1
|
Martins RS, Weber J, Drake L, Latif MJ, Poulikidis K, Razi SS, Luo J, Bhora FY. Improved Composite Hydrogel for Bioengineered Tracheal Graft Demonstrates Effective Early Angiogenesis. J Clin Med 2024; 13:5148. [PMID: 39274364 PMCID: PMC11396371 DOI: 10.3390/jcm13175148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
Background/Objectives: Collagen-agarose hydrogel blends currently used in tracheal graft bioengineering contain relatively high concentrations of collagen to withstand mechanical stresses associated with native trachea function (e.g., breathing). Unfortunately, the high collagen content restricts effective cell infiltration into the hydrogel. In this study, we created an improved hydrogel blend with lower concentrations of collagen (<5 mg/mL) and characterized its capacity for fibroblast invasion and angiogenesis. Methods: Four collagen-agarose hydrogel blends were created: 1 mg/mL type 1 collagen (T1C) and 0.25% agarose, 1 mg/mL T1C and 0.125% agarose, 2 mg/mL T1C and 0.25% agarose, and 2 mg/mL T1C and 0.125% agarose. The hydrogel surface was seeded with fibroblasts, while both endothelial cells and fibroblasts (3:1 ratio) were mixed within the hydrogel matrix. We assessed early angiogenesis by observing fibroblast migration and endothelial cell morphology (elongation and branching) at 7 days. In addition, we performed immunostaining for alpha-smooth muscle actin (aSMA) and explored the gene expression of various angiogenic markers (including vascular endothelial growth factor; VEGF). Results: Gels with lower agarose concentrations (0.125%) with 1 or 2 mg/mL T1C were more effective in allowing early attachment and migration of surface-applied fibroblasts compared to gels with higher (0.25%) agarose concentrations. The low-agarose gels also allowed cells to quickly adopt a spread morphology and self-assemble into elongated structures indicative of early angiogenesis, while demonstrating positive immunostaining for aSMA and increased gene expression of VEGF by day 7. Conclusions: Hydrogel blends with collagen and low agarose concentrations may be effective in allowing early cellular infiltration and angiogenesis, making such gels a suitable cell substrate for use in the development of composite bioengineered tracheal grafts. The collagen-agarose hydrogel blend is meant to be cast around a three-dimensional (3D) printed polycaprolactone support structure and wrapped in porcine small intestine submucosa ECM to create an off-the-shelf bioengineered tracheal implant.
Collapse
Affiliation(s)
- Russell Seth Martins
- Division of Thoracic Surgery, Department of Surgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health (HMH) Network, Edison, NJ 08820, USA
| | - Joanna Weber
- Division of Thoracic Surgery, Department of Surgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health (HMH) Network, Edison, NJ 08820, USA
| | - Lauren Drake
- Department of Surgery, Nuvance Health, Danbury, CT 06810, USA
| | - M Jawad Latif
- Division of Thoracic Surgery, Department of Surgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health (HMH) Network, Edison, NJ 08820, USA
| | - Kostantinos Poulikidis
- Division of Thoracic Surgery, Department of Surgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health (HMH) Network, Edison, NJ 08820, USA
| | - Syed Shahzad Razi
- Division of Thoracic Surgery, Department of Surgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health (HMH) Network, Edison, NJ 08820, USA
| | - Jeffrey Luo
- Division of Thoracic Surgery, Department of Surgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health (HMH) Network, Edison, NJ 08820, USA
| | - Faiz Y Bhora
- Division of Thoracic Surgery, Department of Surgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health (HMH) Network, Edison, NJ 08820, USA
| |
Collapse
|
2
|
Amadeo E, Foti S, Camera S, Rossari F, Persano M, Lo Prinzi F, Vitiello F, Casadei-Gardini A, Rimini M. Developing targeted therapeutics for hepatocellular carcinoma: a critical assessment of promising phase II agents. Expert Opin Investig Drugs 2024; 33:839-849. [PMID: 39039690 DOI: 10.1080/13543784.2024.2377321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/03/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide and the first for primary liver tumors. In recent years greater therapeutic advancement was represented by employment of tyrosine kinase inhibitors (TKIs) either in monotherapy or in combination with immune checkpoint inhibitors (ICIs). AREAS COVERED Major attention was given to target therapies in the last couple of years, especially in those currently under phase II trials. Priority was given either to combinations of novel ICI and TKIs or those targeting alternative mutations of major carcinogenic pathways. EXPERT OPINION As TKIs are playing a more crucial role in HCC therapeutic strategies, it is fundamental to further expand molecular testing and monitoring of acquired resistances. Despite the recent advancement in both laboratory and clinical studies, further research is necessary to face the discrepancy in clinical practice.
Collapse
Affiliation(s)
- Elisabeth Amadeo
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Silvia Foti
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Silvia Camera
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Federico Rossari
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Mara Persano
- Medical Oncology, University and University Hospital of Cagliari, Cagliari, Italy
| | - Federica Lo Prinzi
- Operative Research Unit of Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Francesco Vitiello
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Andrea Casadei-Gardini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Margherita Rimini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| |
Collapse
|
3
|
Chasovskikh NY, Chizhik EE. Bioinformatic analysis of biological pathways in coronary heart disease and Alzheimer’s disease. BULLETIN OF SIBERIAN MEDICINE 2023. [DOI: 10.20538/1682-0363-2022-4-193-204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Aim. Using bioinformatic tools, to perform a pathway enrichment analysis in Alzheimer’s disease and coronary heart disease (CHD).Materials and methods. Genes contributing to susceptibility to CHD and Alzheimer’s disease were obtained from the public database DisGeNET (Database of Gene – Disease Associations). A pathway enrichment analysis was performed in the ClueGO Cytoscape plug-in (version 3.6.0) using hypergeometric distribution and the KEGG and Reactome databases.Results. The identified genes contributing to susceptibility to Alzheimer’s disease and CHD are included in 69 common signaling pathways, grouped into the following subgroups: cell death signaling pathways (1); signaling pathways regulating immune responses (2); signaling pathways responsible for fatty acid metabolism (3); signaling pathways involved in the functioning of the nervous system (4), cardiovascular system (5), and endocrine system (6).Conclusion. Following the performed analysis, we identified possible associations between processes involving genetic factors and their products in CHD and Alzheimer’s disease. In particular, we assumed that susceptibility genes involved in the implementation of these pathways regulate apoptosis, production of inflammatory cytokines and chemokines, lipid metabolism, β-amyloid formation, and angiogenesis.
Collapse
|
4
|
Ruszel KP, Zalewski DP, Stępniewski A, Gałkowski D, Bogucki J, Feldo M, Płachno BJ, Kocki J, Bogucka-Kocka A. Next-Generation Sequencing in the Assessment of the Transcriptomic Landscape of DNA Damage Repair Genes in Abdominal Aortic Aneurysm, Chronic Venous Disease and Lower Extremity Artery Disease. Int J Mol Sci 2022; 24:551. [PMID: 36614026 PMCID: PMC9820637 DOI: 10.3390/ijms24010551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
Vascular diseases are one of the most common causes of death and morbidity. Lower extremity artery disease (LEAD), abdominal aortic aneurysm (AAA) and chronic venous disease (CVD) belong to this group of conditions and exhibit various presentations and courses; thus, there is an urgent need for revealing new biomarkers for monitoring and potential treatment. Next-generation sequencing of mRNA allows rapid and detailed transcriptome analysis, allowing us to pinpoint the most pronounced differences between the mRNA expression profiles of vascular disease patients. Comparison of expression data of 519 DNA-repair-related genes obtained from mRNA next-generation sequencing revealed significant transcriptomic marks characterizing AAA, CVD and LEAD. Statistical, gene set enrichment analysis (GSEA), gene ontology (GO) and literature analyses were applied and highlighted many DNA repair and accompanying processes, such as cohesin functions, oxidative stress, homologous recombination, ubiquitin turnover, chromatin remodelling and DNA double-strand break repair. Surprisingly, obtained data suggest the contribution of genes engaged in the regulatory function of DNA repair as a key component that could be used to distinguish between analyzed conditions. DNA repair-related genes depicted in the presented study as dysregulated in AAA, CVD and LEAD could be utilized in the design of new biomarkers or therapies associated with these diseases.
Collapse
Affiliation(s)
- Karol P. Ruszel
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland
| | - Daniel P. Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland
| | - Andrzej Stępniewski
- Ecotech Complex Analytical and Programme Centre for Advanced Environmentally Friendly Technologies, University of Marie Curie-Skłodowska, 39 Głęboka St., 20-612 Lublin, Poland
| | - Dariusz Gałkowski
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, One Robert Wood Johnson Place, New Brunswick, NJ 08903, USA
| | - Jacek Bogucki
- Chair and Department of Organic Chemistry, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland
| | - Marcin Feldo
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland
| | - Bartosz J. Płachno
- Department of Plant Cytology and Embryology, Institute of Botany, Faculty of Biology, Jagiellonian University in Kraków, 9 Gronostajowa St., 30-387 Kraków, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland
| |
Collapse
|
5
|
Chi Y, Wang F, Zhang Y, Shan Z, Tao W, Lian Y, Xin D, Fan Q, Sun Y. Apatinib inhibits tumour progression and promotes antitumour efficacy of cytotoxic drugs in oesophageal squamous cell carcinoma. J Cell Mol Med 2022; 26:1905-1917. [PMID: 35315581 PMCID: PMC8980885 DOI: 10.1111/jcmm.17209] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/26/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
Apatinib, a highly selective inhibitor of vascular endothelial growth factor receptor-2 (VEGFR-2), inhibits the angiogenesis of tumours. The function and mechanism of apatinib in oesophageal squamous cell carcinoma (ESCC) remain unknown. In present study, we found that the development of ESCC in patients was controlled by treatment of combination of apatinib and a chemotherapeutic drug. Moreover, apatinib efficiently promotes cell apoptosis, inhibits cell proliferation, invasion, epithelial-mesenchymal transition (EMT) and activity of the Akt/mTOR pathway in ESCC cells. Western blot analysis showed that apatinib significantly increased vimentin protein levels, decreased Bcl2, matrix metalloproteinase 9 (MMP9), E-cadherin, p-Akt and p-mTOR protein levels in ESCC cells. Furthermore, apatinib enhanced chemosensitivity of cytotoxic drugs paclitaxel (TAX), 5-fluorouracil (5-FU) and cisplatin (DDP) by upregulating expression of vimentin protein, and downregulating expression of Bcl2, MMP9 and E-cadherin protein in vitro. Compared with single-agent groups, the combination of apatinib with each chemotherapeutic drug significantly repressed tumour growth and angiogenesis through blocking the expression of Ki67 and VEGFR-2 in vivo. Taken together, apatinib efficiently inhibits cell growth through blocking Bcl2 and Akt/mTOR pathway, and suppresses metastasis via inhibiting MMP9 and EMT in ESCC cells. Apatinib promoted antitumour effect of chemotherapeutic agents through promoting cell apoptosis and inhibiting EMT and angiogenesis in ESCC.
Collapse
Affiliation(s)
- Yanyan Chi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yana Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengzheng Shan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weili Tao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujin Lian
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dao Xin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Sun
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Malekan M, Ebrahimzadeh MA. Vascular Endothelial Growth Factor Receptors [VEGFR] as Target in Breast Cancer Treatment: Current Status in Preclinical and Clinical Studies and Future Directions. Curr Top Med Chem 2022; 22:891-920. [PMID: 35260067 DOI: 10.2174/1568026622666220308161710] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 12/09/2022]
Abstract
Breast cancer [BC] is one of the most common cancers among women, one of the leading causes of a considerable number of cancer-related death globally. Among all procedures leading to the formation of breast tumors, angiogenesis has an important role in cancer progression and outcomes. Therefore, various anti-angiogenic strategies have developed so far to enhance treatment's efficacy in different types of BC. Vascular endothelial growth factors [VEGFs] and their receptors are regarded as the most well-known regulators of neovascularization. VEGF binding to vascular endothelial growth factor receptors [VEGFRs] provides cell proliferation and vascular tissue formation by the subsequent tyrosine kinase pathway. VEGF/VEGFR axis displays an attractive target for anti-angiogenesis and anti-cancer drug design. This review aims to describe the existing literature regarding VEGFR inhibitors, focusing on BC treatment reported in the last two decades.
Collapse
Affiliation(s)
- Mohammad Malekan
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Pharmaceutical Sciences Research Center, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
7
|
Sales RR, Nogueira BL, Tosatti JAG, Gomes KB, Luizon MR. Do Genetic Polymorphisms Affect Fetal Hemoglobin (HbF) Levels in Patients With Sickle Cell Anemia Treated With Hydroxyurea? A Systematic Review and Pathway Analysis. Front Pharmacol 2022; 12:779497. [PMID: 35126118 PMCID: PMC8814522 DOI: 10.3389/fphar.2021.779497] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/31/2021] [Indexed: 01/23/2023] Open
Abstract
Hydroxyurea has long been used for the treatment of sickle cell anemia (SCA), and its clinical effectiveness is related to the induction of fetal hemoglobin (HbF), a major modifier of SCA phenotypes. However, there is substantial variability in response to hydroxyurea among patients with SCA. While some patients show an increase in HbF levels and an ameliorated clinical condition under low doses of hydroxyurea, other patients present a poor effect or even develop toxicity. However, the effects of genetic polymorphisms on increasing HbF levels in response to hydroxyurea in patients with SCA (Hb SS) have been less explored. Therefore, we performed a systematic review to assess whether single-nucleotide polymorphisms (SNPs) affect HbF levels in patients with SCA treated with hydroxyurea. Moreover, we performed pathway analysis using the set of genes with SNPs found to be associated with changes in HbF levels in response to hydroxyurea among the included studies. The systematic literature search was conducted on Medline/PubMed, EMBASE, Cochrane Central Register of Controlled Trials, Cumulative Index to Nursing and Allied Health Literature (CINAHL), Scopus, and Web of Science. Seven cohort studies were included following our inclusion and exclusion criteria. From the 728 genetic polymorphisms examined in the included studies, 50 different SNPs of 17 genes were found to be associated with HbF changes in patients with SCA treated with hydroxyurea, which are known to affect baseline HbF but are not restricted to them. Enrichment analysis of this gene set revealed reactome pathways with the lowest adjusted p-values and highest combined scores related to VEGF ligand–receptor interactions (R-HSA-194313; R-HSA-195399) and the urea cycle (R-HSA-70635). Pharmacogenetic studies of response to hydroxyurea therapy in patients with SCA are still scarce and markedly heterogeneous regarding candidate genes and SNPs examined for association with HbF changes and outcomes, suggesting that further studies are needed. The reviewed findings highlighted that similar to baseline HbF, changes in HbF levels upon hydroxyurea therapy are likely to be regulated by multiple loci. There is evidence that SNPs in intron 2 of BCL11A affect HbF changes in response to hydroxyurea therapy, a potential application that might improve the clinical management of SCA. Systematic Review Registration: (https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=208790).
Collapse
Affiliation(s)
- Rahyssa Rodrigues Sales
- Graduate Program in Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Rahyssa Rodrigues Sales, ; Marcelo Rizzatti Luizon,
| | - Bárbara Lisboa Nogueira
- Graduate Program in Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jéssica Abdo Gonçalves Tosatti
- Department of Clinical and Toxicological Analyzes, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Karina Braga Gomes
- Department of Clinical and Toxicological Analyzes, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Rizzatti Luizon
- Graduate Program in Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Rahyssa Rodrigues Sales, ; Marcelo Rizzatti Luizon,
| |
Collapse
|
8
|
Palmer BR, Paterson MA, Frampton CM, Pilbrow AP, Skelton L, Pemberton CJ, Doughty RN, Ellis CJ, Troughton RW, Richards AM, Cameron VA. Vascular endothelial growth factor-A promoter polymorphisms, circulating VEGF-A and survival in acute coronary syndromes. PLoS One 2021; 16:e0254206. [PMID: 34260629 PMCID: PMC8279389 DOI: 10.1371/journal.pone.0254206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 06/22/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Development of a competent collateral circulation in established coronary artery disease is cardio-protective. The vascular endothelial growth factor (VEGF) system plays a key role in this process. We investigated the prognostic performance of circulating VEGF-A and three genetic variants in the VEGFA gene in a clinical coronary cohort. METHODS AND RESULTS The Coronary Disease Cohort Study (CDCS) recruited 2,140 patients, with a diagnosis of acute coronary syndrome (ACS), after admission to Christchurch or Auckland City Hospitals between July 2002 and January 2009. We present data for 1927 patients from the cohort genotyped for three SNPs in the VEGF-A gene, rs699947 (C-2578A), rs2010963 (C405G) and rs3025039 (C936T). Plasma VEGF-A concentrations were assayed in a subgroup (n = 550) of CDCS patients (geometric mean 36.6 [34.7-38.5] pg/ml). VEGF-A levels correlated with patient heart rate at baseline (p = 0.034). None of rs699947, rs3025039, nor rs2010963 genotypes were significantly associated with VEGF-A levels, but rs3025039 genotype was positively associated with collateral vessels perfusion according to the Rentrop classification (p = 0.01) and baseline natriuretic peptide levels (p<0.05). Survival in the CDCS cohort was independently associated with baseline VEGF-A levels and (in males) with rs699947 genotype. CONCLUSIONS This study is strongly suggestive that VEGF-A levels have value as a prognostic biomarker in coronary heart disease patients and SNPs in VEGF-A deserve further investigation as prognostic markers and indicators of angiogenic potential influencing the formation of collateral circulation.
Collapse
Affiliation(s)
- Barry R. Palmer
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
- School of Health Sciences, College of Health, Massey University, Wellington, New Zealand
| | - Melinda A. Paterson
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Chris. M. Frampton
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Anna P. Pilbrow
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Lorraine Skelton
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Chris J. Pemberton
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Robert N. Doughty
- Faculty of Medicine and Health Sciences, Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Chris J. Ellis
- Faculty of Medicine and Health Sciences, Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Richard W. Troughton
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - A. Mark Richards
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Vicky A. Cameron
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
9
|
VEGF-R2/Caveolin-1 Pathway of Undifferentiated ARPE-19 Retina Cells: A Potential Target as Anti-VEGF-A Therapy in Wet AMD by Resvega, an Omega-3/Polyphenol Combination. Int J Mol Sci 2021; 22:ijms22126590. [PMID: 34205419 PMCID: PMC8234996 DOI: 10.3390/ijms22126590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 01/10/2023] Open
Abstract
Age-related macular degeneration (AMD) is one of the main causes of deterioration in vision in adults aged 55 and older. In spite of therapies, the progression of the disease is often observed without reverse vision quality. In the present study, we explored whether, in undifferentiated ARPE-19 retinal cells, a disruption of the VEGF receptors (VEGF-R)/caveolin-1 (Cav-1)/protein kinases pathway could be a target for counteracting VEGF secretion. We highlight that Resvega®, a combination of omega-3 fatty acids with an antioxidant, resveratrol, inhibits VEGF-A secretion in vitro by disrupting the dissociation of the VEGF-R2/Cav-1 complex into rafts and subsequently preventing MAPK activation. Moreover, DNA ChIP analysis reveals that this combination prevents the interaction between AP-1 and vegf-a and vegf-r2 gene promoters. By these pathways, Resvega could present a potential interest as nutritional complementation against AMD.
Collapse
|
10
|
Cheng K, Liu CF, Rao GW. Anti-angiogenic Agents: A Review on Vascular Endothelial Growth Factor Receptor-2 (VEGFR-2) Inhibitors. Curr Med Chem 2021; 28:2540-2564. [PMID: 32407259 DOI: 10.2174/0929867327666200514082425] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/18/2020] [Accepted: 04/23/2020] [Indexed: 11/22/2022]
Abstract
Tumor growth inhibition can be achieved by inhibiting angiogenesis, which has been a field of great concern in recent years. Important targets to inhibit angiogenesis include vascular endothelial growth factor receptor (VEGFR) and its homologous tyrosine kinase receptor. Anti-angiogenic therapy based on inhibition of VEGFR-2 is an effective clinical treatment strategy. The research progress of VEGFR-2 inhibitors is reviewed in this paper from the aspects of drug development and chemical synthesis.
Collapse
Affiliation(s)
- Kang Cheng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chen-Fu Liu
- School of Pharmaceutical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Guo-Wu Rao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
11
|
Fernandes I, Melo-Alvim C, Lopes-Brás R, Esperança-Martins M, Costa L. Osteosarcoma Pathogenesis Leads the Way to New Target Treatments. Int J Mol Sci 2021; 22:E813. [PMID: 33467481 PMCID: PMC7831017 DOI: 10.3390/ijms22020813] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is a rare condition with very poor prognosis in a metastatic setting. Basic research has enabled a better understanding of OS pathogenesis and the discovery of new potential therapeutic targets. Phase I and II clinical trials are already ongoing, with some promising results for these patients. This article reviews OS pathogenesis and new potential therapeutic targets.
Collapse
Affiliation(s)
- Isabel Fernandes
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1600 Lisbon, Portugal
| | - Cecília Melo-Alvim
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
| | - Raquel Lopes-Brás
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
| | - Miguel Esperança-Martins
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1600 Lisbon, Portugal
- Sérgio Dias Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1600 Lisbon, Portugal
| | - Luís Costa
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1600 Lisbon, Portugal
| |
Collapse
|
12
|
Wieczór R, Rość D, Wieczór AM, Kulwas A. VASCULAR-1 and VASCULAR-2 as a New Potential Angiogenesis and Endothelial Dysfunction Markers in Peripheral Arterial Disease. Clin Appl Thromb Hemost 2020; 25:1076029619877440. [PMID: 31564130 PMCID: PMC6829630 DOI: 10.1177/1076029619877440] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The quotient of concentrations concerning the key proangiogenic factor, that is, the vascular endothelial growth factor (VEGF-A) and the angiogenesis inhibitor, namely, its soluble receptors (sVEGFR-1 or sVEGFR-2), seems to reflect increased hypoxia and intensity of compensation angiogenesis. Therefore, it can be an ischemic and endothelial dysfunction marker reflected in intermittent claudication (IC) or critical limb ischemia (CLI) in patients with symptomatic peripheral arterial disease (PAD). The main objective of this study was to evaluate the levels of VEGF-A/sVEGFR-1 and VEGF-A/sVEGFR-2—presented using a novelty acronym VASCULAR-1 and VASCULAR-2—in patients with IC and CLI, as well as displayed in 4 classes of severity of PAD. VASCULAR-1 and VASCULAR-2 were calculated using the plasma of venous blood sampled from 80 patients with IC (n = 65) and CLI (n = 15) and the control group (n = 30). Patients with CLI were reported to have a slightly higher index of VASCULAR-1 and double VASCULAR-2 levels as compared to patients with IC (P = nonsignificant), and these markers were significantly higher than controls (P < .01 and P < .01, respectively). VASCULAR-2 levels were observed to have an increasing tendency in the subsequent degrees of PAD severity according to the Fontaine classification (P = .02). In view of the need to consider the role of the proangiogenic and antiangiogenic factor in the assessment of the so-called “angiogenic potential,” VASCULAR-1 ratio and VASCULAR-2 ratio may be a new useful biomarker of limb ischemia in patients with IC and CLI. However, this requires further studies and evidence on a very large group of patients with PAD.
Collapse
Affiliation(s)
- Radosław Wieczór
- Department of Pathophysiology, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.,Dr Jan Biziel University Hospital No. 2, Bydgoszcz, Poland
| | - Danuta Rość
- Department of Pathophysiology, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Anna Maria Wieczór
- Department of Pathophysiology, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Arleta Kulwas
- Department of Pathophysiology, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
13
|
The Active Compounds of Yixin Ningshen Tablet and Their Potential Action Mechanism in Treating Coronary Heart Disease- A Network Pharmacology and Proteomics Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4912395. [PMID: 32419806 PMCID: PMC7204378 DOI: 10.1155/2020/4912395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/15/2019] [Accepted: 10/10/2019] [Indexed: 12/22/2022]
Abstract
Yixin Ningshen tablet is a CFDA-approved TCM formula for treating coronary heart disease (CHD) clinically. However, its active compounds and mechanism of action in treating CHD are unknown. In this study, a novel strategy with the combination of network pharmacology and proteomics was proposed to identify the active components of Yixin Ningshen tablet and the mechanism by which they treat CHD. With the application of network pharmacology, 62 active compounds in Yixin Ningshen tablet were screened out by text mining, and their 313 potential target proteins were identified by a tool in SwissTargetPrediction. These data were integrated with known CHD-related proteomics results to predict the most possible targets, which reduced the 313 potential target proteins to 218. The STRING database was retrieved to find the enriched pathways and related diseases of these target proteins, which indicated that the Calcium, MAPK, PI3K-Akt, cAMP, Rap1, AGE-RAGE, Relaxin, HIF-1, Prolactin, Sphingolipid, Estrogen, IL-17, Jak-STAT signaling pathway, necroptosis, arachidonic acid metabolism, insulin resistance, endocrine resistance, and steroid hormone biosynthesis might be the main pathways regulated by Yixin Ningshen tablet for the treatment of CHD. Through further enrichment analysis and literature study, EGFR, ERBB2, VGFR2, FGF1, ESR1, LOX15, PGH2, HMDH, ADRB1, and ADRB2 were selected and then validated to be the target proteins of Yixin Ningshen tablet by molecular docking, which indicated that Yixin Ningshen tablet might treat CHD mainly through promoting heart regeneration, new vessels' formation, and the blood supply of the myocardial region and reducing cardiac output, oxygen demand, and inflammation as well as arteriosclerosis (promoting vasodilation and intraplaque neoangiogenesis, lowering blood lipid). This study is expected to benefit the clinical application of Yixin Ningshen tablet for the treatment of CHD.
Collapse
|
14
|
Anosmin-1 activates vascular endothelial growth factor receptor and its related signaling pathway for olfactory bulb angiogenesis. Sci Rep 2020; 10:188. [PMID: 31932617 PMCID: PMC6957483 DOI: 10.1038/s41598-019-57040-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
Anosmin-1 is a secreted glycoprotein encoded by the ANOS1 gene, and its loss of function causes Kallmann syndrome (KS), which is characterized by anosmia and hypogonadism due to olfactory bulb (OB) dysfunction. However, the physiological function of anosmin-1 remains to be elucidated. In KS, disordered angiogenesis is observed in OB, resulting in its hypoplasia. In this study, we examined the involvement of anosmin-1 in angiogenic processes. Anosmin-1 was detected on the vessel-like structure in OB of chick embryos, and promoted the outgrowth of vascular sprouts as shown by assays of OB tissue culture. Cell migration, proliferation, and tube formation of endothelial cells were induced by treatment with anosmin-1 as well as vascular endothelial growth factor-A (VEGF-A), and further enhanced by treatment with both of them. We newly identified that anosmin-1 activated VEGF receptor-2 (VEGFR2) by binding directly to it, and its downstream signaling molecules, phospholipase Cγ1 (PLCγ1) and protein kinase C (PKC). These results suggest that anosmin-1 plays a key role in the angiogenesis of developing OB through the VEGFR2–PLCγ1–PKC axis by enhancing the VEGF function.
Collapse
|
15
|
Hernandez-Pacheco N, Guillen-Guio B, Acosta-Herrera M, Pino-Yanes M, Corrales A, Ambrós A, Nogales L, Muriel A, González-Higueras E, Diaz-Dominguez FJ, Zavala E, Belda J, Ma SF, Villar J, Flores C. A vascular endothelial growth factor receptor gene variant is associated with susceptibility to acute respiratory distress syndrome. Intensive Care Med Exp 2018; 6:16. [PMID: 29987654 PMCID: PMC6037659 DOI: 10.1186/s40635-018-0181-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 06/20/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The acute respiratory distress syndrome (ARDS) is one of the main causes of mortality in adults admitted to intensive care units. Previous studies have demonstrated the existence of genetic variants involved in the susceptibility and outcomes of this syndrome. We aimed to identify novel genes implicated in sepsis-induced ARDS susceptibility. METHODS We first performed a prioritization of candidate genes by integrating our own genomic data from a transcriptomic study in an animal model of ARDS and from the only published genome-wide association study of ARDS study in humans. Then, we selected single nucleotide polymorphisms (SNPs) from prioritized genes to conduct a case-control discovery association study in patients with sepsis-induced ARDS (n = 225) and population-based controls (n = 899). Finally, we validated our findings in an independent sample of 661 sepsis-induced ARDS cases and 234 at-risk controls. RESULTS Three candidate genes were prioritized: dynein cytoplasmic-2 heavy chain-1, fms-related tyrosine kinase 1 (FLT1), and integrin alpha-1. Of those, a SNP from FLT1 gene (rs9513106) was associated with ARDS in the discovery study, with an odds ratio (OR) for the C allele of 0.76, 95% confidence interval (CI) 0.58-0.98 (p = 0.037). This result was replicated in an independent study (OR = 0.78, 95% CI = 0.62-0.98, p = 0.039), showing consistent direction of effects in a meta-analysis (OR = 0.77, 95% CI = 0.65-0.92, p = 0.003). CONCLUSIONS We identified FLT1 as a novel ARDS susceptibility gene and demonstrated that integration of genomic data can be a valid procedure to identify novel susceptibility genes. These results contribute to previous firm associations and functional evidences implicating FLT1 gene in other complex traits that are mechanistically linked, through the key role of endothelium, to the pathophysiology of ARDS.
Collapse
Affiliation(s)
- Natalia Hernandez-Pacheco
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Beatriz Guillen-Guio
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Marialbert Acosta-Herrera
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
- Instituto de Parasitología y Biomedicina López-Neyra, IPBLN-CSIC, P.T.S, Granada, Spain
| | - Maria Pino-Yanes
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, La Laguna, Tenerife Spain
| | - Almudena Corrales
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Ambrós
- Intensive Care Unit, Hospital General de Ciudad Real, Ciudad Real, Spain
| | - Leonor Nogales
- Intensive Care Unit, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Arturo Muriel
- Intensive Care Unit, Hospital Universitario Rio Hortega, Valladolid, Spain
| | | | | | - Elizabeth Zavala
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Intensive Care Unit, Hospital Clinic Barcelona, Barcelona, Spain
| | - Javier Belda
- Department of Anesthesiology, Hospital Clínico Universitario, Universidad de Valencia, Valencia, Spain
| | - Shwu-Fan Ma
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, USA
| | - Jesús Villar
- Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
16
|
Formyl Peptide Receptor 1 Modulates Endothelial Cell Functions by NADPH Oxidase-Dependent VEGFR2 Transactivation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2609847. [PMID: 29743977 PMCID: PMC5884202 DOI: 10.1155/2018/2609847] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
In the vasculature, NADPH oxidase is the main contributor of reactive oxygen species (ROS) which play a key role in endothelial signalling and functions. We demonstrate that ECV304 cells express p47phox, p67phox, and p22phox subunits of NADPH oxidase, as well as formyl peptide receptors 1 and 3 (FPR1/3), which are members of the GPCR family. By RT-PCR, we also detected Flt-1 and Flk-1/KDR in these cells. Stimulation of FPR1 by N-fMLP induces p47phox phosphorylation, which is the crucial event for NADPH oxidase-dependent superoxide production. Transphosphorylation of RTKs by GPCRs is a biological mechanism through which the information exchange is amplified throughout the cell. ROS act as signalling intermediates in the transactivation mechanism. We show that N-fMLP stimulation induces the phosphorylation of cytosolic Y951, Y996, and Y1175 residues of VEGFR2, which constitute the anchoring sites for signalling molecules. These, in turn, activate PI3K/Akt and PLC-γ1/PKC intracellular pathways. FPR1-induced ROS production plays a critical role in this cross-talk mechanism. In fact, inhibition of FPR1 and/or NADPH oxidase functions prevents VEGFR2 transactivation and the triggering of the downstream signalling cascades. N-fMLP stimulation also ameliorates cellular migration and capillary-like network formation ability of ECV304 cells.
Collapse
|
17
|
Oak P, Hilgendorff A. The BPD trio? Interaction of dysregulated PDGF, VEGF, and TGF signaling in neonatal chronic lung disease. Mol Cell Pediatr 2017; 4:11. [PMID: 29116547 PMCID: PMC5676585 DOI: 10.1186/s40348-017-0076-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 10/17/2017] [Indexed: 12/26/2022] Open
Abstract
The development of neonatal chronic lung disease (nCLD), i.e., bronchopulmonary dysplasia (BPD) in preterm infants, significantly determines long-term outcome in this patient population. Risk factors include mechanical ventilation and oxygen toxicity impacting on the immature lung resulting in impaired alveolarization and vascularization. Disease development is characterized by inflammation, extracellular matrix remodeling, and apoptosis, closely intertwined with the dysregulation of growth factor signaling. This review focuses on the causes and consequences of altered signaling in central pathways like transforming growth factor (TGF), platelet-derived growth factor (PDGF), and vascular endothelial growth factor (VEGF) driving these above indicated processes, i.e., inflammation, matrix remodeling, and vascular development. We emphasize the shared and distinct role of these pathways as well as their interconnection in disease initiation and progression, generating important knowledge for the development of future treatment strategies.
Collapse
Affiliation(s)
- Prajakta Oak
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany.
- Department of Neonatology, Perinatal Center Grosshadern, Ludwig-Maximilians University, Munich, Germany.
- Center for Comprehensive Developmental Care, Dr. von Haunersches Children's Hospital University, Hospital Ludwig-Maximilians University, Munich, Germany.
| |
Collapse
|
18
|
Krcek R, Matschke V, Theis V, Adamietz IA, Bühler H, Theiss C. Vascular Endothelial Growth Factor, Irradiation, and Axitinib Have Diverse Effects on Motility and Proliferation of Glioblastoma Multiforme Cells. Front Oncol 2017; 7:182. [PMID: 28879167 PMCID: PMC5572260 DOI: 10.3389/fonc.2017.00182] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/08/2017] [Indexed: 01/18/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor. It is highly aggressive with an unfavorable prognosis for the patients despite therapies including surgery, irradiation, and chemotherapy. One important characteristic of highly vascularized GBM is the strong expression of vascular endothelial growth factor (VEGF). VEGF has become a new target in the treatment of GBM, and targeted therapies such as the VEGF-receptor blocker axitinib are in clinical trials. Most studies focus on VEGF-induced angiogenesis, but only very few investigations analyze autocrine or paracrine effects of VEGF on the tumor cells. In this study, we examined the impact of VEGF, irradiation, and axitinib on cell proliferation and cell motility in human GBM cell lines U-251 and U-373. VEGF receptor 2 was shown to be expressed within both cell lines by using PCR and immunochemistry. Moreover, we performed 24-h videography to analyze motility, and a viability assay for cell proliferation. We observed increasing effects of VEGF and irradiation on cell motility in both cell lines, as well as strong inhibiting effects on cellular motility by VEGF-receptor blockade using axitinib. Moreover, axitinib diminished irradiation induced accelerating effects. While VEGF stimulation or irradiation did not affect cell proliferation, axitinib significantly decreased cell proliferation in both cell lines. Therefore, the impairment of VEGF signaling might have a crucial role in the treatment of GBM.
Collapse
Affiliation(s)
- Reinhardt Krcek
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Verena Theis
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Irenäus Anton Adamietz
- Department of Radiotherapy and Radio-Oncology, University Medical Centre Marien Hospital, Ruhr-University Bochum, Herne, Germany
| | - Helmut Bühler
- Institute for Molecular Oncology, Radio-Biology and Experimental Radiotherapy, University Medical Centre Marien Hospital, Ruhr-University Bochum, Herne, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
19
|
Kilpatrick LE, Friedman-Ohana R, Alcobia DC, Riching K, Peach CJ, Wheal AJ, Briddon SJ, Robers MB, Zimmerman K, Machleidt T, Wood KV, Woolard J, Hill SJ. Real-time analysis of the binding of fluorescent VEGF 165a to VEGFR2 in living cells: Effect of receptor tyrosine kinase inhibitors and fate of internalized agonist-receptor complexes. Biochem Pharmacol 2017; 136:62-75. [PMID: 28392095 PMCID: PMC5457915 DOI: 10.1016/j.bcp.2017.04.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/04/2017] [Indexed: 01/01/2023]
Abstract
Vascular endothelial growth factor (VEGF) is an important mediator of angiogenesis. Here we have used a novel stoichiometric protein-labeling method to generate a fluorescent variant of VEGF (VEGF165a-TMR) labeled on a single cysteine within each protomer of the antiparallel VEGF homodimer. VEGF165a-TMR has then been used in conjunction with full length VEGFR2, tagged with the bioluminescent protein NanoLuc, to undertake a real time quantitative evaluation of VEGFR2 binding characteristics in living cells using bioluminescence resonance energy transfer (BRET). This provided quantitative information on VEGF-VEGFR2 interactions. At longer incubation times, VEGFR2 is internalized by VEGF165a-TMR into intracellular endosomes. This internalization can be prevented by the receptor tyrosine kinase inhibitors (RTKIs) cediranib, sorafenib, pazopanib or vandetanib. In the absence of RTKIs, the BRET signal is decreased over time as a consequence of the dissociation of agonist from the receptor in intracellular endosomes and recycling of VEGFR2 back to the plasma membrane.
Collapse
Affiliation(s)
- Laura E Kilpatrick
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Diana C Alcobia
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Chloe J Peach
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Amanda J Wheal
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Stephen J Briddon
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | | | - Jeanette Woolard
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom.
| | - Stephen J Hill
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
20
|
Association of VEGFR-2 Gene Polymorphisms With Clopidogrel Resistance in Patients With Coronary Heart Disease. Am J Ther 2017; 23:e1663-e1670. [PMID: 25738571 DOI: 10.1097/mjt.0000000000000231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR-2) plays a central role in atherogenesis. We investigated the correlation between VEGFR-2 polymorphisms and the risk of clopidogrel resistance (CR) in patients with coronary heart disease (CHD). The study involved 275 patients with CHD undergoing percutaneous coronary intervention and on antiplatelet clopidogrel therapy. The participants were divided into CR group (n = 59) and non-CR group (NCR, n = 216) based on maximum platelet aggregation measurements. VEGFR-2 gene polymorphisms, +1192C>T (rs2305948), +1416T>A (rs1870377), and -271A>G (rs7667298), were genotyped using polymerase chain reaction-restriction fragment length polymorphism. Enzyme-linked immunosorbent assay was used to measure serum transforming growth factor, beta receptor 2 levels. CR was found in 59 patients (20.45%). A significantly higher proportion of patients in the CR group had a history of diabetes mellitus compared with the NCR group (P < 0.05). Genotype and allele frequency of VEGFR-2 +1192C>T (rs2305948) was significantly higher in the CR group than in the NCR group (all P < 0.01). In the VEGFR-2 +1192C>T (rs2305948), the angina pectoris, recurrent myocardial infarction, and combined end point events were significantly more prevalent in the TT carriers than in the CC + CT carriers. In VEGFR-2 -271A>G (rs7667298), the GG carriers had a lower proportion of target lesion revascularization and angina pectoris in contrast to the AA + AG carriers (all P < 0.05). Based on our results, VEGFR-2 +1192C>T (rs2305948) polymorphism is strongly associated with increased CR and main adverse cardiovascular event incidence in patients with CHD undergoing percutaneous coronary intervention. Additionally, patients with CHD with diabetes mellitus history were more likely to develop CR. The associations of +1416T>A (rs1870377) and -271A>G (rs7667298) polymorphisms with CR were inconclusive and will need to be examined further.
Collapse
|
21
|
Kim SH, Pei QM, Jiang P, Yang M, Qian XJ, Liu JB. Effect of active vitamin D3 on VEGF-induced ADAM33 expression and proliferation in human airway smooth muscle cells: implications for asthma treatment. Respir Res 2017; 18:7. [PMID: 28056993 PMCID: PMC5217212 DOI: 10.1186/s12931-016-0490-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 12/11/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Asthma is a chronic respiratory disease characterized by reversible airway obstruction with persistent airway inflammation and airway remodeling, which is associated with increased airway smooth muscle (ASM) mass. Vascular endothelial growth factor (VEGF) has been implicated in inflammatory and airway blood vessel remodeling in asthma. Recent evidence indicates that a deficiency of 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) may influence asthma pathogenesis. A disintegrin and metalloproteinase (ADAM)33 has been identified as playing a role in the pathophysiology of asthma. ADAM33, which is expressed in ASM cells, is suggested to play a role in the function of these cells. Recent studies show that 1,25-(OH)2D3 exerts direct inhibitory effects on passively sensitized human ASM cells in vitro, including inhibition of ADAM33 expression and cell proliferation; however, the mechanism has not been fully understood. METHODS In order to elucidate the precise mechanism underlying the effect of 1,25(OH)2D3 on VEGF-induced ADAM33 expression and ASM cell proliferation, we tested the effects of 1,25(OH)2D3 on cell cycle progression and evaluated the levels of phospho-VEGF receptor 2 (VEGFR2), phospho-extracellular signal-regulated kinase 1/2 (ERK1/2), and phospho-Akt in VEGF-stimulated ASM cells. RESULTS We found that 1,25(OH)2D3 inhibited VEGF-induced ADAM33 expression and ASM cell proliferation, as well as cell cycle arrest. Additionally, VEGF-induced ADAM33 expression and ASM cell proliferation was suppressed via inhibition of ERK1/2 activity, but not that of Akt. Furthermore, 1,25(OH)2D3 treatment inhibited VEGF-induced activation of VEGFR2 as well as that of ERK and Akt in a concentration-dependent manner. 1,25(OH)2D3 also inhibited transforming growth factor (TGF)-β-induced VEGF secretion by ASM cells. CONCLUSIONS Collectively, our findings suggest that 1,25(OH)2D3 inhibits VEGF-induced ASM cell proliferation by suppressing VEGFR2 and ERK1/2 activation and downregulating ADAM33. Further studies of these mechanisms are needed to facilitate the development of treatments for smooth muscle hyperplasia-associated diseases of the airway such as asthma.
Collapse
Affiliation(s)
- Sung-Ho Kim
- Department of Respiration, Tianjin First Central Hospital, Fukanglu-24, Nankaiqu, Tianjin, 300192, China.
| | - Qing-Mei Pei
- Department of Radiology, Tianjin Hospital of Integrated Traditional Chinese and Western Medicine, Tianjin, China
| | - Ping Jiang
- Department of Respiration, Tianjin First Central Hospital, Fukanglu-24, Nankaiqu, Tianjin, 300192, China
| | - Min Yang
- Department of Respiration, Tianjin First Central Hospital, Fukanglu-24, Nankaiqu, Tianjin, 300192, China
| | - Xue-Jiao Qian
- Department of Respiration, Tianjin First Central Hospital, Fukanglu-24, Nankaiqu, Tianjin, 300192, China
| | - Jiang-Bo Liu
- Department of Respiration, Tianjin First Central Hospital, Fukanglu-24, Nankaiqu, Tianjin, 300192, China
| |
Collapse
|
22
|
Carter JJ, Fretwell LV, Woolard J. Effects of 4 multitargeted receptor tyrosine kinase inhibitors on regional hemodynamics in conscious, freely moving rats. FASEB J 2016; 31:1193-1203. [PMID: 27986807 PMCID: PMC5295730 DOI: 10.1096/fj.201600749r] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/03/2016] [Indexed: 11/17/2022]
Abstract
VEGF inhibitors, including receptor tyrosine kinase inhibitors, are used as adjunct therapies in a number of cancer treatments. An emerging issue with these drugs is that most cause hypertension. To gain insight into the physiological mechanisms involved, we evaluated their regional hemodynamic effects in conscious rats. Male Sprague Dawley rats (350–450 g) were chronically implanted with pulsed Doppler flow probes (renal and mesenteric arteries, and the descending abdominal aorta) and catheters (jugular vein, peritoneal cavity, and distal abdominal aorta). Regional hemodynamics were measured over 4 d, before and after daily administration of cediranib (3 and 6 mg/kg, 3 and 6 mg/kg/h for 1 h, i.v.), sorafenib (10 and 20 mg/kg, 10 and 20 mg kg/h for 1 h, i.v.), pazopanib (30 and100 mg/kg, i.p.), or vandetanib (12.5 and 25 mg/kg, i.p.). All drugs evoked significant increases (P < 0.05; n = 7–8) in mean arterial pressure, which were generally accompanied by significant mesenteric and hindquarters, but not renal, vasoconstrictions. The hypertensive effects of cediranib were unaffected by losartan (10 mg/kg/h), bosentan (20 mg/kg/h), or a combination of phentolamine and propranolol (each 1 mg/kg/h), suggesting a need for new strategies to overcome them.—Carter, J. J., Fretwell, L. V., Woolard, J. Effects of 4 multitargeted receptor tyrosine kinase inhibitors on regional hemodynamics in conscious, freely moving rats.
Collapse
Affiliation(s)
- Joanne J Carter
- Cell Signalling Research Group, School of Life Sciences, Medical School, The University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom; and
| | - Laurice V Fretwell
- Faculty of Health and Life Sciences, De Montfort University, Leicester, United Kingdom
| | - Jeanette Woolard
- Cell Signalling Research Group, School of Life Sciences, Medical School, The University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom; and
| |
Collapse
|
23
|
Pei QM, Jiang P, Yang M, Qian XJ, Liu JB, Zheng H, Zhao LH, Kim SH. Upregulation of a disintegrin and metalloproteinase-33 by VEGF in human airway smooth muscle cells: Implications for asthma. Cell Cycle 2016; 15:2819-2826. [PMID: 27579513 PMCID: PMC5053581 DOI: 10.1080/15384101.2016.1220462] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 07/31/2016] [Indexed: 12/14/2022] Open
Abstract
Asthma is a chronic respiratory disease characterized by reversible airway obstruction with persistent airway inflammation and airway remodeling. Features of airway remodeling include increased airway smooth muscle (ASM) mass. A disintegrin and metalloproteinase (ADAM)-33 has been identified as playing a role in the pathophysiology of asthma. ADAM-33 is expressed in ASM cells and is suggested to play a role in the function of these cells. However, the regulation of ADAM-33 is not fully understood. Vascular endothelial growth factor (VEGF) has been implicated in inflammatory and airway blood vessel remodeling in asthmatics. Although VEGF was initially thought of as an endothelial-specific growth factor, recent reports have found that VEGF can promote proliferation of other cell types, including ASM cells. To investigate the precise mechanism of VEGF's effect on ASM cell proliferation, we tested the expression of ADAM-33, phospho-extracellularsignal-regulated kinase 1/2 (ERK1/2), and phospho-Akt in VEGF-stimulated ASM cells. We found that VEGF up-regulates ADAM-33 mRNA and protein levels in a dose- and time-dependent manner as well as phosphorylation of ERK1/2 and Akt. We also found that VEGF-induced ASM cell proliferation is inhibited by both ADAM-33 knockdown and a selective VEGF receptor 2 (VEGFR2) inhibitor (SU1498). Furthermore, VEGF-induced ADAM-33 expression and ASM cell proliferation were suppressed by inhibiting ERK1/2 activity, but not by inhibiting Akt activity. Collectively, our findings suggest that VEGF enhances ADAM-33 expression and ASM cell proliferation by activating the VEGFR2/ERK1/2 signaling pathway, which might be involved in the pathogenesis of airway remodeling. Further elucidation of the mechanisms underlying these observations might help develop therapeutic strategies for airway diseases associated with smooth muscle hyperplasia such as asthma.
Collapse
Affiliation(s)
- Qing-Mei Pei
- Department of Radiology, Tianjin Hospital of Integrated Traditional Chinese and Western Medicine, Tianjin, China
| | - Ping Jiang
- Department of Respiration, Tianjin First Central Hospital, Tianjin, China
| | - Min Yang
- Department of Respiration, Tianjin First Central Hospital, Tianjin, China
| | - Xue-Jiao Qian
- Department of Respiration, Tianjin First Central Hospital, Tianjin, China
| | - Jiang-Bo Liu
- Department of Respiration, Tianjin First Central Hospital, Tianjin, China
| | - Hong Zheng
- Department of Respiration, Tianjin First Central Hospital, Tianjin, China
| | - Li-Hong Zhao
- Department of Respiration, Tianjin First Central Hospital, Tianjin, China
| | - Sung-Ho Kim
- Department of Respiration, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
24
|
Grasso G, Santoro AM, Magrì A, La Mendola D, Tomasello MF, Zimbone S, Rizzarelli E. The Inorganic Perspective of VEGF: Interactions of Cu2+ with Peptides Encompassing a Recognition Domain of the VEGF Receptor. J Inorg Biochem 2016; 159:149-58. [DOI: 10.1016/j.jinorgbio.2016.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/29/2016] [Accepted: 03/12/2016] [Indexed: 12/19/2022]
|
25
|
Liu D, Song J, Ji X, Liu Z, Cong M, Hu B. Association of Genetic Polymorphisms on VEGFA and VEGFR2 With Risk of Coronary Heart Disease. Medicine (Baltimore) 2016; 95:e3413. [PMID: 27175642 PMCID: PMC4902484 DOI: 10.1097/md.0000000000003413] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Coronary heart disease (CHD) is a cardiovascular disease which is contributed by abnormal neovascularization. VEGFA (vascular endothelial growth factor A) and VEGFR2 (vascular endothelial growth factor receptor 2) have been revealed to be involved in the pathological angiogenesis. This study was intended to confirm whether single nucleotide polymorphisms (SNPs) of VEGFA and VEGFR2 were associated with CHD in a Chinese population, considering pathological features and living habits of CHD patients.Peripheral blood samples were collected from 810 CHD patients and 805 healthy individuals. Six tag SNPs within VEGFA and VEGFR2 were obtained from HapMap Database. Genotyping of SNPs was performed using SNapShot method (Applied Biosystems, Foster, CA). Odd ratios (ORs) and their 95% confidence intervals (95% CIs) were calculated to evaluate the association between SNPs and CHD risk.Under the allelic model, 6 SNPs of VEGFA and VEGFR2 were remarkably associated with the susceptibility to CHD. Genotype CT of rs3025039, TT of rs2305948, and AA of rs1873077 were associated with a reduced risk of CHD when smoking, alcohol intake and diabetes were considered, while homozygote GG of rs1570360 might elevate the susceptibility to CHD (all P < 0.05) for patients who were addicted to smoking or those with hypertension. All of the combined effects of rs699947 (CC/CA) and rs2305948 (TT), rs3025039 (TT) and rs2305948 (TT), rs3025039 (CT) and rs1870377 (AA) had positive effects on the risk of CHD, respectively (all P < 0.05). By contrast, the synthetic effects of rs69947 (CA/AA) and rs1870377 (TA), rs699947 (CA) and rs7667298 (GG), rs699947 (AA) and rs7667298 (GG), rs1570360 (GG) and rs2305948 (TT), as well as rs1570360 (GG) and rs1870377 (AA) all exhibited adverse effects on the risk of CHD, respectively (all P < 0.05).Six polymorphisms in VEGFA and VEGFR2 may have substantial influence on the susceptibility to CHD in a Han Chinese population. Prospective cohort studies should be further designed to confirm the above conclusions.
Collapse
Affiliation(s)
- Dongxing Liu
- From the Emergency Department, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | | | | | | | | | | |
Collapse
|
26
|
Zhang B, Qiangba Y, Shang P, Lu Y, Yang Y, Wang Z, Zhang H. Gene expression of vascular endothelial growth factor A and hypoxic adaptation in Tibetan pig. J Anim Sci Biotechnol 2016; 7:21. [PMID: 27042296 PMCID: PMC4818941 DOI: 10.1186/s40104-016-0082-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 03/22/2016] [Indexed: 02/16/2023] Open
Abstract
Background Vascular endothelial growth factor A (VEGFA) can induce endothelial cell proliferation, promote cell migration, and inhibit apoptosis. These processes play key roles in physiological blood vessel formation and pathological angiogenesis. Methods In this study, we examined VEGFA gene expression in the heart, liver, and kidney of Tibetan pigs (TP), Yorkshire pigs that migrated to high altitudes (YH), and Yorkshire pigs that lived at low altitudes (YL). We used PCR and Sanger sequencing to screen for single nucleotide polymorphisms (SNPs) in 5ʹ-flanking DNA and exons of the VEGFA gene. Quantitative real-time PCR and western blots were used to measure expression levels and PCR products were sequenced. Results Results showed that the VEGFA mRNA and protein expression in heart, liver and kidney of TP was higher than that in YH and YL. In addition, the mRNA sequence of the pig VEGFA gene was conserved among pig breeds, and only five SNPs were found in the 5ʹ-flanking region of the VEGFA gene, the allele frequency distributions of the 5 SNPs were not significantly different between the TP, Yorkshire (YL), and Diannan small-ear (DN) pig populations. Conclusion In conclusion, the Tibetan pig showed high levels of VEGFA gene expression in several hypoxic tissues, which suggests that the VEGFA gene may play a major functional role in hypoxic adaptation.
Collapse
Affiliation(s)
- Bo Zhang
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Yangzong Qiangba
- College of Agriculture and Animal Husbandry, Tibet University, Linzhi, 860000 People's Republic of China
| | - Peng Shang
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193 People's Republic of China ; College of Agriculture and Animal Husbandry, Tibet University, Linzhi, 860000 People's Republic of China
| | - Yunfeng Lu
- School of life science & technology, Nanyang normal University, Nanyang, 473061 Henan Province People's Republic of China
| | - Yuzeng Yang
- Hebei Provincial Husbandry and Veterinary Research Institute, Baoding, Hebei 071001 People's Republic of China
| | - Zhixiu Wang
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Hao Zhang
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193 People's Republic of China
| |
Collapse
|
27
|
Carter JJ, Wheal AJ, Hill SJ, Woolard J. Effects of receptor tyrosine kinase inhibitors on VEGF165 a- and VEGF165 b-stimulated gene transcription in HEK-293 cells expressing human VEGFR2. Br J Pharmacol 2015; 172:3141-50. [PMID: 25684635 PMCID: PMC4459029 DOI: 10.1111/bph.13116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/25/2015] [Accepted: 02/10/2015] [Indexed: 01/03/2023] Open
Abstract
Background and Purpose Receptor tyrosine kinase inhibitors (RTKIs) targeted at VEGF receptor 2 (VEGFR2) have proved to be attractive approaches to cancer therapy based on their ability to reduce angiogenesis. Here we have undertaken a quantitative analysis of the interaction of RTKIs and two VEGF splice variants, VEGF165a and VEGF165b, with VEGFR2 by studying nuclear factor of activated T-cells (NFAT) reporter gene activity in live HEK-293 cells. Experimental Approach HEK-293 cells expressing the human VEGFR2 and a firefly luciferase reporter gene regulated by an NFAT response element were used for quantitative analysis of the effect of RTKIs on VEGF165a- and VEGF165b-stimulated luciferase gene expression. Key Results VEGF165a produced a concentration-dependent activation of the NFAT-luciferase reporter gene in living cells that was inhibited in a non-competitive fashion by four different RTKIs (cediranib, pazopanib, sorafenib and vandetanib). The potency obtained for each RTKI from this analysis was similar to those obtained in binding studies using purified VEGFR2 kinase domains. VEGF165b was a lower-efficacy agonist of the NFAT-luciferase response when compared with VEGF165a. Analysis of the concentration–response data using the operational model of agonism indicated that both VEGF165 isoforms had similar affinity for VEGFR2. Conclusions and Implications Quantitative pharmacological analysis of the interaction of VEGF165 isoforms and RTKIs with VEGFR2 in intact living cells has provided important insights into the relative affinity and efficacy of VEGF165a and VEGF165b for activation of the calcineurin- NFAT signalling pathway by this tyrosine kinase receptor.
Collapse
Affiliation(s)
- Joanne J Carter
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Amanda J Wheal
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Stephen J Hill
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Jeanette Woolard
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
28
|
Billinge ER, Platt M. Multiplexed, label-free detection of biomarkers using aptamers and Tunable Resistive Pulse Sensing (AptaTRPS). Biosens Bioelectron 2015; 68:741-748. [PMID: 25682502 DOI: 10.1016/j.bios.2015.02.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/06/2015] [Indexed: 12/26/2022]
Abstract
Diagnostics that are capable of detecting multiple biomarkers are improving the accuracy and efficiency of bioassays. In previous work we have demonstrated the potential of an aptamer-based sensor (aptasensor) utilising Tunable Resistive Pulse Sensing (TRPS). Here, we have advanced the technique identifying key experimental designs for potential POC assays. The assay utilised superparamagnetic beads, and using TRPS monitored their translocations through a pore. If the surfaces of the beads are modified with an aptamer, the frequency of beads (translocations/min) through the pore can be related to the concentration of specific proteins in the solution. Herein, we have demonstrated the successful use of TRPS to observe the binding of two proteins, to their specific aptamers simultaneously. We describe a series of experiments illustrating key factors which we believe are integral to bead-based assays and demonstrate a general method for a multiplexed assay. In summary, we have explored the effects of beads size, concentration, potential bias, pH and aptamer affinity to enhance the sensitivity and practically of a TRPS aptasensor. The method utilises the fact the binding of the aptamer to the protein results in a change in charge density on the bead surface, the isoelectric point of the protein then dominates the mobility of the beads, creating a multiplexed assay termed AptaTRPS. By alteration of the applied potential to the instrument it is possible to produce a positive signal in a simple multiplexed assay.
Collapse
Affiliation(s)
- Emily R Billinge
- Centre of Analytical Sciences, Department of Chemistry, School of Science, Loughborough University, Loughborough, Leicestershire LE11 3TU, United Kingdom
| | - Mark Platt
- Centre of Analytical Sciences, Department of Chemistry, School of Science, Loughborough University, Loughborough, Leicestershire LE11 3TU, United Kingdom.
| |
Collapse
|
29
|
Zhang J, Li L, Li J, Liu Y, Zhang CY, Zhang Y, Zen K. Protein tyrosine phosphatase 1B impairs diabetic wound healing through vascular endothelial growth factor receptor 2 dephosphorylation. Arterioscler Thromb Vasc Biol 2014; 35:163-74. [PMID: 25395617 DOI: 10.1161/atvbaha.114.304705] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Impaired wound healing is a major complication of diabetes mellitus. The mechanisms that govern wound healing, however, are complex and incompletely understood. In the present study, we determined the inhibitory role of protein tyrosine phosphatase 1B (PTP1B) in the process of diabetic wound healing. APPROACH AND RESULTS First, by comparing the wound healing process in PTP1B knockout (PTP1B(-/-)) mice, ob/ob mice and their wild-type littermates in the presence or absence of streptozotocin treatment, we showed that the inhibition of mouse wound healing in streptozotocin-induced diabetic conditions is because of the upregulation and activation of PTP1B. Second, the impaired wound healing in ob/ob mice and streptozotocin-treated wild-type mice was rescued by a PTP1B inhibitor. Third, PTP1B, which is upregulated under hyperglycemic condition, inhibited the tube formation, proliferation, and migration of human microvascular endothelial cells induced by vascular endothelial growth factor, whereas this inhibition was largely abolished by the PTP1B inhibitor. Finally, mechanism study further indicated that PTP1B likely suppressed the proliferation, migration, and tube formation of vascular endothelial cells through dephosphorylation of vascular endothelial growth factor receptor 2. CONCLUSIONS Our study demonstrated that PTP1B negatively modulated the diabetic wound healing process by dephosphorylating the endothelial cell vascular endothelial growth factor receptor 2 and that the specific inhibitor of PTP1B might serve as a potential novel therapeutic tool for diabetic wound healing.
Collapse
Affiliation(s)
- Jing Zhang
- From the State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China (J.Z., L.L., J.L., C.-Y.Z., Y.Z., K.Z.); and Department of Biology, Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta (Y.L.)
| | - Limin Li
- From the State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China (J.Z., L.L., J.L., C.-Y.Z., Y.Z., K.Z.); and Department of Biology, Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta (Y.L.)
| | - Jing Li
- From the State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China (J.Z., L.L., J.L., C.-Y.Z., Y.Z., K.Z.); and Department of Biology, Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta (Y.L.)
| | - Yuan Liu
- From the State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China (J.Z., L.L., J.L., C.-Y.Z., Y.Z., K.Z.); and Department of Biology, Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta (Y.L.)
| | - Chen-Yu Zhang
- From the State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China (J.Z., L.L., J.L., C.-Y.Z., Y.Z., K.Z.); and Department of Biology, Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta (Y.L.).
| | - Yujing Zhang
- From the State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China (J.Z., L.L., J.L., C.-Y.Z., Y.Z., K.Z.); and Department of Biology, Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta (Y.L.).
| | - Ke Zen
- From the State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China (J.Z., L.L., J.L., C.-Y.Z., Y.Z., K.Z.); and Department of Biology, Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta (Y.L.).
| |
Collapse
|
30
|
Ngo DTM, Farb MG, Kikuchi R, Karki S, Tiwari S, Bigornia SJ, Bates DO, LaValley MP, Hamburg NM, Vita JA, Hess DT, Walsh K, Gokce N. Antiangiogenic actions of vascular endothelial growth factor-A165b, an inhibitory isoform of vascular endothelial growth factor-A, in human obesity. Circulation 2014; 130:1072-80. [PMID: 25116954 DOI: 10.1161/circulationaha.113.008171] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Experimental studies suggest that visceral adiposity and adipose tissue dysfunction play a central role in obesity-related cardiometabolic complications. Impaired angiogenesis in fat has been implicated in the development of adipose tissue hypoxia, capillary rarefaction, inflammation, and metabolic dysregulation, but pathophysiological mechanisms remain unknown. In this study, we examined the role of a novel antiangiogenic isoform of vascular endothelial growth factor-A (VEGF-A), VEGF-A165b, in human obesity. METHODS AND RESULTS We biopsied paired subcutaneous and visceral adipose tissue in 40 obese subjects (body mass index, 45±8 kg/m(2); age, 45±11 years) during bariatric surgery and characterized depot-specific adipose tissue angiogenic capacity using an established ex vivo assay. Visceral adipose tissue exhibited significantly blunted angiogenic growth compared with subcutaneous fat (P<0.001) that was associated with marked tissue upregulation of VEGF-A165b (P=0.004). The extent of VEGF-A165b expression correlated negatively with angiogenic growth (r=-0.6, P=0.006). Although recombinant VEGF-A165b significantly impaired angiogenesis, targeted inhibition of VEGF-A165b with neutralizing antibody stimulated fat pad neovascularization and restored VEGF receptor activation. Blood levels of VEGF-A165b were significantly higher in obese subjects compared with lean control subjects (P=0.02), and surgical weight loss induced a marked decline in serumVEGF-A165b (P=0.003). CONCLUSIONS We demonstrate that impaired adipose tissue angiogenesis is associated with overexpression of a novel antiangiogenic factor, VEGF-A165b, that may play a pathogenic role in human adiposopathy. Moreover, systemic upregulation of VEGF-A165b in circulating blood may have wider-ranging implications beyond the adipose milieu. VEGF-A165b may represent a novel area of investigation to gain further understanding of mechanisms that modulate the cardiometabolic consequences of obesity.
Collapse
Affiliation(s)
- Doan T M Ngo
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Melissa G Farb
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Ryosuke Kikuchi
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Shakun Karki
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Stephanie Tiwari
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Sherman J Bigornia
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - David O Bates
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Michael P LaValley
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Naomi M Hamburg
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Joseph A Vita
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Donald T Hess
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Kenneth Walsh
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Noyan Gokce
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.).
| |
Collapse
|
31
|
Manni S, Kisko K, Schleier T, Missimer J, Ballmer-Hofer K. Functional and structural characterization of the kinase insert and the carboxy terminal domain in VEGF receptor 2 activation. FASEB J 2014; 28:4914-23. [PMID: 25114179 DOI: 10.1096/fj.14-256206] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vascular endothelial growth factors (VEGFs) regulate blood and lymphatic vessel development and homeostasis. VEGF receptor 2 (VEGFR-2) is the major receptor involved in vasculogenesis and angiogenesis and regulates endothelial cell survival, migration, and mitogenesis. Ligand-mediated receptor dimerization instigates transmembrane signaling, thereby promoting activation of the intracellular kinase domain. The intracellular part of the receptor comprises the juxtamembrane domain, the catalytic kinase domain, the kinase insert domain (KID), and the carboxy terminal domain (CD). Here we show that the CD inhibits VEGFR-2 activity in the absence of ligand, whereas the KID, particularly a tyrosine residue in this domain (Y951), is indispensable for downstream signaling by the activated kinase. Because of the lack of crystallographic data for the complete kinase domain, we applied size-exclusion chromatography, multiangle laser scattering, analytical ultracentrifugation, and small-angle X-ray scattering to build and functionally validate structural models. Our data show substantial conformational changes of the kinase when it is switched from the inactive, unphosphorylated state to the active, phosphorylated state. Finally, we structurally characterized recombinantly produced protein complexes between VEGFR-2 and T cell-specific adapter protein, a molecule involved in downstream signaling by VEGFR-2.
Collapse
Affiliation(s)
- Sandro Manni
- Biomolecular Research, Molecular Cell Biology, Paul Scherrer Institute, Villigen, Switzerland
| | - Kaisa Kisko
- Biomolecular Research, Molecular Cell Biology, Paul Scherrer Institute, Villigen, Switzerland
| | - Thomas Schleier
- Biomolecular Research, Molecular Cell Biology, Paul Scherrer Institute, Villigen, Switzerland
| | - Jack Missimer
- Biomolecular Research, Molecular Cell Biology, Paul Scherrer Institute, Villigen, Switzerland
| | - Kurt Ballmer-Hofer
- Biomolecular Research, Molecular Cell Biology, Paul Scherrer Institute, Villigen, Switzerland
| |
Collapse
|
32
|
Nagasawa I, Kaneko A, Suzuki T, Nishio K, Kinoshita K, Shiro M, Koyama K. Potential anti-angiogenesis effects of p-terphenyl compounds from Polyozellus multiplex. JOURNAL OF NATURAL PRODUCTS 2014; 77:963-968. [PMID: 24601669 DOI: 10.1021/np401046z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
One novel p-terphenyl compound, polyozellic acid (1), and its acetone adduct (3), along with a known p-terphenyl compound, thelephoric acid (2), were isolated from the mushroom Polyozellus multiplex. Their molecular structures were determined by spectroscopic analysis, X-ray crystallographic analysis, and chemical modification. In some assays related to angiogenesis, compounds 1 and 2 in particular showed inhibitory effects on proliferation, tubule formation, and invasion of human umbilical vein endothelial cells. The quinone moiety within these molecules possibly contributes to their antiangiogenesis activity.
Collapse
Affiliation(s)
- Ikuko Nagasawa
- Department of Pharmacognosy and Phytochemistry, Meiji Pharmaceutical University , Noshio 2-522-1, Kiyose-shi, Tokyo 204-8588, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Role of plasma membrane caveolae/lipid rafts in VEGF-induced redox signaling in human leukemia cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:857504. [PMID: 24738074 PMCID: PMC3967716 DOI: 10.1155/2014/857504] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/21/2014] [Indexed: 12/02/2022]
Abstract
Caveolae/lipid rafts are membrane-rich cholesterol domains endowed with several functions in signal transduction and caveolin-1 (Cav-1) has been reported to be implicated in regulating multiple cancer-associated processes, ranging from tumor growth to multidrug resistance and angiogenesis. Vascular endothelial growth factor receptor-2 (VEGFR-2) and Cav-1 are frequently colocalized, suggesting an important role played by this interaction on cancer cell survival and proliferation. Thus, our attention was directed to a leukemia cell line (B1647) that constitutively produces VEGF and expresses the tyrosine-kinase receptor VEGFR-2. We investigated the presence of VEGFR-2 in caveolae/lipid rafts, focusing on the correlation between reactive oxygen species (ROS) production and glucose transport modulation induced by VEGF, peculiar features of tumor proliferation. In order to better understand the involvement of VEGF/VEGFR-2 in the redox signal transduction, we evaluated the effect of different compounds able to inhibit VEGF interaction with its receptor by different mechanisms, corroborating the obtained results by immunoprecipitation and fluorescence techniques. Results here reported showed that, in B1647 leukemia cells, VEGFR-2 is present in caveolae through association with Cav-1, demonstrating that caveolae/lipid rafts act as platforms for negative modulation of VEGF redox signal transduction cascades leading to glucose uptake and cell proliferation, suggesting therefore novel potential targets.
Collapse
|
34
|
El Amrani M, Lai D, Debbab A, Aly AH, Siems K, Seidel C, Schnekenburger M, Gaigneaux A, Diederich M, Feger D, Lin W, Proksch P. Protein kinase and HDAC inhibitors from the endophytic fungus Epicoccum nigrum. JOURNAL OF NATURAL PRODUCTS 2014; 77:49-56. [PMID: 24328302 DOI: 10.1021/np4005745] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
A chemical investigation of the endophytic fungus Epicoccum nigrum isolated from leaves of Mentha suaveolens collected in Morocco resulted in the isolation of five new polyketides, epicocconigrones A and B (1 and 2), 3-methoxyepicoccone B (3), 3-methoxyepicoccone (4), and 2,3,4-trihydroxy-6-(methoxymethyl)-5-methylbenzaldehyde (5), together with five known compounds (6-10). The structures of the new compounds were unambiguously determined by extensive analysis of the 1D and 2D NMR and mass spectroscopic data. Compounds 1 and 10 showed potent inhibition of at least 15 protein kinases with IC50 values ranging from 0.07 to 9.00 μM. Moreover, compounds 1 and 10 inhibited histone deacetylase (HDAC) activities with IC50 values of 9.8 and 14.2 μM, respectively. A preliminary structure-activity relationship is discussed. Interestingly, compounds 1 and 10 exert mainly cytostatic effects in human lymphoma RAJI and U-937 cell lines.
Collapse
Affiliation(s)
- Mustapha El Amrani
- Institut für Pharmazeutische Biologie und Biotechnologie, Heinrich-Heine-Universität Düsseldorf , Universitätsstrasse 1, Geb. 26.23, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
García-Martín A, Acitores A, Maycas M, Villanueva-Peñacarrillo ML, Esbrit P. Src kinases mediate VEGFR2 transactivation by the osteostatin domain of PTHrP to modulate osteoblastic function. J Cell Biochem 2013; 114:1404-13. [DOI: 10.1002/jcb.24482] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 12/06/2012] [Indexed: 12/12/2022]
|
36
|
Lin Y, Chiba S, Suzuki A, Yamaguchi S, Nakanishi T, Matsumoto H, Ikeda Y, Ishibashi-Ueda H, Hirano KI, Kato S. Vascular smooth muscle cells isolated from adipose triglyceride lipase-deficient mice exhibit distinct phenotype and phenotypic plasticity. Biochem Biophys Res Commun 2013; 434:534-40. [PMID: 23583398 DOI: 10.1016/j.bbrc.2013.03.109] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 03/19/2013] [Indexed: 10/27/2022]
Abstract
The alteration of triglyceride (TG) metabolism in vascular smooth muscle cells (SMC) is likely to be correlated with certain phenotype, though this has not been elucidated. Adipose triglyceride lipase (ATGL) exerts major TG catalytic activity in both adipotic and non-adipotic cells. In the present study, we isolated SMC from ATGL-deficient mice (ATGL(-/-)mSMC). ATGL(-/-)mSMC showed spontaneous TG accumulation with lower mitogenic response and smooth muscle actin (SMA) expression compared to ATGL (+/+)mSMC. Percentage of senescence-associated β-galactosidase positive cells was also increased in ATGL(-/-)mSMC. Real-time PCR followed by screening with focused DNA array analysis revealed up-regulated expression of glucokinase (1.7-fold), lipoprotein lipase (3.8-fold) and interleukin-6 (3.7-fold) and down-regulated expression of vascular endothelial growth factor-A (0.2-fold), type I collagen (0.5-fold), and transforming growth factor-β (0.4-fold) in ATGL(-/-)mSMC compared to ATGL(+/+)mSMC. Next, ectopic gene transfer of human ATGL was attempted using doxycycline (Dox)-regulatable myc-DDK-tagged adenovirus vector (AdvATGL). AdvATGL infection resulted in a reduction of TG accumulation with elevated mitogenic response and SMA expression, and decreased in senescent cell numbers in ATGL(-/-)mSMC. Moreover, deviated gene expression pattern in ATGL(-/-)mSMC was potentially corrected. Our data suggest that ATGL(-/-)mSMC have a distinct phenotype that may be related to vascular pathogenesis. Plasticity of SMC phenotypes correlated to lipid metabolism could be a therapeutic target.
Collapse
Affiliation(s)
- Yanhui Lin
- Department of Pathology and Cell Biology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Combined strategy of endothelial cells coating, Sertoli cells coculture and infusion improves vascularization and rejection protection of islet graft. PLoS One 2013; 8:e56696. [PMID: 23437215 PMCID: PMC3577699 DOI: 10.1371/journal.pone.0056696] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 01/14/2013] [Indexed: 11/19/2022] Open
Abstract
Improving islet graft revascularization and inhibiting rejection become crucial tasks for prolonging islet graft survival. Endothelial cells (ECs) are the basis of islet vascularization and Sertoli cells (SCs) have the talent to provide nutritional support and exert immunosuppressive effects. We construct a combined strategy of ECs coating in the presence of nutritious and immune factors supplied by SCs in a co-culture system to investigate the effect of vascularization and rejection inhibition for islet graft. In vivo, the combined strategy improved the survival and vascularization as well as inhibited lymphocytes and inflammatory cytokines. In vitro, we found the combinatorial strategy improved the function of islets and the effect of ECs-coating on islets. Combined strategy treated islets revealed higher levels of anti-apoptotic signal molecules (Bcl-2 and HSP-32), survival and function related molecules (PDX-1, Ki-67, ERK1/2 and Akt) and demonstrated increased vascular endothelial growth factor receptor 2 (KDR) and angiogenesis signal molecules (FAk and PLC-γ). SCs effectively inhibited the activation of lymphocyte stimulated by islets and ECs. Predominantly immunosuppressive cytokines could be detected in culture supernatants of the SCs coculture group. These results suggest that ECs-coating and Sertoli cells co-culture or infusion synergistically enhance islet survival and function after transplantation.
Collapse
|
38
|
Locascio LE, Donoghue DJ. KIDs rule: regulatory phosphorylation of RTKs. Trends Biochem Sci 2013; 38:75-84. [PMID: 23312584 DOI: 10.1016/j.tibs.2012.12.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 11/29/2012] [Accepted: 12/03/2012] [Indexed: 01/14/2023]
Abstract
Receptor tyrosine kinases (RTKs) are mediators of multiple cell signaling networks linked to cell growth and differentiation. In general, they exhibit similar overall structure with a ligand-binding extracellular domain and a conserved intracellular tyrosine kinase domain. In many RTKs, the kinase domain is interrupted by a sequence known as the kinase insert domain (KID). In addition to phosphorylation sites within the kinase domain, regulatory phosphorylation also occurs within the KID of several RTKs important in human health and disease. Phosphorylation of specific Tyr or Ser residues within the KID of some RTKs triggers distinct cellular signaling outcomes. Here, we review the functionality of KIDs throughout all RTK families, and provide justification for further study of this often-overlooked domain.
Collapse
Affiliation(s)
- Lauren E Locascio
- Department of Chemistry and Biochemistry, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, USA
| | | |
Collapse
|
39
|
Durand JL, Nawrocki AR, Scherer PE, Jelicks LA. Gender differences in adiponectin modulation of cardiac remodeling in mice deficient in endothelial nitric oxide synthase. J Cell Biochem 2013; 113:3276-87. [PMID: 22644792 DOI: 10.1002/jcb.24206] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Left ventricular hypertrophy (LVH) is a risk factor for cardiovascular disease, a leading cause of death. Alterations in endothelial nitric oxide synthase (eNOS), an enzyme involved in regulating vascular tone, and in adiponectin, an adipocyte-derived secretory factor, are associated with cardiac remodeling. Deficiency of eNOS is associated with hypertension and LVH. Adiponectin exhibits vaso-protective, anti-inflammatory, and anti-atherogenic properties. We hypothesized that increased levels of adiponectin would alleviate cardiac pathology resulting from eNOS deficiency, while decreased levels of adiponectin would exacerbate the pathology. Male and female mice, deficient in eNOS, and either lacking or over-expressing adiponectin, were fed high fat diet (HFD) or normal chow. Cardiac magnetic resonance imaging was performed to serially assess heart morphology and function up to 40 weeks of age. Thirty-two weeks of HFD feeding led to significantly greater LV mass in male mice deficient in eNOS and either lacking or over-expressing adiponectin. Heart function was significantly reduced when the mice were deficient in either eNOS, adiponectin or both eNOS and adiponectin; for female mice, heart function was only reduced when both eNOS and adiponectin were lacking. Thus, while over-expression of adiponectin in the eNOS deficient HFD fed male mice preserved function at the expense of significantly increased LV mass, female mice were protected from decreased function and increased LVH by over-expression of adiponectin. Our results demonstrate a sexual dimorphism in response of the heart to alterations in eNOS and adiponectin during high fat feeding and suggest that adiponectin might require eNOS for some of its metabolic effects.
Collapse
Affiliation(s)
- Jorge L Durand
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
40
|
Resch T, Pircher A, Kähler CM, Pratschke J, Hilbe W. Endothelial progenitor cells: current issues on characterization and challenging clinical applications. Stem Cell Rev Rep 2012; 8:926-39. [PMID: 22095429 DOI: 10.1007/s12015-011-9332-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since their discovery about a decade ago, endothelial precursor cells (EPC) have been subjected to intensive investigation. The vision to stimulate respectively suppress a key player of vasculogenesis opened a plethora of clinical applications. However, as research opened deeper insights into EPC biology, the enthusiasm of the pioneer era has been damped in favour of a more critical view. Recent research is focused on three major questions: The fact that the number of EPC in peripheral blood is exceedingly low has consistently raised suspicion whether these cells can plausibly have an impact on physiological or pathophysiological processes. Secondly, whereas the key role of EPC in tumourigenesis has been strongly emphasized by various groups in the past, recent publications are challenging this hypothesis. Thirdly, the lack of consensus on EPC-defining markers and standardized protocols for their detection have repeatedly led to difficulties concerning comparability between papers. In this current review, an overview on recent findings on EPC biology is given, their challenging clinical implications are discussed and the perplexity underlying the current controversial debate is illustrated.
Collapse
Affiliation(s)
- Thomas Resch
- Center of Operative Medicine, Department of Visceral, Transplant, and Thoracic Surgery, Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria.
| | | | | | | | | |
Collapse
|
41
|
Blirando K, Hneino M, Martelly I, Benderitter M, Milliat F, François A. Mast cells and ionizing radiation induce a synergistic expression of inflammatory genes in endothelial cells by a mechanism involving p38α MAP kinase and (p65) NF-κB activation. Radiat Res 2012; 178:556-67. [PMID: 23088768 DOI: 10.1667/rr3058.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Vascular endothelium is a key compartment involved in the development of normal tissue toxicity associated with cancer radiation therapy, i.e., acute inflammation and late fibrosis. Radiation-induced endothelial cell activation has been extensively studied, and activated endothelial cells are characterized by increased expression of inflammatory mediators and adhesion molecules, and activation of the coagulation and thrombosis pathways. However, little is known about the role of vascular endothelium interaction with resident immune cells, such as mast cells on its response to irradiation. Here, we report that endothelial exposure to mast cell conditioned medium and irradiation induces a synergistic expression of many inflammatory genes including interleukin-6 and interleukin-8, CXCL2 and E-selectin. This synergy is blocked by the histamine H1 receptor antagonist mepyramine and partially mimicked by exogenous histamine addition before irradiation. Using pharmacological and molecular inhibition approaches, we show the p38α MAP kinase and p65 (NF-κB) dependence of the synergy. Moreover, our data show a link between both pathways, with p65 (NF-κB) being downstream of p38. These data highlight the possible exacerbation of the radiation-induced endothelial inflammatory response by its interactions with immune cells. It also suggest that p38α MAP kinase and p65 (NF-κB) inhibition in vascular endothelium may limit excessive tissue inflammation induced by radiation therapy, and thereby limit the associated acute and late tissue damage.
Collapse
Affiliation(s)
- Karl Blirando
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety, 92265 Fontenay-aux-Roses, France
| | | | | | | | | | | |
Collapse
|
42
|
Zhang YM, Dai BL, Zheng L, Zhan YZ, Zhang J, Smith WW, Wang XL, Chen YN, He LC. A novel angiogenesis inhibitor impairs lovo cell survival via targeting against human VEGFR and its signaling pathway of phosphorylation. Cell Death Dis 2012; 3:e406. [PMID: 23059825 PMCID: PMC3481133 DOI: 10.1038/cddis.2012.145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Colorectal cancer represents the fourth commonest malignancy, and constitutes a major cause of significant morbidity and mortality among other diseases. However, the chemical therapy is still under development. Angiogenesis plays an important role in colon cancer development. We developed HMQ18–22 (a novel analog of taspine) with the aim to target angiogenesis. We found that HMQ18–22 significantly reduced angiogenesis of chicken chorioallantoic membrane (CAM) and mouse colon tissue, and inhibited cell migration and tube formation as well. Then, we verified the interaction between HMQ18–22 and VEGFR2 by AlphaScreen P-VEGFR assay, screened the targets on angiogenesis by VEGF Phospho Antibody Array, validated the target by western blot and RNAi in lovo cells. We found HMQ18–22 could decrease phosphorylation of VEGFR2(Tyr1214), VEGFR1(Tyr1333), Akt(Tyr326), protein kinase Cα (PKCα) (Tyr657) and phospholipase-Cγ-1 (PLCγ-1) (Tyr771). Most importantly, HMQ18–22 inhibited proliferation of lovo cell and tumor growth in a human colon tumor xenografted model of athymic mice. Compared with normal lovo cells proliferation, the inhibition on proliferation of knockdown cells (VEGFR2, VEGFR1, Akt, PKCα and PLCγ-1) by HMQ18–22 decreased. These results suggested that HMQ18–22 is a novel angiogenesis inhibitor and can be a useful therapeutic candidate for colon cancer intervention.
Collapse
Affiliation(s)
- Y M Zhang
- Institute of Materia Medica, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Olbrich L, Foehring D, Happel P, Brand-Saberi B, Theiss C. Fast rearrangement of the neuronal growth cone's actin cytoskeleton following VEGF stimulation. Histochem Cell Biol 2012; 139:431-45. [PMID: 23052841 DOI: 10.1007/s00418-012-1036-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2012] [Indexed: 12/12/2022]
Abstract
The neuronal growth cone plays a crucial role in the development of the nervous system. This highly motile structure leads the axon to its final destination by translating guidance cues into cytoskeletal rearrangements. Recently, vascular endothelial growth factor (VEGF), which is essential for angiogenesis and vascular sprouting, has been found to exert a trophic activity also on neurons, leading to an increased axonal outgrowth, similar to the well-known nerve growth factor (NGF). The neurotrophic properties of VEGF are likely to be promoted via the VEGF receptor 2 (VEGFR-2) and neuropilin-1 (NRP-1). In the long term, VEGF attracts and influences the growth cone velocity and leads to growth cone enlargement. The present study focuses on immediate VEGF effects using RFP-actin and GFP-NF-M microinjected chicken dorsal root ganglia for live cell imaging of the neuronal growth cone. We analyzed actin and neurofilament dynamics following VEGF and NGF treatment and compared the effects. Furthermore, key signaling pathways of VEGF were investigated by specific blocking of VEGFR-2 or NRP-1. With the aid of confocal laser scanning microscopy and stimulated emission depletion microscopy, we show for the first time that VEGF has a quick effect on the actin-cytoskeleton, since actin rearrangements were identifiable within a few minutes, leading to a dramatically increased motion. Moreover, these effects were strongly enhanced by adding both VEGF and NGF. Most notably, the effects were inhibited by blocking VEGFR-2, therefore we propose that the immediate effects of VEGF on the actin-cytoskeleton are mediated through VEGFR-2.
Collapse
Affiliation(s)
- Laura Olbrich
- Institute of Anatomy and Molecular Embryology, Faculty of Medicine, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | | | | | | | | |
Collapse
|
44
|
Zheng L, He X, Ma W, Dai B, Zhan Y, Zhang Y. Ta1722, an anti-angiogenesis inhibitor targeted on VEGFR-2 against human hepatoma. Biomed Pharmacother 2012; 66:499-505. [DOI: 10.1016/j.biopha.2012.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 05/28/2012] [Indexed: 10/28/2022] Open
|
45
|
Chiu WC, Lin JY, Lee TS, You LR, Chiang AN. β₂-glycoprotein I inhibits VEGF-induced endothelial cell growth and migration via suppressing phosphorylation of VEGFR2, ERK1/2, and Akt. Mol Cell Biochem 2012; 372:9-15. [PMID: 22956423 DOI: 10.1007/s11010-012-1440-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 08/25/2012] [Indexed: 12/26/2022]
Abstract
β(2)-glycoprotein I (β(2)-GPI) is a plasma glycoprotein with diverse functions, but the impact and molecular effects of β(2)-GPI on vascular biology are as yet unclear. Based on the limited information available on the contribution of β(2)-GPI to endothelial cells, we investigated the effect of β(2)-GPI on cell growth and migration in human aortic endothelial cells (HAECs). The regulation of β(2)-GPI as part of intracellular signaling in HAECs was also examined. Vascular endothelial growth factor (VEGF) is a pro-angiogenic factor that may regulate endothelial functions. We found that β(2)-GPI dose-dependently inhibited VEGF-induced endothelial cell growth using the 3-(4,5-dimethylthiazol-2-yl)-2,5-dipenyl tetrazolium bromide assay and cell counts. Using wound healing and Boyden chamber assays, β(2)-GPI remarkably reduced VEGF-increased cell migration at the physiological concentration. Furthermore, β(2)-GPI suppressed VEGF-induced phosphorylation of VEGF receptor 2 (VEGFR2), extracellular signal-regulated kinase 1/2 (ERK1/2), and Akt. These results suggest that β(2)-GPI plays an essential role in the down-regulation of VEGF-induced endothelial responses and may be a useful component for anti-angiogenic therapy.
Collapse
Affiliation(s)
- Wen-Chin Chiu
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155 Section 2, Li-Nong Street, Shih-Pai, Taipei 11221, Taiwan, Republic of China
| | | | | | | | | |
Collapse
|
46
|
Lyzogubov VV, Tytarenko RG, Bora NS, Bora PS. Inhibitory role of adiponectin peptide I on rat choroidal neovascularization. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1264-72. [PMID: 22633972 DOI: 10.1016/j.bbamcr.2012.05.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 03/18/2012] [Accepted: 05/16/2012] [Indexed: 10/28/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of central blindness in the elderly population. The wet type of AMD is characterized by extensive growth of new vessels. One of the effective strategies to treat wet AMD is to limit the choroidal neovascularization (CNV). We studied the effects of adiponectin peptide I (APNpI) on new vessel growth in laser-induced rat model of wet AMD and on rat choroidal endothelial cell (CEC) culture. CNV size and vessel density were investigated by microscopy. Immunohistochemical staining (IHC) for von Willebrand Factor (vWF), APN, APN receptors 1 (AdipoR1), 2 (AdipoR2), VEGF, VEGF receptor 2 (VEGF-R2), proliferating cell nuclear antigen (PCNA) was performed in CNV area. The mRNA expression of VEGF and VEGF-R2 in RPE-choroid was investigated by RT-PCR and real-time PCR. APNpI inhibited area of CNV by 4 fold, number of vWF positive vessels by 99% and area of subretinal tissue by 40%. The expression of VEGF and VEGF-R2 at mRNA and protein levels decreased after APNpI treatment in vivo. Proliferative index (PCNA) was 5 folds less in laser spots of APNpI treated rats compared to controls. In conclusion, APNpI inhibited formation of new vessels in rat model of CNV by decreasing VEGF, VEGF-R2 expression and cell proliferation. Thus, APNpI may have potential therapeutic use for AMD treatment since it significantly inhibited CNV.
Collapse
Affiliation(s)
- Valeriy V Lyzogubov
- Department of Ophthalmology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | | | | |
Collapse
|
47
|
Bruneau S, Woda CB, Daly KP, Boneschansker L, Jain NG, Kochupurakkal N, Contreras AG, Seto T, Briscoe DM. Key Features of the Intragraft Microenvironment that Determine Long-Term Survival Following Transplantation. Front Immunol 2012; 3:54. [PMID: 22566935 PMCID: PMC3342046 DOI: 10.3389/fimmu.2012.00054] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/02/2012] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss how changes in the intragraft microenvironment serve to promote or sustain the development of chronic allograft rejection. We propose two key elements within the microenvironment that contribute to the rejection process. The first is endothelial cell proliferation and angiogenesis that serve to create abnormal microvascular blood flow patterns as well as local tissue hypoxia, and precedes endothelial-to-mesenchymal transition. The second is the overexpression of local cytokines and growth factors that serve to sustain inflammation and, in turn, function to promote a leukocyte-induced angiogenesis reaction. Central to both events is overexpression of vascular endothelial growth factor (VEGF), which is both pro-inflammatory and pro-angiogenic, and thus drives progression of the chronic rejection microenvironment. In our discussion, we focus on how inflammation results in angiogenesis and how leukocyte-induced angiogenesis is pathological. We also discuss how VEGF is a master control factor that fosters the development of the chronic rejection microenvironment. Overall, this review provides insight into the intragraft microenvironment as an important paradigm for future direction in the field.
Collapse
Affiliation(s)
- Sarah Bruneau
- The Division of Nephrology, Transplantation Research Center, Children's Hospital Boston Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Foehring D, Brand-Saberi B, Theiss C. VEGF-induced growth cone enhancement is diminished by inhibiting tyrosine-residue 1214 of VEGFR-2. Cells Tissues Organs 2012; 196:195-205. [PMID: 22433970 DOI: 10.1159/000334600] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2011] [Indexed: 01/10/2023] Open
Abstract
Axonal outgrowth is of paramount significance for establishing the intricate neuronal network both during embryogenesis and nerve regeneration. Vascular endothelial growth factor (VEGF), which is known for its essential role in vascular sprouting and its involvement in cancer, has recently been found to exert a trophic activity on neurons leading to an increased axonal outgrowth. Although two receptors, VEGFR-2 and neuropilin-1, were identified on neurons, the signaling pathways associated with them are not well understood. The aim of this study was to analyze the influence of VEGF on the growth cone morphology and motility of dorsal root ganglia (DRG) neurons. Moreover, we aimed for a deeper understanding of VEGFR-2 on growth cones that potentially mediates the stimulating and attractive effects. We cultivated chicken DRG in medium containing mouse VEGF and analyzed growth cone size. The data presented here show a positive effect of VEGF on growth cone size. Furthermore, we interrupted the activity of VEGFR-2 by either blocking the tyrosine residue 1214 (tyr1214) or by inhibiting the receptor phosphorylation with axitinib, a novel small molecule, which has recently entered phase III trials for cancer treatment. Disruption of the VEGFR-2 leads to a significantly diminished growth cone size. Based on these findings, we propose a positive effect of VEGF on peripheral nervous system growth cone size and show for the first time quantitative data to underline this hypothesis. Additionally, we propose that VEGFR-2 and especially the tyr1214-dependent pathway of VEGFR-2 are of importance in VEGF signaling in the growth cone of DRG neurons.
Collapse
Affiliation(s)
- Daniel Foehring
- Institute of Anatomy and Molecular Embryology, Ruhr-University Bochum, Bochum, Germany
| | | | | |
Collapse
|
49
|
Wuestefeld R, Chen J, Meller K, Brand-Saberi B, Theiss C. Impact of vegf on astrocytes: analysis of gap junctional intercellular communication, proliferation, and motility. Glia 2012; 60:936-47. [PMID: 22431192 DOI: 10.1002/glia.22325] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 02/16/2012] [Indexed: 12/12/2022]
Abstract
The purpose of the present study was to investigate the effects of vascular endothelial growth factor (VEGF) on gap junctional intercellular communication (GJIC), cell proliferation, and cell dynamics in primary astrocytes. VEGF is known as a dimeric polypeptide that potentially binds to two receptors, VEGFR-1 and VEGFR-2, however many effects are mediated by VEGFR-2, for example, actin polymerization, forced cell migration, angiogenesis, and cell proliferation. Recently it has been shown that in case of hypoxia, ischemia or injury VEGF is upregulated to stimulate angiogenesis and cell proliferation. Besides this, VEGF reveals a potent therapeutical target for averting tumor vascularization, emerging in bevacizumab, the first humanized anti-VEGF-A antibody for treating recurrent Glioblastoma multiforme. To expand our knowledge about VEGF effects in glial cells, we cultivated rat astrocytes in medium containing VEGF for 1 and 2 days. To investigate the effects of VEGF on GJIC, we microinjected neurobiotin into a single cell and monitored dye-spreading into adjacent cells. These experiments showed that VEGF significantly enhances astrocytic GJIC compared with controls. Cell proliferation measured by BrdU-labeling also revealed a significant increase of astrocytic mitose rates subsequent to 1 day of VEGF exposure, whereas longer VEGF treatment for 2 days did not have additive effects. To study cell-dynamics of astrocytes subsequent to VEGF treatment, we additionally transfected astrocytes with LifeAct-RFP. Live-cell imaging and quantitative analysis of these cells with aid of confocal laser scanning microscopy revealed higher process movement of VEGF-treated astrocytes. In conclusion, VEGF strongly affects cell proliferation, GJIC, and motility in astrocytes.
Collapse
Affiliation(s)
- Ricarda Wuestefeld
- Institute of Anatomy and Molecular Embryology, Ruhr-University Bochum, Bochum, Germany
| | | | | | | | | |
Collapse
|
50
|
Conway D, Schwartz MA. Lessons from the endothelial junctional mechanosensory complex. F1000 BIOLOGY REPORTS 2012; 4:1. [PMID: 22238515 PMCID: PMC3251317 DOI: 10.3410/b4-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mechanotransduction plays a key role in both normal physiology and in diseases such as cancer, atherosclerosis and hypertension. Nowhere is this more evident than in the vascular system, where fluid shear stress from blood flow plays a critical role in shaping the blood vessels and in determining their function and dysfunction. Responses to flow are mediated in part by a complex of proteins comprised of PECAM-1, VE-cadherin and VEGFR2 at endothelial cell-cell junctions; all proteins that clearly have other, non-mechanical functions. We review recent progress toward understanding the functions and mechanisms of mechanotransduction by this complex and suggest some principles that may apply more broadly.
Collapse
Affiliation(s)
- Daniel Conway
- Department of Microbiology and Cardiovascular Research Center, University of VirginiaCharlottesville VA 22908USA
| | - Martin A. Schwartz
- Department of Microbiology and Cardiovascular Research Center, University of VirginiaCharlottesville VA 22908USA
- Departments of Cell Biology and Biomedical Engineering, University of VirginiaCharlottesville VA 22908USA
- Departments of Medicine and Cell Biology, Yale UniversityNew Haven CT 06511USA
| |
Collapse
|