used as single reagents enhanced FVIII in activated endothelial cells, while the immune complexes suppressed the upregulation. Following rhuMab VEGF treatment, two distinct activated endothelial subpopulations that differ in their ability to internalize rhuMab VEGF and express interleukin-1 (IL-1) were identified. IL-1beta but not IL-1alpha appeared to be acting as a mediator between the two endothelial subpopulations as the FVIII upregulation induced by rhuMab VEGF could be totally abolished by treatment with type II soluble IL-1 receptor (sIL-1RII).
Collapse
Affiliation(s)
- Wenlu Li
- Department of Toxicology, Genentech, Inc, 1 DNA Way, South San Francisco, CA 94080, USA.
| | | |
Collapse
|
49
|
|
50
|
Olsnes S, Klingenberg O, Wiedłocha A. Transport of exogenous growth factors and cytokines to the cytosol and to the nucleus. Physiol Rev 2003; 83:163-82. [PMID: 12506129 DOI: 10.1152/physrev.00021.2002] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In recent years a number of growth factors, cytokines, protein hormones, and other proteins have been found in the nucleus after having been added externally to cells. This review evaluates the evidence that translocation takes place and discusses possible mechanisms. As a demonstration of the principle that extracellular proteins can penetrate cellular membranes and reach the cytosol, a brief overview of the penetration mechanism of protein toxins with intracellular sites of action is given. Then problems and pitfalls in attempts to demonstrate the presence of proteins in the cytosol and in the nucleus as opposed to intracellular vesicular compartments are discussed, and some new approaches to study this are described. A detailed overview of the evidence for translocation of fibroblast growth factor, HIV-Tat, interferon-gamma, and other proteins where there is evidence for intracellular action is given, and translocation mechanisms are discussed. It is concluded that although there are many pitfalls, the bulk of the experiments indicate that certain proteins are indeed able to enter the cytosol and nucleus. Possible roles of the internalized proteins are discussed.
Collapse
Affiliation(s)
- Sjur Olsnes
- Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, Oslo, Norway.
| | | | | |
Collapse
|