1
|
Lee C, Kim MJ, Kumar A, Lee HW, Yang Y, Kim Y. Vascular endothelial growth factor signaling in health and disease: from molecular mechanisms to therapeutic perspectives. Signal Transduct Target Ther 2025; 10:170. [PMID: 40383803 PMCID: PMC12086256 DOI: 10.1038/s41392-025-02249-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/09/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
Vascular endothelial growth factor (VEGF) signaling is a critical regulator of vasculogenesis, angiogenesis, and lymphangiogenesis, processes that are vital for the development of vascular and lymphatic systems, tissue repair, and the maintenance of homeostasis. VEGF ligands and their receptors orchestrate endothelial cell proliferation, migration, and survival, playing a pivotal role in dynamic vascular remodeling. Dysregulated VEGF signaling drives diverse pathological conditions, including tumor angiogenesis, cardiovascular diseases, and ocular disorders. Excessive VEGF activity promotes tumor growth, invasion, and metastasis, while insufficient signaling contributes to impaired wound healing and ischemic diseases. VEGF-targeted therapies, such as monoclonal antibodies and tyrosine kinase inhibitors, have revolutionized the treatment of diseases involving pathological angiogenesis, offering significant clinical benefits in oncology and ophthalmology. These therapies inhibit angiogenesis and slow disease progression, but they often face challenges such as therapeutic resistance, suboptimal efficacy, and adverse effects. To further explore these issues, this review provides a comprehensive overview of VEGF ligands and receptors, elucidating their molecular mechanisms and regulatory networks. It evaluates the latest progress in VEGF-targeted therapies and examines strategies to address current challenges, such as resistance mechanisms. Moreover, the discussion includes emerging therapeutic strategies such as innovative drug delivery systems and combination therapies, highlighting the continuous efforts to improve the effectiveness and safety of VEGF-targeted treatments. This review highlights the translational potential of recent discoveries in VEGF biology for improving patient outcomes.
Collapse
Affiliation(s)
- Chunsik Lee
- Department of R&D, GEMCRO Inc, Seoul, Republic of Korea.
| | - Myung-Jin Kim
- Department of Biological Sciences and Research Institute of Women's Health, Sookmyung Women's University, Seoul, Republic of Korea
| | - Anil Kumar
- Center for Research and Innovations, Adichunchanagiri University, Mandya, Karnataka, India
| | - Han-Woong Lee
- Department of R&D, GEMCRO Inc, Seoul, Republic of Korea
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yonghwan Kim
- Department of Biological Sciences and Research Institute of Women's Health, Sookmyung Women's University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Sargunas PR, Ariail E, Lima E Silva R, Patil A, Zhang M, Shen J, Lopes BS, Oh Y, McCue AC, Ramasubramanian R, Stephenson AC, Popel AS, Campochiaro PA, Spangler JB. Bispecific receptor decoy proteins block ocular neovascularization via simultaneous blockade of vascular endothelial growth factor A and C. Mol Ther 2025:S1525-0016(25)00201-1. [PMID: 40143548 DOI: 10.1016/j.ymthe.2025.03.027] [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: 09/26/2024] [Revised: 01/23/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Several debilitating eye diseases that lead to vision loss are driven by ocular neovascularization, which entails abnormal blood vessel growth in the eye. Neovascularization is often induced by the upregulation of vascular endothelial growth factor (VEGF) ligands, which activate angiogenesis through engagement of VEGF receptor (VEGFR) proteins on endothelial cells. Therapeutic interventions that block ocular neovascularization by targeting VEGF ligands, particularly VEGF-A, have revolutionized eye disease treatment. However, a significant population of patients are either non-responders or develop resistance, which can be driven by the upregulation of other VEGF family ligands such as VEGF-C. Here, we engineered two bispecific receptor decoy fusion proteins that incorporate domains of VEGFR-1 and VEGFR-2 for more effective and comprehensive inhibition of VEGF ligands. We demonstrated that our engineered proteins bind all VEGF ligands and can sequester two ligands simultaneously. We further showed that these molecules block VEGF activity to potently inhibit proliferation, migration, and survival of human endothelial cells. Moreover, these receptor decoy proteins significantly reduced ocular neovascularization in two mouse models at doses wherein the current standard-of-care anti-VEGF therapy is ineffective. Collectively, our engineered receptor decoy proteins present a new architecture for VEGF pathway inhibition, offering a promising treatment paradigm for ocular diseases.
Collapse
Affiliation(s)
- Paul R Sargunas
- Department of Chemical and Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Emily Ariail
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Raquel Lima E Silva
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Akash Patil
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Mingliang Zhang
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Jikui Shen
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Beatriz Silva Lopes
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21231, USA
| | - Yuseong Oh
- Department of Chemical and Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Amelia C McCue
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | - A Carson Stephenson
- Department of Chemical and Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21231, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Peter A Campochiaro
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jamie B Spangler
- Department of Chemical and Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
3
|
Das UN. Lipoxin A4 (LXA4) as a Potential Drug for Diabetic Retinopathy. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:177. [PMID: 40005295 PMCID: PMC11857424 DOI: 10.3390/medicina61020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 02/27/2025]
Abstract
The purpose of this review is to propose that lipoxin A4 (LXA4), derived from arachidonic acid (AA), a potent anti-inflammatory, cytoprotective, and wound healing agent, may be useful to prevent and manage diabetic retinopathy (DR). LXA4 suppresses inappropriate angiogenesis and the production of pro-inflammatory prostaglandin E2 (PGE2), leukotrienes (LTs), 12-HETE (12-hydroxyeicosatetraenoic acid), derived from AA by the action of 12-lioxygenase (12-LOX)) interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), as well as the expression of NF-κB, inducible NO (nitric oxide) synthase (iNOS), cyclooxygenase-2 (COX-2), intracellular adhesion molecule-1 (ICAM-1), and vascular endothelial growth factor (VEGF)-factors that play a role in DR. Thus, the intravitreal injection of LXA4 may form a new approach to the treatment of DR and other similar conditions such as AMD (age-associated macular degeneration) and SARS-CoV-2-associated hyperinflammatory immune response in the retina. The data for this review are derived from our previous work conducted in individuals with DR and from various publications on LXA4, inflammation, and DR.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St, Battle Ground, WA 98604, USA
| |
Collapse
|
4
|
Advances in Molecular Regulation of Prostate Cancer Cells by Top Natural Products of Malaysia. Curr Issues Mol Biol 2023; 45:1536-1567. [PMID: 36826044 PMCID: PMC9954984 DOI: 10.3390/cimb45020099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Prostate cancer (PCa) remains both a global health burden and a scientific challenge. We present a review of the molecular targets driving current drug discovery to fight this disease. Moreover, the preventable nature of most PCa cases represents an opportunity for phytochemicals as chemopreventive when adequately integrated into nutritional interventions. With a renovated interest in natural remedies as a commodity and their essential role in cancer drug discovery, Malaysia is looking towards capitalizing on its mega biodiversity, which includes the oldest rainforest in the world and an estimated 1200 medicinal plants. We here explore whether the list of top Malay plants prioritized by the Malaysian government may fulfill the potential of becoming newer, sustainable sources of prostate cancer chemotherapy. These include Andrographis paniculate, Centella asiatica, Clinacanthus nutans, Eurycoma longifolia, Ficus deltoidea, Hibiscus sabdariffa, Marantodes pumilum (syn. Labisia pumila), Morinda citrifolia, Orthosiphon aristatus, and Phyllanthus niruri. Our review highlights the importance of resistance factors such as Smac/DIABLO in cancer progression, the role of the CXCL12/CXCR4 axis in cancer metastasis, and the regulation of PCa cells by some promising terpenes (andrographolide, Asiatic acid, rosmarinic acid), flavonoids (isovitexin, gossypin, sinensetin), and alkylresorcinols (labisiaquinones) among others.
Collapse
|
5
|
Tanaka H, Tanaka K, Takakura S, Enomoto N, Maki S, Ikeda T. Placental growth factor level is correlated with intrapartum fetal heart rate findings. BMC Pregnancy Childbirth 2022; 22:215. [PMID: 35300623 PMCID: PMC8932326 DOI: 10.1186/s12884-022-04562-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 03/10/2022] [Indexed: 11/10/2022] Open
Abstract
Objective Here, we tested the correlation between maternal placental growth factor (PlGF) and fetal heart rate (FHR) monitoring findings. Methods We included 35 women with single pregnancies from 35 to 42 weeks of gestation who were hospitalized owing to onset of labor. Blood samples were collected at the start of labor. Intrapartum FHR monitoring parameters included total deceleration area, average deceleration area (mean deceleration area per 10 min), and five-tier classification level. Results Of the 35 women, 26 (74%) had vaginal delivery and 9 (26%) had cesarean section. After excluding 2 women who had cesarean section for arrest of labor, we analyzed 26 women who had vaginal delivery (VD group) and 7 who had cesarean section for fetal indications (CSF group). PlGF level was significantly higher in the VD group (157 ± 106 pg/ml) than in the CSF group (74 ± 62 pg/ml) (P = 0.03). There were no significant correlations between PlGF and total (r = -0.07) or average (r = -0.08) deceleration area. There was a significant negative correlation (r = -0.42, P = 0.01) between PlGF and the percentage of level 3 or higher in the five-level classification. Conclusion PlGF was correlated with FHR monitoring findings and might be a promising biomarker of intrapartum fetal function.
Collapse
Affiliation(s)
- Hiroaki Tanaka
- Department of Obstetrics and Gynecology, Mie University, Tsu, Japan.
| | - Kayo Tanaka
- Department of Obstetrics and Gynecology, Mie University, Tsu, Japan
| | - Sho Takakura
- Department of Obstetrics and Gynecology, Mie University, Tsu, Japan
| | - Naosuke Enomoto
- Department of Obstetrics and Gynecology, Mie University, Tsu, Japan
| | - Shintaro Maki
- Department of Obstetrics and Gynecology, Mie University, Tsu, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University, Tsu, Japan
| |
Collapse
|
6
|
Ye X, Gaucher JF, Vidal M, Broussy S. A Structural Overview of Vascular Endothelial Growth Factors Pharmacological Ligands: From Macromolecules to Designed Peptidomimetics. Molecules 2021; 26:6759. [PMID: 34833851 PMCID: PMC8625919 DOI: 10.3390/molecules26226759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/27/2022] Open
Abstract
The vascular endothelial growth factor (VEGF) family of cytokines plays a key role in vasculogenesis, angiogenesis, and lymphangiogenesis. VEGF-A is the main member of this family, alongside placental growth factor (PlGF), VEGF-B/C/D in mammals, and VEGF-E/F in other organisms. To study the activities of these growth factors under physiological and pathological conditions, resulting in therapeutic applications in cancer and age-related macular degeneration, blocking ligands have been developed. These have mostly been large biomolecules like antibodies. Ligands with high affinities, at least in the nanomolar range, and accurate structural data from X-ray crystallography and NMR spectroscopy have been described. They constitute the main focus of this overview, which evidences similarities and differences in their binding modes. For VEGF-A ligands, and to a limited extent also for PlGF, a transition is now observed towards developing smaller ligands like nanobodies and peptides. These include unnatural amino acids and chemical modifications for designed and improved properties, such as serum stability and greater affinity. However, this review also highlights the scarcity of such small molecular entities and the striking lack of small organic molecule ligands. It also shows the gap between the rather large array of ligands targeting VEGF-A and the general absence of ligands binding other VEGF members, besides some antibodies. Future developments in these directions are expected in the upcoming years, and the study of these growth factors and their promising therapeutic applications will be welcomed.
Collapse
Affiliation(s)
- Xiaoqing Ye
- Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, 75006 Paris, France; (X.Y.); (M.V.)
| | - Jean-François Gaucher
- Laboratoire de Cristallographie et RMN Biologiques, Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, 75006 Paris, France;
| | - Michel Vidal
- Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, 75006 Paris, France; (X.Y.); (M.V.)
- Service Biologie du Médicament, Toxicologie, AP-HP, Hôpital Cochin, 75014 Paris, France
| | - Sylvain Broussy
- Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, 75006 Paris, France; (X.Y.); (M.V.)
| |
Collapse
|
7
|
Uemura A, Fruttiger M, D'Amore PA, De Falco S, Joussen AM, Sennlaub F, Brunck LR, Johnson KT, Lambrou GN, Rittenhouse KD, Langmann T. VEGFR1 signaling in retinal angiogenesis and microinflammation. Prog Retin Eye Res 2021; 84:100954. [PMID: 33640465 PMCID: PMC8385046 DOI: 10.1016/j.preteyeres.2021.100954] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
Five vascular endothelial growth factor receptor (VEGFR) ligands (VEGF-A, -B, -C, -D, and placental growth factor [PlGF]) constitute the VEGF family. VEGF-A binds VEGF receptors 1 and 2 (VEGFR1/2), whereas VEGF-B and PlGF only bind VEGFR1. Although much research has been conducted on VEGFR2 to elucidate its key role in retinal diseases, recent efforts have shown the importance and involvement of VEGFR1 and its family of ligands in angiogenesis, vascular permeability, and microinflammatory cascades within the retina. Expression of VEGFR1 depends on the microenvironment, is differentially regulated under hypoxic and inflammatory conditions, and it has been detected in retinal and choroidal endothelial cells, pericytes, retinal and choroidal mononuclear phagocytes (including microglia), Müller cells, photoreceptor cells, and the retinal pigment epithelium. Whilst the VEGF-A decoy function of VEGFR1 is well established, consequences of its direct signaling are less clear. VEGFR1 activation can affect vascular permeability and induce macrophage and microglia production of proinflammatory and proangiogenic mediators. However the ability of the VEGFR1 ligands (VEGF-A, PlGF, and VEGF-B) to compete against each other for receptor binding and to heterodimerize complicates our understanding of the relative contribution of VEGFR1 signaling alone toward the pathologic processes seen in diabetic retinopathy, retinal vascular occlusions, retinopathy of prematurity, and age-related macular degeneration. Clinically, anti-VEGF drugs have proven transformational in these pathologies and their impact on modulation of VEGFR1 signaling is still an opportunity-rich field for further research.
Collapse
Affiliation(s)
- Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, 20 Staniford Street, Boston, MA, 02114, USA.
| | - Sandro De Falco
- Angiogenesis Laboratory, Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", Via Pietro Castellino 111, 80131 Naples, Italy; ANBITION S.r.l., Via Manzoni 1, 80123, Naples, Italy.
| | - Antonia M Joussen
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200 Berlin, and Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
| | - Lynne R Brunck
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kristian T Johnson
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - George N Lambrou
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kay D Rittenhouse
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 9, 50931, Cologne, Germany.
| |
Collapse
|
8
|
Meakin AS, Cuffe JSM, Darby JRT, Morrison JL, Clifton VL. Let's Talk about Placental Sex, Baby: Understanding Mechanisms That Drive Female- and Male-Specific Fetal Growth and Developmental Outcomes. Int J Mol Sci 2021; 22:6386. [PMID: 34203717 PMCID: PMC8232290 DOI: 10.3390/ijms22126386] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 02/06/2023] Open
Abstract
It is well understood that sex differences exist between females and males even before they are born. These sex-dependent differences may contribute to altered growth and developmental outcomes for the fetus. Based on our initial observations in the human placenta, we hypothesised that the male prioritises growth pathways in order to maximise growth through to adulthood, thereby ensuring the greatest chance of reproductive success. However, this male-specific "evolutionary advantage" likely contributes to males being less adaptable to shifts in the in-utero environment, which then places them at a greater risk for intrauterine morbidities or mortality. Comparatively, females are more adaptable to changes in the in-utero environment at the cost of growth, which may reduce their risk of poor perinatal outcomes. The mechanisms that drive these sex-specific adaptations to a change in the in-utero environment remain unclear, but an increasing body of evidence within the field of developmental biology would suggest that alterations to placental function, as well as the feto-placental hormonal milieu, is an important contributing factor. Herein, we have addressed the current knowledge regarding sex-specific intrauterine growth differences and have examined how certain pregnancy complications may alter these female- and male-specific adaptations.
Collapse
Affiliation(s)
- Ashley S. Meakin
- Early Origins of Adult Health Research Group, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.S.M.); (J.R.T.D.); (J.L.M.)
| | - James S. M. Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.S.M.); (J.R.T.D.); (J.L.M.)
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.S.M.); (J.R.T.D.); (J.L.M.)
| | - Vicki L. Clifton
- Mater Medical Research Institute, The University of Queensland, Brisbane, QLD 4000, Australia
| |
Collapse
|
9
|
Ma Y, Wang W, Liu L, Liu Y, Bi W. Co-expression of VEGF-B and FLT-1 correlates with malignancy and prognosis of gastric cancer. Biomark Med 2021; 15:481-488. [PMID: 33856262 DOI: 10.2217/bmm-2020-0608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background: This study aims to investigate the correlation of VEGF-B and FLT-1 co-expression with the prognosis of gastric cancer (GC). Materials & methods: Primary GC samples and adjacent tissues were obtained from 96 patients. Results: Both VEGF-B and FLT-1 were testified to be upregulated in the human GC compared with adjacent tissues. Spearman's rank correlation analysis indicated that VEGF-B and FLT-1 expression were correlated (r = 0.321, p = 0.0015). High VEGF-B and FLT-1 co-expression patients showed poor prognosis when compared with low VEGF-B and FLT-1 co-expression patients (p = 0.0169). Conclusion: The high co-expression of VEGF-B and FLT-1 in GC shows a poor prognosis of overall survival, and targeted therapy against the interaction between VEGF-B and FLT-1 is worth further detailed analysis.
Collapse
Affiliation(s)
- Yanpeng Ma
- Department of General Surgery of East District, Second Hospital of Hebei Medical University, no. 215 Heping East Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
| | - Wenyao Wang
- Department of General Surgery of East District, Second Hospital of Hebei Medical University, no. 215 Heping East Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
| | - Longlong Liu
- Department of General Surgery of East District, Second Hospital of Hebei Medical University, no. 215 Heping East Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
| | - Yang Liu
- Department of Vascular Surgery, Second Hospital of Hebei Medical University, no. 215 Heping East Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
| | - Wei Bi
- Department of Vascular Surgery, Second Hospital of Hebei Medical University, no. 215 Heping East Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
| |
Collapse
|
10
|
Synergistic interactions of PlGF and VEGF contribute to blood-retinal barrier breakdown through canonical NFκB activation. Exp Cell Res 2020; 397:112347. [PMID: 33130176 DOI: 10.1016/j.yexcr.2020.112347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/26/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
Abstract
To investigate the role of placental growth factor/vascular endothelial growth factor (PlGF-VEGF) heterodimers are involved in the blood-retinal barrier (BRB) breakdown and the associated mechanism, human retinal endothelial cells (HRECs) were treated with recombinant human (rh)PlGF-VEGF heterodimers and rhPlGF and studied in normal and high-glucose conditions. HREC barrier function was evaluated by the measurement of trans-endothelial electrical resistance (TEER). Adeno-Associated Virus Type 5 (AAV5) vectors overexpressed PlGF in the retina by intravitreal injection into the C57BL6 mouse eye. AAV5-GFP vector and naïve animals were used as controls. Immunofluorescence (IF) and western blots examined the protein expression of PlGF-VEGF heterodimers, VEGF, PlGF, NFκB, p-IκBα, ZO-1, and VE-cadherin in HREC and mouse retina. PlGF-VEGF heterodimers were detected predominantly in the HREC cell nuclei based on IF and cytoplasmic and nuclear fractionation experiments. High glucose treatment increased PlGF-VEGF nuclear abundance. Dot immunoblotting demonstrated a strong affinity of the 5D11D4 antibody to PlGF-VEGF heterodimers. rhPlGF-VEGF disrupted the barrier function of HREC, which was prevented by the neutralization of PlGF-VEGF by the 5D11D4 antibody. Stimulation of HRECs with rhPlGF also led to an increase in the nuclear signals for PlGF-VEGF, p-IκBα, and colocalization of NFκB p65 and PlGF-VEGF in the nuclei. The selective IKK2 inhibitor IMD0354 disrupted the nuclear colocalization. Treatment with IMD0354 restored the barrier function of HREC, as indicated by the ZO-1 and VE-cadherin expression. In the mouse retinas, PlGF overexpression by AAV5 vector reduced ZO-1 expression and increased abundance of pIκBα. PIGF/VEGF heterodimers mediate BRB breakdown potentially through the canonical NFκB activation.
Collapse
|
11
|
Yang SH, Wang XL, Cai J, Wang SH. Diagnostic Value of Circulating PIGF in Combination with Flt-1 in Early Cervical Cancer. Curr Med Sci 2020; 40:973-978. [PMID: 33123910 DOI: 10.1007/s11596-020-2269-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/21/2020] [Indexed: 12/29/2022]
Abstract
The utility of placental growth factor (PlGF) and its receptor VEGFR-1 (Flt-1) as biomarkers for cervical cancer has not been clarified yet. To address this issue, we investigated the levels of soluble PlGF (sPlGF) and soluble Flt-1 (sFlt-1) in the serum from patients with early cervical cancer, cervical intraepithelial neoplasia (CIN) and controls in this study. sPlGF and sFlt-1 were detected in 44 preoperative patients with cervical cancer, 18 cases with CIN, and 20 controls by ELISA. It was found that both sPlGF and sFlt-1 were significantly increased in the cervical cancer group as compared with those in CIN and control groups. sPlGF presented a high diagnostic ability of cervical cancer, with a sensitivity of 61.36% and a specificity of 89.47%; and sFlt-1 with a sensitivity of 50.00% and a specificity of 92.11%. Importantly, the combined use of sPlGF and sFlt-1 could increase the diagnostic rate of cervical cancer, with a sensitivity of 70.45% and a specificity of 92.11%. These results indicated that both sPlGF and sFlt-1 in circulation can serve as possible valuable diagnostic biomarkers for cervical cancer, and the combined use of them can be more valuable to diagnose the patients with early cervical cancer.
Collapse
Affiliation(s)
- Shou-Hua Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao-Ling Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shao-Hai Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
12
|
Riccardi C, Napolitano E, Platella C, Musumeci D, Melone MAB, Montesarchio D. Anti-VEGF DNA-based aptamers in cancer therapeutics and diagnostics. Med Res Rev 2020; 41:464-506. [PMID: 33038031 DOI: 10.1002/med.21737] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/12/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
The vascular endothelial growth factor (VEGF) family and its receptors play fundamental roles not only in physiological but also in pathological angiogenesis, characteristic of cancer progression. Aiming at finding putative treatments for several malignancies, various small molecules, antibodies, or protein-based drugs have been evaluated in vitro and in vivo as VEGF inhibitors, providing efficient agents approved for clinical use. Due to the high clinical importance of VEGF, also a great number of anti-VEGF nucleic acid-based aptamers-that is, oligonucleotides able to bind with high affinity and specificity a selected biological target-have been developed as promising agents in anticancer strategies. Notable research efforts have been made in optimization processes of the identified aptamers, searching for increased target affinity and/or bioactivity by exploring structural analogues of the lead compounds. This review is focused on recent studies devoted to the development of DNA-based aptamers designed to target VEGF. Their therapeutic potential as well as their significance in the construction of highly selective biosensors is here discussed.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.,Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Inter-University Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ettore Napolitano
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Chiara Platella
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.,Institute of Biostructures and Bioimages, Naples, Italy
| | - Mariarosa A B Melone
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Inter-University Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
13
|
Mtshali Z, Moodley J, Naicker T. An Insight into the Angiogenic and Lymphatic Interplay in Pre-eclampsia Comorbid with HIV Infection. Curr Hypertens Rep 2020; 22:35. [PMID: 32200445 DOI: 10.1007/s11906-020-01040-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW To provide insight on the imbalance of angiogenic and lymphangiogenic factors in pre-eclampsia, as well as highlight polymorphism in genes related to angiogenesis and lymphangiogenesis. RECENT FINDINGS The pregnancy-specific disorder pre-eclampsia is diagnosed by the presence of hypertension with/without proteinuria, after 20 weeks of gestation. The pathogenesis of pre-eclampsia remains ambiguous, but research over the years has identified an imbalance in maternal and foetal factors. Familial predisposition and gene variation are also linked to pre-eclampsia development. The sFlt-1/PIGF ratio has attracted great attention over the years; more recently several researchers have reported that a sFlt-1/PIGF ratio of ≤ 38 can be used to predict short-term absence of pre-eclampsia. This ratio has the potential to prevent adverse pregnancy outcomes and reduce healthcare costs significantly. Genome-wide studies have additionally identified variation in the foetal gene near Flt-1. The development of preeclampsia is not limited to the maternal interface, but foetal involvement as well as genetic interplay is associated with the disorder.
Collapse
Affiliation(s)
- Zamahlabangane Mtshali
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.
| | - Jagidesa Moodley
- Department of Obstetrics and Gynaecology and Women's Health and HIV Research Group, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
14
|
Yang W, Jiang Y, Wang Y, Zhang T, Liu Q, Wang C, Swisher G, Wu N, Chao C, Prasadan K, Gittes GK, Xiao X. Placental growth factor in beta cells plays an essential role in gestational beta-cell growth. BMJ Open Diabetes Res Care 2020; 8:e000921. [PMID: 32144129 PMCID: PMC7059504 DOI: 10.1136/bmjdrc-2019-000921] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Pancreatic beta cells proliferate in response to metabolic requirements during pregnancy, while failure of this response may cause gestational diabetes. A member of the vascular endothelial growth factor family, placental growth factor (PlGF), typically plays a role in metabolic disorder and pathological circumstance. The expression and function of PlGF in the endocrine pancreas have not been reported and are addressed in the current study. RESEARCH DESIGN AND METHODS PlGF levels in beta cells were determined by immunostaining or ELISA in purified beta cells in non-pregnant and pregnant adult mice. An adeno-associated virus (AAV) serotype 8 carrying a shRNA for PlGF under the control of a rat insulin promoter (AAV-rat insulin promoter (RIP)-short hairpin small interfering RNA for PlGF (shPlGF)) was prepared and infused into mouse pancreas through the pancreatic duct to specifically knock down PlGF in beta cells, and its effects on beta-cell growth were determined by beta-cell proliferation, beta-cell mass and insulin release. A macrophage-depleting reagent, clodronate, was coapplied into AAV-treated mice to study crosstalk between beta cells and macrophages. RESULTS PlGF is exclusively produced by beta cells in the adult mouse pancreas. Moreover, PlGF expression in beta cells was significantly increased during pregnancy. Intraductal infusion of AAV-RIP-shPlGF specifically knocked down PlGF in beta cells, resulting in compromised beta-cell proliferation, reduced growth in beta-cell mass and impaired glucose tolerance during pregnancy. Mechanistically, PlGF depletion in beta cells reduced islet infiltration of trophic macrophages, which appeared to be essential for gestational beta-cell growth. CONCLUSIONS Our study suggests that increased expression of PlGF in beta cells may trigger gestational beta-cell growth through recruited macrophages.
Collapse
Affiliation(s)
- Weixia Yang
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yinan Jiang
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yan Wang
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ting Zhang
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Qun Liu
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Endocrinology, the First Affiliated Hospital of NanChang University, Nanchang, China
| | - Chaoban Wang
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pediatric Endocrinology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Grant Swisher
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nannan Wu
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Endocrinology, Lu He Hospital, Capital Medical University, Beijing, China
| | - Chelsea Chao
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Krishna Prasadan
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - George K Gittes
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Xiangwei Xiao
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
Hendrata M, Sudiono J. Multiscale modeling of tumor response to vascular endothelial growth factor (VEGF) inhibitor. In Silico Biol 2020; 14:71-88. [PMID: 35001886 PMCID: PMC8842763 DOI: 10.3233/isb-210235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Vascular endothelial growth factor (VEGF) has been known as a key mediator of angiogenesis in cancer. Bevacizumab is anti-VEGF monoclonal antibody that has been approved by the FDA as a first-line treatment in many types of cancer. In this paper, we extend a previously validated multiscale tumor model to comprehensively include the multiple roles of VEGF during the course of angiogenesis and its binding mechanism with bevacizumab. We use the model to simulate tumor system response under various bevacizumab concentrations, both in stand-alone treatment and in combination with chemotherapy. Our simulation indicates that periodic administration of bevacizumab with lower concentration can achieve greater efficacy than a single treatment with higher concentration. The simulation of the combined therapy also shows that the continuous administration of bevacizumab during the maintenance phase can lead to antitumor activity which further suppresses its growth. Agreement with experimental results indicates the potential of the model in predicting the efficacy of anti-VEGF therapies and could therefore contribute to developing prospective clinical trials.
Collapse
Affiliation(s)
- Melisa Hendrata
- Department of Mathematics, California State University, Los Angeles, CA, USA
| | - Janti Sudiono
- Department of Oral Pathology, Faculty of Dentistry, Trisakti University, Jakarta, Indonesia
| |
Collapse
|
16
|
Hendrata M, Sudiono J. A hybrid multiscale model for investigating tumor angiogenesis and its response to cell-based therapy. In Silico Biol 2019; 13:1-20. [PMID: 29226860 PMCID: PMC6597970 DOI: 10.3233/isb-170469] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Angiogenesis, a formation of blood vessels from an existing vasculature, plays a key role in tumor growth and its progression into cancer. The lining of blood vessels consists of endothelial cells (ECs) which proliferate and migrate, allowing the capillaries to sprout towards the tumor to deliver the needed oxygen. Various treatments aiming to suppress or even inhibit angiogenesis have been explored. Mesenchymal stem cells (MSCs) have recently been undergoing development in cell-based therapy for cancer due to their ability to migrate towards the capillaries and induce the apoptosis of the ECs, causing capillary degeneration. However, further investigations in this direction are needed as it is usually difficult to preclinically assess the efficacy of such therapy. We develop a hybrid multiscale model that integrates molecular, cellular, tissue and extracellular components of tumor system to investigate angiogenesis and tumor growth under MSC-mediated therapy. Our simulations produce angiogenesis and vascular tumor growth profiles as observed in the experiments. Furthermore, the simulations show that the effectiveness of MSCs in inducing EC apoptosis is density dependent and its full effect is reached within several days after MSCs application. Quantitative agreements with experimental data indicate the predictive potential of our model for evaluating the efficacy of cell-based therapies targeting angiogenesis.
Collapse
Affiliation(s)
- Melisa Hendrata
- Department of Mathematics, California StateUniversity, Los Angeles, CA, USA
| | - Janti Sudiono
- Department of Oral Pathology, Faculty of Dentistry, Trisakti University, Jakarta, Indonesia
| |
Collapse
|
17
|
Abstract
Receptor tyrosine kinases (RTKs) play important roles in cell growth, motility, differentiation, and survival. These single-pass membrane proteins are grouped into subfamilies based on the similarity of their extracellular domains. They are generally thought to be activated by ligand binding, which promotes homodimerization and then autophosphorylation in trans. However, RTK interactions are more complicated, as RTKs can interact in the absence of ligand and heterodimerize within and across subfamilies. Here, we review the known cross-subfamily RTK heterointeractions and their possible biological implications, as well as the methodologies which have been used to study them. Moreover, we demonstrate how thermodynamic models can be used to study RTKs and to explain many of the complicated biological effects which have been described in the literature. Finally, we discuss the concept of the RTK interactome: a putative, extensive network of interactions between the RTKs. This RTK interactome can produce unique signaling outputs; can amplify, inhibit, and modify signaling; and can allow for signaling backups. The existence of the RTK interactome could provide an explanation for the irreproducibility of experimental data from different studies and for the failure of some RTK inhibitors to produce the desired therapeutic effects. We argue that a deeper knowledge of RTK interactome thermodynamics can lead to a better understanding of fundamental RTK signaling processes in health and disease. We further argue that there is a need for quantitative, thermodynamic studies that probe the strengths of the interactions between RTKs and their ligands and between different RTKs.
Collapse
Affiliation(s)
- Michael D. Paul
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore MD 21218
| | - Kalina Hristova
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore MD 21218
| |
Collapse
|
18
|
The role of placental growth factor (PlGF) and its receptor system in retinal vascular diseases. Prog Retin Eye Res 2018; 69:116-136. [PMID: 30385175 DOI: 10.1016/j.preteyeres.2018.10.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/23/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022]
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family. Upon binding to VEGF- and neuropilin-receptor sub-types, PlGF modulates a range of neural, glial and vascular cell responses that are distinct from VEGF-A. As PlGF expression is selectively associated with pathological angiogenesis and inflammation, its blockade does not affect the healthy vasculature. PlGF actions have been extensively described in tumor biology but more recently there has been accumulating preclinical evidence that indicates that this growth factor could have an important role in retinal diseases. High levels of PlGF have been found in aqueous humor, vitreous and/or retina of patients exhibiting retinopathies, especially those with diabetic retinopathy (DR) and neovascular age-related macular degeneration (nvAMD). Expression of this growth factor seems to correlate closely with many of the key pathogenic features of early and late retinopathy in preclinical models. For example, studies using genetic modification and/or pharmacological treatment to block PlGF in the laser-induced choroidal neovascularization (CNV) model, oxygen-induced retinopathy model, as well as various murine diabetic models, have shown that PlGF deletion or inhibition can reduce neovascularization, retinal leakage, inflammation and gliosis, without affecting vascular development or inducing neuronal degeneration. Moreover, an inhibitory effect of PlGF blockade on retinal scarring in the mouse CNV model has also been recently demonstrated and was found to be unique for PlGF inhibition, as compared to various VEGF inhibition strategies. Together, these preclinical results suggest that anti-PlGF therapy might have advantages over anti-VEGF treatment, and that it may have clinical applications as a standalone treatment or in combination with anti-VEGF. Additional clinical studies are clearly needed to further elucidate the role of PlGF and its potential as a therapeutic target in ocular diseases.
Collapse
|
19
|
Karaman S, Leppänen VM, Alitalo K. Vascular endothelial growth factor signaling in development and disease. Development 2018; 145:145/14/dev151019. [DOI: 10.1242/dev.151019] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
ABSTRACT
Vascular endothelial growth factors (VEGFs) are best known for their involvement in orchestrating the development and maintenance of the blood and lymphatic vascular systems. VEGFs are secreted by a variety of cells and they bind to their cognate tyrosine kinase VEGF receptors (VEGFRs) in endothelial cells to elicit various downstream effects. In recent years, there has been tremendous progress in elucidating different VEGF/VEGFR signaling functions in both the blood and lymphatic vascular systems. Here, and in the accompanying poster, we present key elements of the VEGF/VEGFR pathway and highlight the classical and newly discovered functions of VEGF signaling in blood and lymphatic vessel development and pathology.
Collapse
Affiliation(s)
- Sinem Karaman
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki 00290, Finland
| | - Veli-Matti Leppänen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki 00290, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki 00290, Finland
| |
Collapse
|
20
|
Apicella I, Cicatiello V, Acampora D, Tarallo V, De Falco S. Full Functional Knockout of Placental Growth Factor by Knockin with an Inactive Variant Able to Heterodimerize with VEGF-A. Cell Rep 2018; 23:3635-3646. [DOI: 10.1016/j.celrep.2018.05.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/17/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022] Open
|
21
|
Placental growth factor: A review of literature and future applications. Pregnancy Hypertens 2018; 14:260-264. [PMID: 29555222 DOI: 10.1016/j.preghy.2018.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 12/11/2022]
|
22
|
Di Pietro M, Pascuali N, Parborell F, Abramovich D. Ovarian angiogenesis in polycystic ovary syndrome. Reproduction 2018; 155:R199-R209. [PMID: 29386378 DOI: 10.1530/rep-17-0597] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/30/2018] [Indexed: 12/16/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent endocrine pathology among women in reproductive age. Its main symptoms are oligo or amenorrhea, hyperandrogenism and the presence of ovarian cysts. It is also associated with infertility, obesity and insulin resistance. Mainly due to its heterogeneity, PCOS treatments are directed to manage its symptoms and to prevent associated diseases. The correct formation and regression of blood vessels during each ovarian cycle is indispensable for proper follicular development, ovulation and corpus luteum formation. The importance of these processes opened a new and promising field: ovarian angiogenesis. Vascular alterations characterize numerous pathologies, either with increased, decreased or abnormal angiogenesis. In the last years, several anomalies of ovarian angiogenesis have been described in women with PCOS. Therefore, it has been suggested that these alterations may be associated with the decreased - or lack of - ovulation rates and for the formation of cysts in the PCOS ovaries. Restoration of a proper vessel formation in the ovaries may lead to improved follicular development and ovulation in these patients. In the present review, we attempt to summarize the alterations in ovarian angiogenesis that have been described in women with PCOS. We also discuss the therapeutic approaches aimed to correct these alterations and their beneficial effects on the treatment of infertility in PCOS.
Collapse
Affiliation(s)
- Mariana Di Pietro
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| |
Collapse
|
23
|
Foxp3 + Tregs are recruited to the retina to repair pathological angiogenesis. Nat Commun 2017; 8:748. [PMID: 28963474 PMCID: PMC5622066 DOI: 10.1038/s41467-017-00751-w] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 07/25/2017] [Indexed: 02/03/2023] Open
Abstract
Neovascular retinopathies are major causes of vision loss; yet treatments to prevent the condition are inadequate. The role of regulatory T cells in neovascular retinopathy is unknown. Here we show that in retinopathy regulatory T cells are transiently increased in lymphoid organs and the retina, but decline when neovascularization is established. The decline is prevented following regulatory T cells expansion with an IL-2/anti-IL-2 mAb complex or the adoptive transfer of regulatory T cells. Further, both approaches reduce vasculopathy (vaso-obliteration, neovascularization, vascular leakage) and alter the activation of Tmem119+ retinal microglia. Our in vitro studies complement these findings, showing that retinal microglia co-cultured with regulatory T cells exhibit a reduction in co-stimulatory molecules and pro-inflammatory mediators that is attenuated by CTLA-4 blockade. Collectively, we demonstrate that regulatory T cells are recruited to the retina and, when expanded in number, repair the vasculature. Manipulation of regulatory T cell numbers is a previously unrecognized, and promising avenue for therapies to prevent blinding neovascular retinopathies. The local immune responses in the eye are attenuated to preserve sight. Surprisingly, Deliyanti et al. show that regulatory T cells (Tregs) take an active role in protecting the eye from neovascularization in oxygen-induced retinopathy, and that interventions that augment the retinal Treg numbers reduce neovascular retinopathy in mice.
Collapse
|
24
|
Zimna A, Wiernicki B, Kolanowski T, Rozwadowska N, Malcher A, Labedz W, Trzeciak T, Chojnacka K, Bednarek-Rajewska K, Majewski P, Kurpisz M. Biological and Pro-Angiogenic Properties of Genetically Modified Human Primary Myoblasts Overexpressing Placental Growth Factor in In Vitro and In Vivo Studies. Arch Immunol Ther Exp (Warsz) 2017; 66:145-159. [PMID: 28951939 PMCID: PMC5851700 DOI: 10.1007/s00005-017-0486-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 07/11/2017] [Indexed: 01/01/2023]
Abstract
Cardiovascular diseases are a growing problem in developing countries; therefore, there is an ongoing intensive search for new approaches to treat these disorders. Currently, cellular therapies are focused on healing the damaged heart by implanting stem cells modified with pro-angiogenic factors. This approach ensures that the introduced cells are capable of fulfilling the complex requirements of the environment, including the replacement of the post-infarction scar with cells that are able to contract and promote the formation of new blood vessels that can supply the ischaemic region with nutrients and oxygen. This study focused on the genetic modification of human skeletal muscle cells (SkMCs). We chose myoblast cells due to their close biological resemblance to cardiomyocytes and the placental growth factor (PlGF) gene due to its pro-angiogenic potential. In our in vitro studies, we transfected SkMCs with the PlGF gene using electroporation, which has previously been proven to be efficient and generate robust overexpression of the PlGF gene and elevate PlGF protein secretion. Moreover, the functionality of the secreted pro-angiogenic proteins was confirmed using an in vitro capillary development assay. We have also examined the influence of PlGF overexpression on VEGF-A and VEGF-B, which are well-known factors described in the literature as the most potent activators of blood vessel formation. We were able to confirm the overexpression of VEGF-A in myoblasts transfected with the PlGF gene. The results obtained in this study were further verified in an animal model. These data were able to confirm the potential therapeutic effects of the applied treatments.
Collapse
Affiliation(s)
- Agnieszka Zimna
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - Bartosz Wiernicki
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - Tomasz Kolanowski
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - Natalia Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - Agnieszka Malcher
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - Wojciech Labedz
- Department of Orthopaedics and Traumatology, W. Dega University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Tomasz Trzeciak
- Department of Orthopaedics and Traumatology, W. Dega University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Chojnacka
- Department of Clinical Pathomorphology, H. Swiecicki University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Bednarek-Rajewska
- Department of Clinical Pathomorphology, H. Swiecicki University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Przemyslaw Majewski
- Department of Clinical Pathomorphology, H. Swiecicki University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Kurpisz
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland.
| |
Collapse
|
25
|
Atzori MG, Tentori L, Ruffini F, Ceci C, Lisi L, Bonanno E, Scimeca M, Eskilsson E, Daubon T, Miletic H, Ricci Vitiani L, Pallini R, Navarra P, Bjerkvig R, D'Atri S, Lacal PM, Graziani G. The anti-vascular endothelial growth factor receptor-1 monoclonal antibody D16F7 inhibits invasiveness of human glioblastoma and glioblastoma stem cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:106. [PMID: 28797294 PMCID: PMC5553938 DOI: 10.1186/s13046-017-0577-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/02/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Glioblastoma (GBM) is a highly migratory, invasive, and angiogenic brain tumor. Like vascular endothelial growth factor-A (VEGF-A), placental growth factor (PlGF) promotes GBM angiogenesis. VEGF-A is a ligand for both VEGF receptor-1 (VEGFR-1) and VEGFR-2, while PlGF interacts exclusively with VEGFR-1. We recently generated the novel anti-VEGFR-1 monoclonal antibody (mAb) D16F7 that diminishes VEGFR-1 homodimerization/activation without affecting VEGF-A and PlGF binding. METHODS In the present study, we evaluated the expression of VEGFR-1 in human GBM tissue samples (n = 42) by immunohistochemistry, in cell lines (n = 6) and GBM stem cells (GSCs) (n = 18) by qRT-PCR and/or western blot analysis. In VEGFR-1 positive GBM or GSCs we also analyzed the ability of D16F7 to inhibit GBM invasiveness in response to VEGF-A and PlGF. RESULTS Most of GBM specimens stained positively for VEGFR-1 and all but one GBM cell lines expressed VEGFR-1. On the other hand, in GSCs the expression of the receptor was heterogeneous. D16F7 reduced migration and invasion of VEGFR-1 positive GBM cell lines and patient-derived GSCs in response to VEGF-A and PlGF. Interestingly, this effect was also observed in VEGFR-1 positive GSCs transfected to over-express wild-type EGFR (EGFRwt+) or mutant EGFR (ligand binding domain-deficient EGFRvIII+). Furthermore, D16F7 suppressed intracellular signal transduction in VEGFR-1 over-expressing GBM cells by reducing receptor auto-phosphorylation at tyrosine 1213 and downstream Erk1/2 activation induced by receptor ligands. CONCLUSION The results from this study suggest that VEGFR-1 is a relevant target for GBM therapy and that D16F7-derived humanized mAbs warrant further investigation.
Collapse
Affiliation(s)
- Maria Grazia Atzori
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Lucio Tentori
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Federica Ruffini
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-IRCCS, Via dei Monti di Creta, 104, 00167, Rome, Italy
| | - Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Lucia Lisi
- Istituto di Farmacologia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Roma, Italia
| | - Elena Bonanno
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Manuel Scimeca
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Eskil Eskilsson
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas Daubon
- INSERM U1029, University of Bordeaux, Pessac, France.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Lucia Ricci Vitiani
- Department of Hematology, Oncology and Molecular Medicine, "Istituto Superiore di Sanità" (ISS), Rome, Italy
| | - Roberto Pallini
- Department of Neurosurgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Pierluigi Navarra
- Istituto di Farmacologia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Roma, Italia.,UOC di Farmacologia, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168, Roma, Italia
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Stefania D'Atri
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-IRCCS, Via dei Monti di Creta, 104, 00167, Rome, Italy
| | - Pedro Miguel Lacal
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-IRCCS, Via dei Monti di Creta, 104, 00167, Rome, Italy.
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
26
|
Kay VR, Tayade C, Carmeliet P, Croy BA. Influences of placental growth factor on mouse retinal vascular development. Dev Dyn 2017. [PMID: 28646507 DOI: 10.1002/dvdy.24540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Placental growth factor (PGF) is important for wound-healing and vascular collaterogenesis. PGF deficiency is associated with preeclampsia, a hypertensive disease of human pregnancy. Offspring born to preeclamptic mothers display cognitive impairments and brain vascular and neurostructural deviations. Low PGF production during development may contribute to alterations in offspring cerebrovascular beds. Retina is a readily accessible part of the central nervous system with a well-described pattern of vascular development in mice. Impacts of PGF deficiency were addressed during mouse retinal vascularization. RESULTS Retinal vessels were compared between Pgf-/- and congenic C57BL/6 (B6) mice. PGF deficiency altered neonatal retinal vascularization patterns. Some anatomic alterations persisted into adulthood, particularly in males. Greater arterial wall collagen IV expression was found in adult Pgf-/- females. Pregnancy (studied in adult females at gestational days 11.5 or 18.5) induced subtle changes upon the mother's retinal vasculature but these pregnancy-induced changes did not differ between genotypes. Significant sex-related differences occurred between adult male and female B6 although sexually dimorphic retinal vascular differences were absent in B6 neonates. CONCLUSIONS Overall, PGF has a role in retinal vascular angiogenesis and vessel organization during development but does not affect retinal vessel adaptations in adult females during pregnancy. Developmental Dynamics 246:700-712, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Chandrakant Tayade
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB - Vesalius Research Center, University of Leuven, Department of Oncology, Leuven, Belgium
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
27
|
Clegg LE, Mac Gabhann F. A computational analysis of in vivo VEGFR activation by multiple co-expressed ligands. PLoS Comput Biol 2017; 13:e1005445. [PMID: 28319199 PMCID: PMC5378411 DOI: 10.1371/journal.pcbi.1005445] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 04/03/2017] [Accepted: 03/08/2017] [Indexed: 12/16/2022] Open
Abstract
The splice isoforms of vascular endothelial growth A (VEGF) each have different affinities for the extracellular matrix (ECM) and the coreceptor NRP1, which leads to distinct vascular phenotypes in model systems expressing only a single VEGF isoform. ECM-immobilized VEGF can bind to and activate VEGF receptor 2 (VEGFR2) directly, with a different pattern of site-specific phosphorylation than diffusible VEGF. To date, the way in which ECM binding alters the distribution of isoforms of VEGF and of the related placental growth factor (PlGF) in the body and resulting angiogenic signaling is not well-understood. Here, we extend our previous validated cell-level computational model of VEGFR2 ligation, intracellular trafficking, and site-specific phosphorylation, which captured differences in signaling by soluble and immobilized VEGF, to a multi-scale whole-body framework. This computational systems pharmacology model captures the ability of the ECM to regulate isoform-specific growth factor distribution distinctly for VEGF and PlGF, and to buffer free VEGF and PlGF levels in tissue. We show that binding of immobilized growth factor to VEGF receptors, both on endothelial cells and soluble VEGFR1, is likely important to signaling in vivo. Additionally, our model predicts that VEGF isoform-specific properties lead to distinct profiles of VEGFR1 and VEGFR2 binding and VEGFR2 site-specific phosphorylation in vivo, mediated by Neuropilin-1. These predicted signaling changes mirror those observed in murine systems expressing single VEGF isoforms. Simulations predict that, contrary to the 'ligand-shifting hypothesis,' VEGF and PlGF do not compete for receptor binding at physiological concentrations, though PlGF is predicted to slightly increase VEGFR2 phosphorylation when over-expressed by 10-fold. These results are critical to design of appropriate therapeutic strategies to control VEGF availability and signaling in regenerative medicine applications.
Collapse
Affiliation(s)
- Lindsay E. Clegg
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Feilim Mac Gabhann
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
28
|
Newell LF, Holtan SG. Placental growth factor: What hematologists need to know. Blood Rev 2017; 31:57-62. [PMID: 27608972 PMCID: PMC5916812 DOI: 10.1016/j.blre.2016.08.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/19/2016] [Accepted: 08/23/2016] [Indexed: 12/16/2022]
Abstract
Although first identified in placenta, the angiogenic factor known as placental growth factor (PlGF) can be widely expressed in ischemic or damaged tissues. Recent studies have indicated that PlGF is a relevant factor in the pathobiology of blood diseases including hemoglobinopathies and hematologic malignancies. Therapies for such blood diseases may one day be based upon these and ongoing investigations into the role of PlGF in sickle cell disease, acute and chronic leukemias, and complications related to hematopoietic cell transplantation. In this review, we summarize recent studies regarding the potential role of PlGF in blood disorders and suggest avenues for future research.
Collapse
Affiliation(s)
- Laura F Newell
- Oregon Health and Science University, Center for Hematologic Malignancies, Portland, OR, USA.
| | - Shernan G Holtan
- University of Minnesota, Blood and Marrow Transplant Program, Minneapolis, MN, USA.
| |
Collapse
|
29
|
|
30
|
Expression and Function of Placenta Growth Factor: Implications for Abnormal Placentation. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/s1071-55760300048-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Vilsmaier T, Rack B, Janni W, Jeschke U, Weissenbacher T. Angiogenic cytokines and their influence on circulating tumour cells in sera of patients with the primary diagnosis of breast cancer before treatment. BMC Cancer 2016; 16:547. [PMID: 27464822 PMCID: PMC4964055 DOI: 10.1186/s12885-016-2612-7] [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: 11/02/2015] [Accepted: 07/25/2016] [Indexed: 12/05/2022] Open
Abstract
Background Circulating tumour cells (CTCs) have been found to be a prognostic marker for reduced disease free survival, breast cancer–specific survival, and overall survival before the start of systemic treatment. Methods A total of 200 patients’ sera were included in this study, 100 patients being CTC positive and 100 patients being CTC negative. Matching criteria were histo-pathological grading, lymph node metastasis, hormone receptor status, TNM classification and survived breast cancer patients vs. deceased tumor associated patients. A multi cytokine/chemokine array was used to screen the sera for the angiogenic markers. Results Statistical significant correlation was exposed for sFlt1 values in regard to the CTC-Status. CTC negative patients displayed increased sFlt1 expression opposed to CTC positive breast cancer patients. Furthermore, significant enhanced PIGF values were also disclosed in CTC negative patients compared to patients being CTC positive. Analyzing the living patient collective we found significant differences in sFlt1 and PlGF values in regard to CTC negative and CTC positive patients. Conclusion Both vascular markers showed enhanced expression in the CTC negative patient collective. To continue, the collective graded G2 showed significantly enhanced sFlt1 expressions amongst patients with no CTCs. Moreover, the patient collective with no lymph node metastasis and CTC negativity indicated statistically significant increased sFlt1 values. A functional interaction of sFlt1 and PlGF was found, suggesting that their overexpression in tumour cells inhibits CTCs entering the peripheral blood. Furthermore, in regard to CTC negativity, sFlt1 and PlGF values may potentially serve as predictive markers. Trial registration The TRN of this study is NCT02181101 and the date of registration was the 4th of June 2014. The study was retrospectively registered.
Collapse
Affiliation(s)
- Theresa Vilsmaier
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Maistrasse 11, 80337, Munich, Germany
| | - Brigitte Rack
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Maistrasse 11, 80337, Munich, Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Maistrasse 11, 80337, Munich, Germany.
| | - Tobias Weissenbacher
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Maistrasse 11, 80337, Munich, Germany
| | | |
Collapse
|
32
|
Makris A, Yeung KR, Lim SM, Sunderland N, Heffernan S, Thompson JF, Iliopoulos J, Killingsworth MC, Yong J, Xu B, Ogle RF, Thadhani R, Karumanchi SA, Hennessy A. Placental Growth Factor Reduces Blood Pressure in a Uteroplacental Ischemia Model of Preeclampsia in Nonhuman Primates. Hypertension 2016; 67:1263-72. [PMID: 27091894 DOI: 10.1161/hypertensionaha.116.07286] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/04/2016] [Indexed: 01/04/2023]
Abstract
An imbalance in the angiogenesis axis during pregnancy manifests as clinical preeclampsia because of endothelial dysfunction. Circulating soluble fms-like tyrosine kinase 1 (sFLT-1) increases and placental growth factor (PlGF) reduces before and during disease. We investigated the clinical and biochemical effects of replenishing the reduced circulating PlGF with recombinant human PlGF (rhPlGF) and thus restoring the angiogenic balance. Hypertensive proteinuria was induced in a nonhuman primate (Papio hamadryas) by uterine artery ligation at 136 days gestation (of a 182-day pregnancy). Two weeks after uteroplacental ischemia, rhPlGF (rhPlGF, n=3) or normal saline (control, n=4) was administered by subcutaneous injection (100 μg/kg per day) for 5 days. Blood pressure was monitored by intra-arterial radiotelemetry and sFLT-1 and PlGF by ELISA. Uteroplacental ischemia resulted in experimental preeclampsia evidenced by increased blood pressure, proteinuria, and endotheliosis on renal biopsy and elevated sFLT-1. PlGF significantly reduced after uteroplacental ischemia. rhPlGF reduced systolic blood pressure in the treated group (-5.2±0.8 mm Hg; from 132.6±6.6 mm Hg to 124.1±7.6 mm Hg) compared with an increase in systolic blood pressure in controls (6.5±3 mm Hg; from 131.3±1.5 mm Hg to 138.6±1.5 mm Hg). Proteinuria reduced in the treated group (-72.7±55.7 mg/mmol) but increased in the control group. Circulating levels of total sFLT-1 were not affected by the administration of PlGF; however, a reduction in placental sFLT-1 mRNA expression was demonstrated. There was no significant difference between the weights or lengths of the neonates in the rhPlGF or control group; however, this study was not designed to assess fetal safety or outcomes. Increasing circulating PlGF by the administration of rhPlGF improves clinical parameters in a primate animal model of experimental preeclampsia.
Collapse
Affiliation(s)
- Angela Makris
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.).
| | - Kristen R Yeung
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Shirlene M Lim
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Neroli Sunderland
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Scott Heffernan
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - John F Thompson
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Jim Iliopoulos
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Murray C Killingsworth
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Jim Yong
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Bei Xu
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Robert F Ogle
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Ravi Thadhani
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - S Ananth Karumanchi
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Annemarie Hennessy
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| |
Collapse
|
33
|
Talia DM, Deliyanti D, Agrotis A, Wilkinson-Berka JL. Inhibition of the Nuclear Receptor RORγ and Interleukin-17A Suppresses Neovascular Retinopathy: Involvement of Immunocompetent Microglia. Arterioscler Thromb Vasc Biol 2016; 36:1186-96. [PMID: 27055905 DOI: 10.1161/atvbaha.115.307080] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/28/2016] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Although inhibitors of vascular endothelial growth factor (VEGF) provide benefit for the management of neovascular retinopathies, their use is limited to end-stage disease and some eyes are resistant. We hypothesized that retinoic acid-related orphan nuclear receptor γ (RORγ) and its downstream effector, interleukin (IL)-17A, upregulate VEGF and hence are important treatment targets for neovascular retinopathies. APPROACH AND RESULTS Utilizing a model of oxygen-induced retinopathy, confocal microscopy and flow cytometry, we identified that retinal immunocompetent cells, microglia, express IL-17A. This was confirmed in primary cultures of rat retinal microglia, where hypoxia increased IL-17A protein as well as IL-17A, RORγ, and tumor necrosis factor-α mRNA, which were reduced by the RORγ inhibitor, digoxin, and the RORα/RORγ inverse agonist, SR1001. By contrast, retinal macroglial Müller cells and ganglion cells, key sources of VEGF in oxygen-induced retinopathy, did not produce IL-17A when exposed to hypoxia and IL-1β. However, they expressed IL-17 receptors, and in response to IL-17A, secreted VEGF. This suggested that RORγ and IL-17A inhibition might attenuate neovascular retinopathy. Indeed, digoxin and SR1001 reduced retinal vaso-obliteration, neovascularization, and vascular leakage as well as VEGF and VEGF-related placental growth factor. Digoxin and SR1001 reduced microglial-derived IL-17A and Müller cell and ganglion cell damage. The importance of IL-17A in oxygen-induced retinopathy was confirmed by IL-17A neutralization reducing vasculopathy, VEGF, placental growth factor, tumor necrosis factor-α, microglial density and Müller cell, and ganglion cell injury. CONCLUSIONS Our findings indicate that an RORγ/IL-17A axis influences VEGF production and neovascular retinopathy by mechanisms involving neuroglia. Inhibition of RORγ and IL-17A may have potential for the improved treatment of neovascular retinopathies.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antibodies, Monoclonal/pharmacology
- Cells, Cultured
- Digoxin/pharmacology
- Disease Models, Animal
- Ependymoglial Cells/drug effects
- Ependymoglial Cells/immunology
- Ependymoglial Cells/metabolism
- Hyperoxia/complications
- Interleukin-17/antagonists & inhibitors
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Mice, Inbred C57BL
- Microglia/drug effects
- Microglia/immunology
- Microglia/metabolism
- Microglia/pathology
- Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Placenta Growth Factor/metabolism
- Rats, Sprague-Dawley
- Retina/drug effects
- Retina/immunology
- Retina/metabolism
- Retina/pathology
- Retinal Ganglion Cells/drug effects
- Retinal Ganglion Cells/immunology
- Retinal Ganglion Cells/metabolism
- Retinal Neovascularization/immunology
- Retinal Neovascularization/metabolism
- Retinal Neovascularization/pathology
- Retinal Neovascularization/prevention & control
- Retinopathy of Prematurity/immunology
- Retinopathy of Prematurity/metabolism
- Retinopathy of Prematurity/pathology
- Retinopathy of Prematurity/prevention & control
- Signal Transduction/drug effects
- Sulfonamides/pharmacology
- Thiazoles/pharmacology
- Tumor Necrosis Factor-alpha/metabolism
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Dean M Talia
- From the Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | - Devy Deliyanti
- From the Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | - Alex Agrotis
- From the Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|
34
|
Szabo E, Schneider H, Seystahl K, Rushing EJ, Herting F, Weidner KM, Weller M. Autocrine VEGFR1 and VEGFR2 signaling promotes survival in human glioblastoma models in vitro and in vivo. Neuro Oncol 2016; 18:1242-52. [PMID: 27009237 PMCID: PMC4998998 DOI: 10.1093/neuonc/now043] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 02/18/2016] [Indexed: 12/28/2022] Open
Abstract
Background Although the vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) system has become a prime target for antiangiogenic treatment, its biological role in glioblastoma beyond angiogenesis has remained controversial. Methods Using neutralizing antibodies to VEGF or placental growth factor (PlGF) or the tyrosine kinase inhibitor, cediranib, or lentiviral gene silencing, we delineated autocrine signaling in glioma cell lines. The in vivo effects of VEGFR1 and VEGFR2 depletion were evaluated in orthotopic glioma xenograft models. Results VEGFR1 and VEGFR2 modulated glioma cell clonogenicity, viability, and invasiveness in vitro in an autocrine, cell–line-specific manner. VEGFR1 silencing promoted mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling, whereas VEGFR2 silencing resulted in cell-type dependent activation of the protein kinase B (PKB)/AKT and MAPK/ERK pathways. These responses may represent specific escape mechanisms from VEGFR inhibition. The survival of orthotopic glioma-bearing mice was prolonged upon VEGFR1 silencing in the LNT-229, LN-308, and U87MG models and upon VEGFR2 silencing in LN-308 and U87MG. Disruption of VEGFR1 and VEGFR2 signaling was associated with decreased tumor size, increased tumor necrosis, or loss of matrix metalloproteinase 9 (MMP9) immunoreactivity. Neutralizing VEGF and PlGF by specific antibodies was superior to either antibody treatment alone in the VEGFR1-dependent LNT-229 model. Conclusions Differential dependence on autocrine signaling through VEGFR1 and VEGFR2 suggests a need for biomarker–stratified VEGF(R)-based therapeutic approaches to glioblastoma.
Collapse
Affiliation(s)
- Emese Szabo
- Laboratory of Molecular Neuro-Oncology, Department of Neurology (E.S., H.S., K.S., M.W.), and Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.J.R); Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Nonnenwald 2, Penzberg D-82372, Germany (F.H., K.M.W.)
| | - Hannah Schneider
- Laboratory of Molecular Neuro-Oncology, Department of Neurology (E.S., H.S., K.S., M.W.), and Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.J.R); Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Nonnenwald 2, Penzberg D-82372, Germany (F.H., K.M.W.)
| | - Katharina Seystahl
- Laboratory of Molecular Neuro-Oncology, Department of Neurology (E.S., H.S., K.S., M.W.), and Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.J.R); Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Nonnenwald 2, Penzberg D-82372, Germany (F.H., K.M.W.)
| | - Elisabeth Jane Rushing
- Laboratory of Molecular Neuro-Oncology, Department of Neurology (E.S., H.S., K.S., M.W.), and Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.J.R); Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Nonnenwald 2, Penzberg D-82372, Germany (F.H., K.M.W.)
| | - Frank Herting
- Laboratory of Molecular Neuro-Oncology, Department of Neurology (E.S., H.S., K.S., M.W.), and Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.J.R); Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Nonnenwald 2, Penzberg D-82372, Germany (F.H., K.M.W.)
| | - K Michael Weidner
- Laboratory of Molecular Neuro-Oncology, Department of Neurology (E.S., H.S., K.S., M.W.), and Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.J.R); Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Nonnenwald 2, Penzberg D-82372, Germany (F.H., K.M.W.)
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology (E.S., H.S., K.S., M.W.), and Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.J.R); Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Nonnenwald 2, Penzberg D-82372, Germany (F.H., K.M.W.)
| |
Collapse
|
35
|
Drolet DW, Green LS, Gold L, Janjic N. Fit for the Eye: Aptamers in Ocular Disorders. Nucleic Acid Ther 2016; 26:127-46. [PMID: 26757406 PMCID: PMC4900223 DOI: 10.1089/nat.2015.0573] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
For any new class of therapeutics, there are certain types of indications that represent a natural fit. For nucleic acid ligands in general, and aptamers in particular, the eye has historically been an attractive site for therapeutic intervention. In this review, we recount the discovery and early development of three aptamers designated for use in ophthalmology, one approved (Macugen), and two in late-stage development (Fovista and Zimura). Every one of these molecules was originally intended for other indications. Key improvements in technology, specifically with regard to libraries used for in vitro selection and subsequent chemical optimization of aptamers, have played an important role in allowing the identification of development candidates with suitable properties. The lessons learned from the selection of these molecules are valuable for informing us about the many remaining opportunities for aptamer-based therapeutics in ophthalmology as well as for identifying additional indications for which aptamers as a class of therapeutics have distinct advantages.
Collapse
|
36
|
Iwamoto H, Zhang Y, Seki T, Yang Y, Nakamura M, Wang J, Yang X, Torimura T, Cao Y. PlGF-induced VEGFR1-dependent vascular remodeling determines opposing antitumor effects and drug resistance to Dll4-Notch inhibitors. SCIENCE ADVANCES 2015; 1:e1400244. [PMID: 26601163 PMCID: PMC4640632 DOI: 10.1126/sciadv.1400244] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/11/2015] [Indexed: 06/05/2023]
Abstract
Inhibition of Dll4 (delta-like ligand 4)-Notch signaling-mediated tumor angiogenesis is an attractive approach in cancer therapy. However, inhibition of Dll4-Notch signaling has produced different effects in various tumors, and no biomarkers are available for predicting the anti-Dll4-Notch-associated antitumor activity. We show that human and mouse tumor cell-derived placental growth factor (PlGF) is a key determinant of the Dll4-Notch-induced vascular remodeling and tumor growth. In natural PlGF-expressing human tumors, inhibition of Dll4-Notch signaling markedly accelerated tumor growth by increasing blood perfusion in nonleaking tumor vasculatures. Conversely, in PlGF-negative tumors, Dll4 inhibition suppressed tumor growth by the formation of nonproductive and leaky vessels. Surprisingly, genetic inactivation of vascular endothelial growth factor receptor 1 (VEGFR1) completely abrogated the PlGF-modulated vascular remodeling and tumor growth, indicating a crucial role for VEGFR1-mediated signals in modulating Dll4-Notch functions. These findings provide mechanistic insights on PlGF-VEGFR1 signaling in the modulation of the Dll4-Notch pathway in angiogenesis and tumor growth, and have therapeutic implications of PlGF as a biomarker for predicting the antitumor benefits of Dll4 and Notch inhibitors.
Collapse
Affiliation(s)
- Hideki Iwamoto
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Yin Zhang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Takahiro Seki
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Yunlong Yang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Masaki Nakamura
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Jian Wang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Xiaojuan Yang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
- Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology, Tongji University, Shanghai, People’s Republic of China
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 831 0011 Kurume, Japan
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
- Department of Medicine and Health Sciences, Linköping University, 581 83 Linköping, Sweden
- Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| |
Collapse
|
37
|
Xiang L, Varshney R, Rashdan NA, Shaw JH, Lloyd PG. Placenta growth factor and vascular endothelial growth factor a have differential, cell-type specific patterns of expression in vascular cells. Microcirculation 2015; 21:368-79. [PMID: 24410720 DOI: 10.1111/micc.12113] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 01/07/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE PLGF, a VEGF-A related protein, mediates collateral enlargement via monocytes but plays little role in capillary proliferation. In contrast, VEGF-A mediates both collateral enlargement and capillary proliferation. PLGF has been less thoroughly studied than VEGF-A, and questions remain regarding its regulation and function. Therefore, our goal was to characterize the expression of PLGF by vascular cells. We hypothesized that vascular SMC would express more PLGF than EC, since VEGF-A is primarily expressed by non-EC. METHODS We compared PLGF and VEGF-A across eight EC and SMC lines, then knocked down PLGF and evaluated cell function. We also assessed the effect of hypoxia on PLGF expression and promoter activity. RESULTS PLGF was most highly expressed in EC, whereas VEGF-A was most highly expressed in SMC. PLGF knockdown did not affect EC number, migration, or tube formation, but reduced monocyte migration toward EC. Monocyte migration was rescued by exogenous PLGF. Hypoxia increased PLGF protein without activating PLGF gene transcription. CONCLUSIONS PLGF and VEGF-A have distinct patterns of expression in vascular cells. EC derived PLGF may function primarily in communication between EC and circulating cells. Hypoxia increases EC PLGF expression posttranscriptionally.
Collapse
Affiliation(s)
- Lingjin Xiang
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma, USA
| | | | | | | | | |
Collapse
|
38
|
Huang H, He J, Johnson D, Wei Y, Liu Y, Wang S, Lutty GA, Duh EJ, Semba RD. Deletion of placental growth factor prevents diabetic retinopathy and is associated with Akt activation and HIF1α-VEGF pathway inhibition. Diabetes 2015; 64:200-12. [PMID: 25187372 PMCID: PMC4274802 DOI: 10.2337/db14-0016] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A new diabetic mouse strain, the Akita.PlGF knockout ((-/-)), was generated to study the role of placental growth factor (PlGF) in the pathogenesis of diabetic retinopathy (DR). PlGF deletion did not affect blood glucose but reduced the body weight of Akita.PlGF(-/-) mice. Diabetes-induced retinal cell death, capillary degeneration, pericyte loss, and blood-retinal barrier breakdown were prevented in these mice. Protein expression of PlGF was upregulated by diabetes, particularly in vascular cells. Diabetes-induced degradation of ZO-1 and VE-cadherin was reversed due to PlGF deficiency; their expression was correlated with that of sonic hedgehog and angiopoietin-1. PlGF deletion in Akita mice resulted in an increased Akt phosphorylation. Diabetes-activated hypoxia-inducible factor (HIF)1α-vascular endothelial growth factor (VEGF) pathway, including expression of HIF1α, VEGF, VEGFR1-3, and the extent of phospho (p)-VEGFR1, p-VEGFR2, and p-endothelial nitric oxide synthase, was inhibited in the retinas of diabetic PlGF(-/-) mice. However, expression of intercellular adhesion molecule-1, vascular cell adhesion molecule-1, CD11b, and CD18 was not inhibited by PlGF deletion, nor was retinal leukostasis. These results suggest that PlGF is critical for the development of DR, and its genetic deletion protects the retina from diabetic damage. Protective mechanisms are associated with Akt activation and HIF1α-VEGF pathway inhibition, but independent of retinal leukostasis in the retinas of diabetic PlGF(-/-) mice.
Collapse
Affiliation(s)
- Hu Huang
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jianbo He
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD Guangxi Tumor Hospital and Institute, Nanning, Guangxi, China
| | - Da'Kuawn Johnson
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yanhong Wei
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ying Liu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD Aier Eye Hospital, Changsha, Hunan, China
| | - Shuang Wang
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD China Japan Union Hospital, Changchun, Jilin, China
| | - Gerard A Lutty
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elia J Duh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
39
|
Alasztics B, Gullai N, Molvarec A, Rigó Jr. J. The role of angiogenic factors in preeclampsia. Orv Hetil 2014; 155:1860-6. [DOI: 10.1556/oh.2014.30042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Preeclampsia is one of the most common and most serious complications of pregnancy and the management of this condition still challenges obstetricians. Despite intensive research the etiology of preeclampsia still remains unclear. At the beginning of the 2000s preeclampsia-related research was directed towards factors that influence angiogenesis. Most studies have been carried out on the placental growth factor and soluble fms-like tyrosine kinase-1. Most publications confirm the increased concentrations of antiangiogenic factors and decreased concentrations of proangiogenic factors in maternal blood samples in preeclampsia even before the onset of clinical symptoms. According to our current knowledge antiangiogenic proteins are responsible for the endothelial dysfunction in the symptomatic stage of the disease. Placental growth factor and soluble fms-like tyrosine kinase-1 may have important roles in the prediction and treatment of the disease. The point of care detection of placental growth factor and soluble fms-like tyrosine kinase-1 may be used to predict preeclampsia. Rapid tests are available to determine the serum levels of the two proteins. Removal of soluble fms-like tyrosine kinase-1 from maternal circulation is a potential treatment option for early onset preeclampsia. Orv. Hetil., 2014, 155(47), 1860–1866.
Collapse
Affiliation(s)
- Bálint Alasztics
- Semmelweis Egyetem, Általános Orvostudományi Kar I. Szülészeti és Nőgyógyászati Klinika Budapest Baross u. 27. 1082
| | - Nóra Gullai
- Semmelweis Egyetem, Általános Orvostudományi Kar I. Szülészeti és Nőgyógyászati Klinika Budapest Baross u. 27. 1082
| | - Attila Molvarec
- Semmelweis Egyetem, Általános Orvostudományi Kar I. Szülészeti és Nőgyógyászati Klinika Budapest Baross u. 27. 1082
| | - János Rigó Jr.
- Semmelweis Egyetem, Általános Orvostudományi Kar I. Szülészeti és Nőgyógyászati Klinika Budapest Baross u. 27. 1082
| |
Collapse
|
40
|
Hentges CR, Silveira RC, Ferrelli RS, Procianoy RS. Influence of maternal pre-eclampsia on VEGF/PlGF heterodimer levels in preterm infants. J Matern Fetal Neonatal Med 2014; 28:2166-71. [PMID: 25354293 DOI: 10.3109/14767058.2014.980231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To measure VEGF/PlGF heterodimer levels in preterm infants born to mothers with preeclampsia. METHODS Neonates with birth weight <2000 g and gestational age ≤34 weeks were divided into two groups: born to mothers with Preeclampsia (PE) and controls. Neonates transferred from outside after the 72nd hour of life, death before blood collection, major congenital malformations or inborn errors of metabolism, and mothers with multiple pregnancies, STORCH complex infections, HIV or autoimmune conditions were excluded. Blood was collected within 72 h of birth and again at 28 days. VEGF/PlGF heterodimer levels were measured by ELISA. RESULTS We included 73 neonates (24 born to mothers with PE and 49 without PE). Mean gestational age was 30.32 ± 2.88 weeks and mean birth weight was 1288.62 ± 462.22 g. Median VEGF/PlGF levels were significantly higher in infants born to mothers with PE. VEGF/PlGF levels were inversely proportional to birth weight. There were no between-group differences in blood samples collected at age 28 days. CONCLUSION Higher VEGF/PlGF levels were higher in neonates exposed to PE, and there was a significant negative correlation between birth weight and VEGF/PlGF levels. Further studies to elucidate the role of this substance in the fetal and neonatal period are needed.
Collapse
Affiliation(s)
- Cláudia R Hentges
- a Newborn Section, Department of Pediatrics , Universidade Federal do Rio Grande do Sul and Hospital de Clinicas de Porto Alegre , RS , Brazil
| | - Rita C Silveira
- a Newborn Section, Department of Pediatrics , Universidade Federal do Rio Grande do Sul and Hospital de Clinicas de Porto Alegre , RS , Brazil
| | - Régis S Ferrelli
- a Newborn Section, Department of Pediatrics , Universidade Federal do Rio Grande do Sul and Hospital de Clinicas de Porto Alegre , RS , Brazil
| | - Renato S Procianoy
- a Newborn Section, Department of Pediatrics , Universidade Federal do Rio Grande do Sul and Hospital de Clinicas de Porto Alegre , RS , Brazil
| |
Collapse
|
41
|
Domigan CK, Ziyad S, Iruela-Arispe ML. Canonical and noncanonical vascular endothelial growth factor pathways: new developments in biology and signal transduction. Arterioscler Thromb Vasc Biol 2014; 35:30-9. [PMID: 25278287 DOI: 10.1161/atvbaha.114.303215] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The past 5 years have witnessed a significant expansion in our understanding of vascular endothelial growth factor (VEGF) signaling. In particular, the process of canonical activation of VEGF receptor tyrosine kinases by homodimeric VEGF molecules has now been broadened by the realization that heterodimeric ligands and receptors are also active participants in the signaling process. Although heterodimer receptors were described 2 decades ago, their impact, along with the effect of additional cell surface partners and novel autocrine VEGF signaling pathways, are only now starting to be clarified. Furthermore, ligand-independent signaling (noncanonical) has been identified through galectin and gremlin binding and upon rise of intracellular levels of reactive oxygen species. Activation of the VEGF receptors in the absence of ligand holds immediate implications for therapeutic approaches that exclusively target VEGF. The present review provides a concise summary of the recent developments in both canonical and noncanonical VEGF signaling and places these findings in perspective to their potential clinical and biological ramifications.
Collapse
Affiliation(s)
- Courtney K Domigan
- From the Department of Molecular, Cell, and Developmental Biology (C.K.D., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles
| | - Safiyyah Ziyad
- From the Department of Molecular, Cell, and Developmental Biology (C.K.D., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles
| | - M Luisa Iruela-Arispe
- From the Department of Molecular, Cell, and Developmental Biology (C.K.D., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles.
| |
Collapse
|
42
|
McCoy RJ, Widaa A, Watters KM, Wuerstle M, Stallings RL, Duffy GP, O'Brien FJ. Orchestrating osteogenic differentiation of mesenchymal stem cells--identification of placental growth factor as a mechanosensitive gene with a pro-osteogenic role. Stem Cells 2014; 31:2420-31. [PMID: 23897668 DOI: 10.1002/stem.1482] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 06/17/2013] [Accepted: 07/01/2013] [Indexed: 01/09/2023]
Abstract
Skeletogenesis is initiated during fetal development and persists through adult life as either a remodeling process in response to homeostatic regulation or as a regenerative process in response to physical injury. Mesenchymal stem cells (MSCs) play a crucial role providing progenitor cells from which osteoblasts, bone matrix forming cells are differentiated. The mechanical environment plays an important role in regulating stem cell differentiation into osteoblasts, however, the mechanisms by which MSCs respond to mechanical stimuli are yet to be fully elucidated. To increase understanding of MSC mechanotransuction and osteogenic differentiation, this study aimed to identify novel, mechanically augmented genes and pathways with pro-osteogenic functionality. Using collagen glycoaminoglycan scaffolds as mimics of native extracellular matrix, to create a 3D environment more representative of that found in bone, MSC-seeded constructs were mechanically stimulated in a flow-perfusion bioreactor. Global gene expression profiling techniques were used to identify potential candidates warranting further investigation. Of these, placental growth factor (PGF) was selected and expression levels were shown to strongly correlate to both the magnitude and duration of mechanical stimulation. We demonstrated that PGF gene expression was modulated through an actin polymerization-mediated mechanism. The functional role of PGF in modulating MSC osteogenic differentiation was interrogated, and we showed a concentration-dependent response whereby low concentrations exhibited the strongest pro-osteogenic effect. Furthermore, pre-osteoclast migration and differentiation, as well as endothelial cell tubule formation also maintained concentration-dependent responses to PGF, suggesting a potential role for PGF in bone resorption and angiogenesis, processes key to bone remodeling and fracture repair.
Collapse
Affiliation(s)
- Ryan J McCoy
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin 2, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland
| | | | | | | | | | | | | |
Collapse
|
43
|
Brennan LJ, Morton JS, Davidge ST. Vascular dysfunction in preeclampsia. Microcirculation 2014; 21:4-14. [PMID: 23890192 DOI: 10.1111/micc.12079] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/22/2013] [Indexed: 12/30/2022]
Abstract
Preeclampsia is a complex disorder which affects an estimated 5% of all pregnancies worldwide. It is diagnosed by hypertension in the presence of proteinuria after the 20th week of pregnancy and is a prominent cause of maternal morbidity and mortality. As delivery is currently the only known treatment, preeclampsia is also a leading cause of preterm delivery. Preeclampsia is associated with maternal vascular dysfunction, leading to serious cardiovascular risk both during and following pregnancy. Endothelial dysfunction, resulting in increased peripheral resistance, is an integral part of the maternal syndrome. While the cause of preeclampsia remains unknown, placental ischemia resulting from aberrant placentation is a fundamental characteristic of the disorder. Poor placentation is believed to stimulate the release of a number of factors including pro- and antiangiogenic factors and inflammatory activators into the maternal systemic circulation. These factors are critical mediators of vascular function and impact the endothelium in distinctive ways, including enhanced endothelial oxidative stress. The mechanisms of action and the consequences on the maternal vasculature will be discussed in this review.
Collapse
Affiliation(s)
- Lesley J Brennan
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, Alberta, Canada; Women and Children's Health Research Institute and the Cardiovascular Research Centre, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
44
|
Tal R, Seifer DB, Grazi RV, Malter HE. Follicular fluid placental growth factor is increased in polycystic ovarian syndrome: correlation with ovarian stimulation. Reprod Biol Endocrinol 2014; 12:82. [PMID: 25141961 PMCID: PMC4150963 DOI: 10.1186/1477-7827-12-82] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/15/2014] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) is characterized by increased ovarian angiogenesis and vascularity. Accumulating evidence indicates that vascular endothelial growth factor (VEGF) is increased in PCOS and may play an important role in these vascular changes and the pathogenesis of this disease. Placental growth factor (PlGF), a VEGF family member, has not been previously characterized in PCOS women. We investigated levels and temporal expression patterns of PlGF and its soluble receptor sFlt-1 (soluble Fms-like tyrosine kinase) in serum and follicular fluid (FF) of women with PCOS during controlled ovarian stimulation. METHODS This was a prospective cohort study of 14 PCOS women (Rotterdam criteria) and 14 matched controls undergoing controlled ovarian stimulation. Serum was collected on day 3, day of hCG and day of oocyte retrieval. FF was collected on retrieval day. PlGF, sFlt-1 and anti-mullerian hormone (AMH) protein concentrations were measured using ELISA. Since sFlt-1 binds free PlGF, preventing its signal transduction, we calculated PlGF bioavailability as PlGF/sFlt-1 ratio. RESULTS Serum PlGF and sFlt-1 levels were constant throughout controlled ovarian stimulation, and no significant differences were observed in either factor in PCOS women compared with non-PCOS controls at all three measured time points. However, FF PlGF levels were increased 1.5-fold in PCOS women compared with controls (p < 0.01). Moreover, FF PlGF correlated positively with number of oocytes retrieved and the ovarian reserve marker anti-mullerian hormone (AMH) and negatively with age. In addition, FF sFlt-1 levels were decreased 1.4-fold in PCOS women compared to controls (p = 0.04). PlGF bioavailability in FF was significantly greater (2-fold) in PCOS women compared with non-PCOS controls (p < 0.01). CONCLUSIONS These data provide evidence that FF PlGF correlates with ovarian stimulation and that its bioavailability is increased in women with PCOS undergoing controlled ovarian stimulation. This suggests that PlGF may play a role in PCOS pathogenesis and its angiogenic dysregulation.
Collapse
Affiliation(s)
- Reshef Tal
- Division of Reproductive Endocrinology and Infertility, Genesis Fertility & Reproductive Medicine, Maimonides Medical Center, Brooklyn, NY USA
| | - David B Seifer
- Division of Reproductive Endocrinology and Infertility, Genesis Fertility & Reproductive Medicine, Maimonides Medical Center, Brooklyn, NY USA
| | - Richard V Grazi
- Division of Reproductive Endocrinology and Infertility, Genesis Fertility & Reproductive Medicine, Maimonides Medical Center, Brooklyn, NY USA
| | - Henry E Malter
- Division of Reproductive Endocrinology and Infertility, Genesis Fertility & Reproductive Medicine, Maimonides Medical Center, Brooklyn, NY USA
| |
Collapse
|
45
|
Zhou AY, Bai YJ, Zhao M, Yu WZ, Huang LZ, Li XX. Placental growth factor expression is reversed by antivascular endothelial growth factor therapy under hypoxic conditions. World J Pediatr 2014; 10:262-70. [PMID: 25124978 DOI: 10.1007/s12519-014-0502-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 09/09/2013] [Indexed: 01/13/2023]
Abstract
BACKGROUND Clinical trials have revealed that the antivascular endothelial growth factor (VEGF) therapies are effective in retinopathy of prematurity (ROP). But the low level of VEGF was necessary as a survival signal in healthy conditions, and endogenous placental growth factor (PIGF) is redundant for development. The purpose of this study was to elucidate the PIGF expression under hypoxia as well as the influence of anti-VEGF therapy on PIGF. METHODS CoCl2-induced hypoxic human umbilical vein endothelial cells (HUVECs) were used for an in vitro study, and oxygen-induced retinopathy (OIR) mice models were used for an in vivo study. The expression patterns of PIGF under hypoxic conditions and the influence of anti-VEGF therapy on PIGF were evaluated by quantitative reverse transcription-polymerase chain reaction (RTPCR). The retinal avascular areas and neovascularization (NV) areas of anti-VEGF, anti-PIGF and combination treatments were calculated. Retina PIGF concentration was evaluated by ELISA after treatment. The vasoactive effects of exogenous PIGF on HUVECs were investigated by proliferation and migration studies. RESULTS PIGF mRNA expression was reduced by hypoxia in OIR mice, in HUVECs under hypoxia and anti-VEGF treatment. However, PIGF expression was reversed by anti-VEGF therapy in the OIR model and in HUVECs under hypoxia. Exogenous PIGF significantly inhibited HUVECs proliferation and migration under normal conditions, but it stimulated cell proliferation and migration under hypoxia. Anti-PIGF treatment was effective for neovascular tufts in OIR mice (P<0.05). CONCLUSION The finding that PIGF expression is iatrogenically up-regulated by anti-VEGF therapy provides a consideration to combine it with anti-PIGF therapy.
Collapse
Affiliation(s)
- Ai-Yi Zhou
- Department of Ophthalmology, the Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | | | | | | | | | | |
Collapse
|
46
|
Arroyo J, Price M, Straszewski-Chavez S, Torry RJ, Mor G, Torry DS. XIAP protein is induced by placenta growth factor (PLGF) and decreased during preeclampsia in trophoblast cells. Syst Biol Reprod Med 2014; 60:263-73. [DOI: 10.3109/19396368.2014.927540] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
47
|
The role of angiogenic factors in endometrial cancer. MENOPAUSE REVIEW 2014; 13:122-6. [PMID: 26327841 PMCID: PMC4520350 DOI: 10.5114/pm.2014.42714] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/04/2014] [Accepted: 02/17/2014] [Indexed: 01/09/2023]
Abstract
Endometrial cancer is the most common malignancy within the female reproductive system (37.7%). The incidence increases with age. Frequently this type of cancer is diagnosed in peri- and post-menopausal women. 60-70% of cancers occur in women over 60 years of age, and less than 5% in women below 40 years of age. Angiogenesis is a process of formation of new microvessels from existing capillaries. There are four different mechanisms of new vessel growth: sprouting, intussusception, vessel elongation and incorporation of endothelial progenitor cells into new microvessels. Angiogenesis plays important roles in growth of endometrial cancers. This process is controlled by many angiogenic factors, for example vascular endothelial growth factor (VEGF). VEGF is the most powerful and most specific endothelial cell growth factor. It plays a crucial role in the initiation of physiological and pathological angiogenesis, lymphangiogenesis, and vasculogenesis. The VEGF family consists of VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, VEGF-F and PLGF (placental growth factor). The effects of VEGF are mediated through binding to the two specific and homologous receptors VEGFR-1 (FLT-1) and VEGFR-2 (KDR). Placental growth factor (PLGF) belongs to the VEGF family and it is also a very important growth factor. So far four isoforms of PLGF have been identified: PLGF-1 (PLGF131), PLGF-2 (PLGF152), PLGF-3 (PLGF203) and PLGF-4 (PLGF224).
Collapse
|
48
|
Cavallaro G, Filippi L, Bagnoli P, La Marca G, Cristofori G, Raffaeli G, Padrini L, Araimo G, Fumagalli M, Groppo M, Dal Monte M, Osnaghi S, Fiorini P, Mosca F. The pathophysiology of retinopathy of prematurity: an update of previous and recent knowledge. Acta Ophthalmol 2014; 92:2-20. [PMID: 23617889 DOI: 10.1111/aos.12049] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Retinopathy of prematurity (ROP) is a disease that can cause blindness in very low birthweight infants. The incidence of ROP is closely correlated with the weight and the gestational age at birth. Despite current therapies, ROP continues to be a highly debilitating disease. Our advancing knowledge of the pathogenesis of ROP has encouraged investigations into new antivasculogenic therapies. The purpose of this article is to review the findings on the pathophysiological mechanisms that contribute to the transition between the first and second phases of ROP and to investigate new potential therapies. Oxygen has been well characterized for the key role that it plays in retinal neoangiogenesis. Low or high levels of pO2 regulate the normal or abnormal production of hypoxia-inducible factor 1 and vascular endothelial growth factors (VEGF), which are the predominant regulators of retinal angiogenesis. Although low oxygen saturation appears to reduce the risk of severe ROP when carefully controlled within the first few weeks of life, the optimal level of saturation still remains uncertain. IGF-1 and Epo are fundamentally required during both phases of ROP, as alterations in their protein levels can modulate disease progression. Therefore, rhIGF-1 and rhEpo were tested for their abilities to prevent the loss of vasculature during the first phase of ROP, whereas anti-VEGF drugs were tested during the second phase. At present, previous hypotheses concerning ROP should be amended with new pathogenetic theories. Studies on the role of genetic components, nitric oxide, adenosine, apelin and β-adrenergic receptor have revealed new possibilities for the treatment of ROP. The genetic hypothesis that single-nucleotide polymorphisms within the β-ARs play an active role in the pathogenesis of ROP suggests the concept of disease prevention using β-blockers. In conclusion, all factors that can mediate the progression from the avascular to the proliferative phase might have significant implications for the further understanding and treatment of ROP.
Collapse
Affiliation(s)
- Giacomo Cavallaro
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico - Università degli Studi di Milano, Milan, ItalyNICU, Medical and Surgical Feto-Neonatal Department, "A. Meyer" University Children's Hospital, Florence, ItalyDepartment of Biology, Unit of General Physiology, University of Pisa, Pisa, ItalyNeurometabolic Unit, Department of Pediatric Neurosciences, "A. Meyer" University Children's Hospital, Florence, ItalyDepartment of Ophthalmology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Siegfried G, Khatib AM. Processing of VEGF-C and -D by the Proprotein Convertases: Importance in Angiogenesis, Lymphangiogenesis, and Tumorigenesis. ACTA ACUST UNITED AC 2013. [DOI: 10.4199/c00097ed1v01y201310pac006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
Szajewski M, Kruszewski WJ, Lakomy J, Ciesielski M, Kawecki K, Jankun J, Buczek T, Szefel J. VEGF-C and VEGF-D overexpression is more common in left-sided and well-differentiated colon adenocarcinoma. Oncol Rep 2013; 31:125-30. [PMID: 24173916 DOI: 10.3892/or.2013.2821] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 10/08/2013] [Indexed: 11/05/2022] Open
Abstract
Tumour vessel network formation, including blood and lymph vessels, is a major step involved in the process of carcinogenesis. The discovery of vascular growth factors has led to a better understanding of tumour biology, thus, creating new possibilities for cancer treatment that targets angiogenesis within tumour-associated stroma, including therapy for colon cancer patients. The present study evaluated the relationships between increased expression of lymphangiogenic factors (VEGF-C and VEGF-D) and vessel density in the tumour-surrounding stroma, patient survival and other standard prognostic factors. The expression of VEGF-C and VEGF-D and vessel density were immunohistochemically assessed in 114 primary tumour specimens from colon adenocarcinoma patients after surgical resection between January 1, 2003 and December 31, 2008. Concomittant overexpression of VEGF-C and VEGF-D was found in 51 (44.7%) colon tumours and low expression was observed in 63 (55.3%) cases. Mean vessel density was 52.87/field. A significant correlation was found between the expression of factors influencing lymph vessel growth and increased vessel density in the tumour-surrounding stroma (p=0.03). A relationship between lymphangiogenic factor overexpression and left-sided tumour location was also found (p=0.00002). Overexpression of these factors was likely to occur in well-differentiated tumours (p=0.003). No association between patient survival and the expression levels of lymphangiogenic factors was observed. The study results indicate that the overexpression of lymphangiogenic factors tends to be associated with tumours of favourable prognosis, i.e. well-differentiated and those localized in the left-side of the colon.
Collapse
Affiliation(s)
- Mariusz Szajewski
- Department of Surgical Oncology, Gdynia Oncology Centre, PCK's Maritime Hospital in Gdynia, 81-519 Gdynia, Poland
| | | | | | | | | | | | | | | |
Collapse
|