1
|
Ceci C, Lacal PM, Barbaccia ML, Mercuri NB, Graziani G, Ledonne A. The VEGFs/VEGFRs system in Alzheimer's and Parkinson's diseases: Pathophysiological roles and therapeutic implications. Pharmacol Res 2024; 201:107101. [PMID: 38336311 DOI: 10.1016/j.phrs.2024.107101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The vascular endothelial growth factors (VEGFs) and their cognate receptors (VEGFRs), besides their well-known involvement in physiological angiogenesis/lymphangiogenesis and in diseases associated to pathological vessel formation, play multifaceted functions in the central nervous system (CNS). In addition to shaping brain development, by controlling cerebral vasculogenesis and regulating neurogenesis as well as astrocyte differentiation, the VEGFs/VEGFRs axis exerts essential functions in the adult brain both in physiological and pathological contexts. In this article, after describing the physiological VEGFs/VEGFRs functions in the CNS, we focus on the VEGFs/VEGFRs involvement in neurodegenerative diseases by reviewing the current literature on the rather complex VEGFs/VEGFRs contribution to the pathogenic mechanisms of Alzheimer's (AD) and Parkinson's (PD) diseases. Thereafter, based on the outcome of VEGFs/VEGFRs targeting in animal models of AD and PD, we discuss the factual relevance of pharmacological VEGFs/VEGFRs modulation as a novel and potential disease-modifying approach for these neurodegenerative pathologies. Specific VEGFRs targeting, aimed at selective VEGFR-1 inhibition, while preserving VEGFR-2 signal transduction, appears as a promising strategy to hit the molecular mechanisms underlying AD pathology. Moreover, therapeutic VEGFs-based approaches can be proposed for PD treatment, with the aim of fine-tuning their brain levels to amplify neurotrophic/neuroprotective effects while limiting an excessive impact on vascular permeability.
Collapse
Affiliation(s)
- Claudia Ceci
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Maria Luisa Barbaccia
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; IRCCS Santa Lucia Foundation, Department of Experimental Neuroscience, Rome, Italy; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Grazia Graziani
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Ada Ledonne
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; IRCCS Santa Lucia Foundation, Department of Experimental Neuroscience, Rome, Italy; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
2
|
Bokhari SMZ, Hamar P. Vascular Endothelial Growth Factor-D (VEGF-D): An Angiogenesis Bypass in Malignant Tumors. Int J Mol Sci 2023; 24:13317. [PMID: 37686121 PMCID: PMC10487419 DOI: 10.3390/ijms241713317] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Vascular endothelial growth factors (VEGFs) are the key regulators of vasculogenesis in normal and oncological development. VEGF-A is the most studied angiogenic factor secreted by malignant tumor cells under hypoxic and inflammatory stress, which made VEGF-A a rational target for anticancer therapy. However, inhibition of VEGF-A by monoclonal antibody drugs led to the upregulation of VEGF-D. VEGF-D was primarily described as a lymphangiogenic factor; however, VEGF-D's blood angiogenic potential comparable to VEGF-A has already been demonstrated in glioblastoma and colorectal carcinoma. These findings suggested a role for VEGF-D in facilitating malignant tumor growth by bypassing the anti-VEGF-A antiangiogenic therapy. Owing to its high mitogenic ability, higher affinity for VEGFR-2, and higher expression in cancer, VEGF-D might even be a stronger angiogenic driver and, hence, a better therapeutic target than VEGF-A. In this review, we summarized the angiogenic role of VEGF-D in blood vasculogenesis and its targetability as an antiangiogenic therapy in cancer.
Collapse
Affiliation(s)
| | - Peter Hamar
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary;
| |
Collapse
|
3
|
Sasai K, Tabu K, Saito T, Matsuba Y, Saido TC, Tanaka S. Difference in the malignancy between RAS and GLI1-transformed astrocytes is associated with frequency of p27 KIP1-positive cells in xenograft tissues. Pathol Res Pract 2021; 223:153465. [PMID: 33989885 DOI: 10.1016/j.prp.2021.153465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/02/2021] [Accepted: 05/02/2021] [Indexed: 10/21/2022]
Abstract
We demonstrate that the introduction of GLI1 is sufficient for immortalized human astrocytes to be transformed whereas FOXM1 fails to induce malignant transformation, suggesting differences between GLI1 and FOXM1 in terms of transforming ability despite both transcription factors being overexpressed in malignant gliomas. Moreover, in investigations of mechanisms underlying relatively less-malignant features of GLI1-transformed astrocytes, we found that p27KIP1-positive cells were frequently observed in xenografts derived from GLI1-transformed astrocytes compared to those from RAS-transformed cells. As shRNA-mediated knockdown of p27KIP1 accelerates tumor progression of GLI1-transformed astrocytes, downregulation of p27KIP1 contributes to malignant features of transformed astrocytes. We propose that the models using immortalized/transformed astrocytes are useful to identify the minimal and most crucial set of changes required for glioma formation.
Collapse
Affiliation(s)
- Ken Sasai
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Kouichi Tabu
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Yukio Matsuba
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan; WPI Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21 W10, Kita-ku, Sapporo, 001-0021, Japan
| |
Collapse
|
4
|
Kalya M, Kel A, Wlochowitz D, Wingender E, Beißbarth T. IGFBP2 Is a Potential Master Regulator Driving the Dysregulated Gene Network Responsible for Short Survival in Glioblastoma Multiforme. Front Genet 2021; 12:670240. [PMID: 34211498 PMCID: PMC8239365 DOI: 10.3389/fgene.2021.670240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/06/2021] [Indexed: 01/01/2023] Open
Abstract
Only 2% of glioblastoma multiforme (GBM) patients respond to standard therapy and survive beyond 36 months (long-term survivors, LTS), while the majority survive less than 12 months (short-term survivors, STS). To understand the mechanism leading to poor survival, we analyzed publicly available datasets of 113 STS and 58 LTS. This analysis revealed 198 differentially expressed genes (DEGs) that characterize aggressive tumor growth and may be responsible for the poor prognosis. These genes belong largely to the Gene Ontology (GO) categories “epithelial-to-mesenchymal transition” and “response to hypoxia.” In this article, we applied an upstream analysis approach that involves state-of-the-art promoter analysis and network analysis of the dysregulated genes potentially responsible for short survival in GBM. Binding sites for transcription factors (TFs) associated with GBM pathology like NANOG, NF-κB, REST, FRA-1, PPARG, and seven others were found enriched in the promoters of the dysregulated genes. We reconstructed the gene regulatory network with several positive feedback loops controlled by five master regulators [insulin-like growth factor binding protein 2 (IGFBP2), vascular endothelial growth factor A (VEGFA), VEGF165, platelet-derived growth factor A (PDGFA), adipocyte enhancer-binding protein (AEBP1), and oncostatin M (OSMR)], which can be proposed as biomarkers and as therapeutic targets for enhancing GBM prognosis. A critical analysis of this gene regulatory network gives insights into the mechanism of gene regulation by IGFBP2 via several TFs including the key molecule of GBM tumor invasiveness and progression, FRA-1. All the observations were validated in independent cohorts, and their impact on overall survival has been investigated.
Collapse
Affiliation(s)
- Manasa Kalya
- Department of Medical Bioinformatics, University Medical Center Göttingen, Göttingen, Germany.,geneXplain GmbH, Wolfenbüttel, Germany
| | - Alexander Kel
- geneXplain GmbH, Wolfenbüttel, Germany.,Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Darius Wlochowitz
- Department of Medical Bioinformatics, University Medical Center Göttingen, Göttingen, Germany
| | | | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
5
|
Cignarella A, Fadini GP, Bolego C, Trevisi L, Boscaro C, Sanga V, Seccia TM, Rosato A, Rossi GP, Barton M. Clinical Efficacy and Safety of Angiogenesis Inhibitors: Sex Differences and Current Challenges. Cardiovasc Res 2021; 118:988-1003. [PMID: 33739385 DOI: 10.1093/cvr/cvab096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
Vasoactive molecules, such as vascular endothelial growth factor (VEGF) and endothelins, share cytokine-like activities and regulate endothelial cell (EC) growth, migration and inflammation. Some endothelial mediators and their receptors are targets for currently approved angiogenesis inhibitors, drugs that are either monoclonal antibodies raised towards VEGF, or inhibitors of vascular receptor protein kinases and signaling pathways. Pharmacological interference with the protective functions of ECs results in a similar spectrum of adverse effects. Clinically, the most common side effects of VEGF signaling pathway inhibition include an increase in arterial pressure, left ventricular (LV) dysfunction ultimately causing heart failure, and thromboembolic events, including pulmonary embolism, stroke, and myocardial infarction. Sex steroids such as androgens, progestins, and estrogen and their receptors (ERα, ERβ, GPER; PR-A, PR-B; AR) have been identified as important modifiers of angiogenesis, and sex differences have been reported for anti-angiogenic drugs. This review article discusses the current challenges clinicians are facing with regard to angiogenesis inhibitor treatments, including the need to consider sex differences affecting clinical efficacy and safety. We also propose areas for future research taking into account the role of sex hormone receptors and sex chromosomes. Development of new sex-specific drugs with improved target and cell-type selectivity likely will open the way personalized medicine in men and women requiring antiangiogenic therapy and result in reduced adverse effects and improved therapeutic efficacy.
Collapse
Affiliation(s)
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Lucia Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Carlotta Boscaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Viola Sanga
- Department of Medicine, University of Padova, Italy
| | | | - Antonio Rosato
- Venetian Cancer Institute IOV - IRCCS, Padova, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy
| | | | - Matthias Barton
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy.,Molecular Internal Medicine, University of Zürich, Switzerland.,Andreas Grüntzig Foundation, Zürich, Switzerland
| |
Collapse
|
6
|
Lymphatic metastasis of bladder cancer: Molecular mechanisms, diagnosis and targeted therapy. Cancer Lett 2021; 505:13-23. [PMID: 33610730 DOI: 10.1016/j.canlet.2021.02.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/05/2021] [Accepted: 02/11/2021] [Indexed: 12/24/2022]
Abstract
Bladder cancer is the most common and lethal cancer of the urinary system. Lymphatic metastasis is the primary and main metastatic type of bladder cancer, leading to an extremely poor prognosis in patients. Therefore, a better understanding of molecular mechanisms may provide potential targets for the diagnosis and treatment of lymphatic metastasis in bladder cancer. Herein, we summarize the current knowledge of molecular mechanisms of the lymphatic metastasis in bladder cancer, including lymphangiogenesis and its regulators, noncoding RNAs, and microenvironment-associated molecules. Novel radiomics and genomics approaches have substantially improved the preoperative diagnostic accuracy of lymph node metastasis in patients with bladder cancer. Newly discovered targets may lead to promising therapeutic strategies for clinical intervention in lymphatic metastasis of bladder cancer. More basic and translational studies need to be conducted to further clarify the molecular mechanisms, and identify predictive markers and therapeutic targets of lymphatic metastasis for bladder cancer patients.
Collapse
|
7
|
Loureiro LVM, Neder L, Callegaro-Filho D, de Oliveira Koch L, Stavale JN, Malheiros SMF. The immunohistochemical landscape of the VEGF family and its receptors in glioblastomas. SURGICAL AND EXPERIMENTAL PATHOLOGY 2020. [DOI: 10.1186/s42047-020-00060-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Abstract
Background
Angiogenesis is one of the hallmarks of cancer. This complex mechanism of tumor progression provides tumors cells with essential nutrients. There have been a limited number of investigations of markers of angiogenesis in Glioblastomas (GBMs), and most previous studies have focused on VEGF-A. Recent evidence suggests that there is a complex lymphatic system in central nervous system (CNS), which suggests VEGF-C and VEGF–D as interesting biomarker candidates. This study was designed to evaluate the expressions of VEGF-A, −C, −D and their co-receptors, VEGFR-1, VEGFR-2, and VEGFR-3 by immunohistochemistry (IHC) using a series of GBMs. In addition, we evaluate any putative correlations between IHC expression levels of VEGF and clinical data of patients.
Methods
Tumor samples of 70 GBM patients (64 isocitrate dehydrogenase-1 wildtype (wtIDH-1) and 6 mutant (mutIDH-1)) were assessed by IHC using tissue microarray platforms for VEGF subunits and their co-receptors. The medical records were reviewed for clinical and therapeutic data.
Results
All VEGF subunits and receptors were highly expressed in GBMs: 57 out of 62 (91.9%), 53 out of 56 (94.6%) and 55 out of 63 cases (87.3%) showed VEGF-A, VEGF-C and -D imunoexpression, respectively. Interestingly, we had found both nuclear and cytoplasmic localization of VEGF-C staining in GBM tumor cells. The frequency of immunoexpression of VEGF receptors was the following: VEGFR-1, 65 out of 66 cases (98.5%); VEGFR-2, 63 out of 64 cases (98.4%); VEGFR-3, 49 out of 50 cases (90.0%). There were no significant differences in the patient overall survival (OS) related to the VEGF staining. A weak and monotonous correlation was observed between VEGF and its cognate receptors. The pattern of VEGF IHC was found to be similar when GBM mutIDH-1 subtypes were compared to wtIDH-1.
Conclusion
Both VEGF-C and –D, together with their receptors, were found to be overexpressed in the majority GBMs, and the IHC expression levels did not correlate with OS or IDH status. To understand the significance of the interactions and increased expression of VEGF-C, VEGF-D, VEGFR-2, and VEGFR-3 axis in GBM requires more extensive studies. Also, functional assays using a larger series of GBM is also necessary to better address the biological meaning of nuclear VEGF-C expression in tumor cells.
Collapse
|
8
|
Erkan EP, Ströbel T, Dorfer C, Sonntagbauer M, Weinhäusel A, Saydam N, Saydam O. Circulating Tumor Biomarkers in Meningiomas Reveal a Signature of Equilibrium Between Tumor Growth and Immune Modulation. Front Oncol 2019; 9:1031. [PMID: 31649887 PMCID: PMC6795693 DOI: 10.3389/fonc.2019.01031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/24/2019] [Indexed: 12/31/2022] Open
Abstract
Meningiomas are primary central nervous system (CNS) tumors that originate from the arachnoid cells of the meninges. Recurrence occurs in higher grade meningiomas and a small subset of Grade I meningiomas with benign histology. Currently, there are no established circulating tumor markers which can be used for diagnostic and prognostic purposes in a non-invasive way for meningiomas. Here, we aimed to identify potential biomarkers of meningioma in patient sera. For this purpose, we collected preoperative (n = 30) serum samples from the meningioma patients classified as Grade I (n = 23), Grade II (n = 4), or Grade III (n = 3). We used a high-throughput, multiplex immunoassay cancer panel comprising of 92 cancer-related protein biomarkers to explore the serum protein profiles of meningioma patients. We detected 14 differentially expressed proteins in the sera of the Grade I meningioma patients in comparison to the age- and gender-matched control subjects (n = 12). Compared to the control group, Grade I meningioma patients showed increased serum levels of amphiregulin (AREG), CCL24, CD69, prolactin, EGF, HB-EGF, caspase-3, and decreased levels of VEGFD, TGF-α, E-Selectin, BAFF, IL-12, CCL9, and GH. For validation studies, we utilized an independent set of meningioma tumor tissue samples (Grade I, n = 20; Grade II, n = 10; Grade III, n = 6), and found that the expressions of amphiregulin and Caspase3 are significantly increased in all grades of meningiomas either at the transcriptional or protein level, respectively. In contrast, the gene expression of VEGF-D was significantly lower in Grade I meningioma tissue samples. Taken together, our study identifies a meningioma-specific protein signature in blood circulation of meningioma patients and highlights the importance of equilibrium between tumor-promoting factors and anti-tumor immunity.
Collapse
Affiliation(s)
- Erdogan Pekcan Erkan
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Thomas Ströbel
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Christian Dorfer
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Markus Sonntagbauer
- Austrian Institute of Technology, Molecular Diagnostics Center for Health and Bioresources, Vienna, Austria
| | - Andreas Weinhäusel
- Austrian Institute of Technology, Molecular Diagnostics Center for Health and Bioresources, Vienna, Austria
| | - Nurten Saydam
- Department of Biochemistry, Molecular Biology, and Biophysics, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Okay Saydam
- Division of Hematology and Oncology, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
9
|
Increased Expression of Vascular Endothelial Growth Factor-D Following Brain Injury. Int J Mol Sci 2019; 20:ijms20071594. [PMID: 30935023 PMCID: PMC6479775 DOI: 10.3390/ijms20071594] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 01/02/2023] Open
Abstract
Alterations in the expression of the vascular endothelial growth factors (VEGF) A and B occur during blood–brain barrier (BBB) breakdown and angiogenesis following brain injury. In this study, the temporal and spatial expression of VEGF-D and VEGF receptors-2 and -3 (VEGFR-2 and VEGFR-3, respectively) was determined at the mRNA and protein level in the rat cortical cold-injury model over a period of 0.5 to 6 days post-injury. In order to relate endothelial VEGF-D protein expression with BBB breakdown, dual labeling immunofluorescence was performed using antibodies to VEGF-D and to fibronectin, a marker of BBB breakdown. In control rats, VEGF-D signal was only observed in scattered perivascular macrophages in the cerebral cortex. The upregulation of VEGF-D mRNA expression was observed in the injury site between days 0.5 to 4, coinciding with the period of BBB breakdown and angiogenesis. At the protein level, intracerebral vessels with BBB breakdown to fibronectin in the lesion on days 0.5 to 4 failed to show endothelial VEGF-D. Between days 0.5 to 6, an increased VEGF-D immunoreactivity was noted in the endothelium of pial vessels overlying the lesion site, in neutrophils, macrophages, and free endothelial cells within the lesion. The upregulation of VEGFR-2 and -3 mRNA and protein expression was observed early post-injury on day 0.5. Although there was concurrent expression of VEGF-A, VEGF-B, and VEGF-D post-injury, differences in their spatial expression during BBB breakdown and angiogenesis suggest that they have specific and separate roles in these processes.
Collapse
|
10
|
Sun L, Guo Z, Sun J, Li J, Dong Z, Zhang Y, Chen J, Kan Q, Yu Z. MiR-133a acts as an anti-oncogene in Hepatocellular carcinoma by inhibiting FOSL2 through TGF-β/Smad3 signaling pathway. Biomed Pharmacother 2018; 107:168-176. [DOI: 10.1016/j.biopha.2018.07.151] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 01/27/2023] Open
|
11
|
Jiang J, Wang S, Chen Y, Wang C, Qu C, Liu Y. Immunohistochemical characterization of lymphangiogenesis-related biomarkers in primary and recurrent gliomas: A STROBE compliant article. Medicine (Baltimore) 2018; 97:e12458. [PMID: 30278527 PMCID: PMC6181622 DOI: 10.1097/md.0000000000012458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Glial tumors constitute the majority of primary intracranial brain tumors. The expression of specific markers of lymphangiogenesis in gliomas still remains unclear.A total of 40 surgical specimens from 20 patients with recurrent gliomas were included in the study. The expression of D2-40, vascular endothelial growth factor (VEGF)-C, VEGF-D, and VEGF receptor-3 (VEGR-3) was detected by immunohistochemistry (IHC). The clinicopathologic data (p53 and Ki67) were also collected and analyzed.At relapse malignant transformation rate was 65% (13/20 cases). D2-40, VEGF-C, VEGF-D, and VEGFR-3 were expressed in 20%, 30%, 60%, and 20% of primary and 45%, 30%, 75%, and 35% of recurrent glioma tumors (P < .01, P = 1.00, P = .03, P = .03). In 13 cases with increased malignancy grade, the expression of Ki67 and p53 were higher at relapse compared with the primary tumors (P = .001, P = .045). Multivariate survival analysis showed VEGF-D was an independent prognostic factor for malignant transformation (HR = 0.376, P = .045).Glioma is easy to relapse with tumor progression. VEGF-D was an independent prognostic factor for malignant transformation.
Collapse
Affiliation(s)
- Jun Jiang
- Department of Neurosurgery, The Second Hospital, Shandong University
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute of Shandong University
| | | | - Yuan Chen
- Central Laboratory, The Second Hospital, Shandong University, Jinan, Shandong, China
| | - Chengwei Wang
- Department of Neurosurgery, The Second Hospital, Shandong University
| | - Chuncheng Qu
- Department of Neurosurgery, The Second Hospital, Shandong University
| | - Yuguang Liu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute of Shandong University
| |
Collapse
|
12
|
Santi A, Kugeratski FG, Zanivan S. Cancer Associated Fibroblasts: The Architects of Stroma Remodeling. Proteomics 2018; 18:e1700167. [PMID: 29280568 PMCID: PMC5900985 DOI: 10.1002/pmic.201700167] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/15/2017] [Indexed: 12/24/2022]
Abstract
Fibroblasts have exceptional phenotypic plasticity and capability to secrete vast amount of soluble factors, extracellular matrix components and extracellular vesicles. While in physiological conditions this makes fibroblasts master regulators of tissue homeostasis and healing of injured tissues, in solid tumors cancer associated fibroblasts (CAFs) co-evolve with the disease, and alter the biochemical and physical structure of the tumor microenvironment, as well as the behavior of the surrounding stromal and cancer cells. Thus CAFs are fundamental regulators of tumor progression and influence response to therapeutic treatments. Increasing efforts are devoted to better understand the biology of CAFs to bring insights to develop complementary strategies to target this cell type in cancer. Here we highlight components of the tumor microenvironment that play key roles in cancer progression and invasion, and provide an extensive overview of past and emerging understanding of CAF biology as well as the contribution that MS-based proteomics has made to this field.
Collapse
Affiliation(s)
- Alice Santi
- Cancer Research UK Beatson InstituteGlasgowUK
| | | | - Sara Zanivan
- Cancer Research UK Beatson InstituteGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
13
|
Davydova N, Harris NC, Roufail S, Paquet-Fifield S, Ishaq M, Streltsov VA, Williams SP, Karnezis T, Stacker SA, Achen MG. Differential Receptor Binding and Regulatory Mechanisms for the Lymphangiogenic Growth Factors Vascular Endothelial Growth Factor (VEGF)-C and -D. J Biol Chem 2016; 291:27265-27278. [PMID: 27852824 PMCID: PMC5207153 DOI: 10.1074/jbc.m116.736801] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/14/2016] [Indexed: 12/31/2022] Open
Abstract
VEGF-C and VEGF-D are secreted glycoproteins that induce angiogenesis and lymphangiogenesis in cancer, thereby promoting tumor growth and spread. They exhibit structural homology and activate VEGFR-2 and VEGFR-3, receptors on endothelial cells that signal for growth of blood vessels and lymphatics. VEGF-C and VEGF-D were thought to exhibit similar bioactivities, yet recent studies indicated distinct signaling mechanisms (e.g. tumor-derived VEGF-C promoted expression of the prostaglandin biosynthetic enzyme COX-2 in lymphatics, a response thought to facilitate metastasis via the lymphatic vasculature, whereas VEGF-D did not). Here we explore the basis of the distinct bioactivities of VEGF-D using a neutralizing antibody, peptide mapping, and mutagenesis to demonstrate that the N-terminal α-helix of mature VEGF-D (Phe93–Arg108) is critical for binding VEGFR-2 and VEGFR-3. Importantly, the N-terminal part of this α-helix, from Phe93 to Thr98, is required for binding VEGFR-3 but not VEGFR-2. Surprisingly, the corresponding part of the α-helix in mature VEGF-C did not influence binding to either VEGFR-2 or VEGFR-3, indicating distinct determinants of receptor binding by these growth factors. A variant of mature VEGF-D harboring a mutation in the N-terminal α-helix, D103A, exhibited enhanced potency for activating VEGFR-3, was able to promote increased COX-2 mRNA levels in lymphatic endothelial cells, and had enhanced capacity to induce lymphatic sprouting in vivo. This mutant may be useful for developing protein-based therapeutics to drive lymphangiogenesis in clinical settings, such as lymphedema. Our studies shed light on the VEGF-D structure/function relationship and provide a basis for understanding functional differences compared with VEGF-C.
Collapse
Affiliation(s)
- Natalia Davydova
- From the Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000
| | - Nicole C Harris
- From the Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000
| | - Sally Roufail
- From the Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000
| | - Sophie Paquet-Fifield
- From the Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000
| | - Musarat Ishaq
- From the Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000
| | - Victor A Streltsov
- the Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, Victoria 3052, and
| | - Steven P Williams
- From the Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000
| | - Tara Karnezis
- From the Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000
| | - Steven A Stacker
- From the Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000.,the Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
| | - Marc G Achen
- From the Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, .,the Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
14
|
Tarantini S, Tucsek Z, Valcarcel-Ares MN, Toth P, Gautam T, Giles CB, Ballabh P, Wei JY, Wren JD, Ashpole NM, Sonntag WE, Ungvari Z, Csiszar A. Circulating IGF-1 deficiency exacerbates hypertension-induced microvascular rarefaction in the mouse hippocampus and retrosplenial cortex: implications for cerebromicrovascular and brain aging. AGE (DORDRECHT, NETHERLANDS) 2016; 38:273-289. [PMID: 27613724 PMCID: PMC5061685 DOI: 10.1007/s11357-016-9931-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 07/12/2016] [Indexed: 05/03/2023]
Abstract
Strong epidemiological and experimental evidence indicate that both age and hypertension lead to significant functional and structural impairment of the cerebral microcirculation, predisposing to the development of vascular cognitive impairment (VCI) and Alzheimer's disease. Preclinical studies establish a causal link between cognitive decline and microvascular rarefaction in the hippocampus, an area of brain important for learning and memory. Age-related decline in circulating IGF-1 levels results in functional impairment of the cerebral microvessels; however, the mechanistic role of IGF-1 deficiency in impaired hippocampal microvascularization remains elusive. The present study was designed to characterize the additive/synergistic effects of IGF-1 deficiency and hypertension on microvascular density and expression of genes involved in angiogenesis and microvascular regression in the hippocampus. To achieve that goal, we induced hypertension in control and IGF-1 deficient mice (Igf1 f/f + TBG-Cre-AAV8) by chronic infusion of angiotensin II. We found that circulating IGF-1 deficiency is associated with decreased microvascular density and exacerbates hypertension-induced microvascular rarefaction both in the hippocampus and the neocortex. The anti-angiogenic hippocampal gene expression signature observed in hypertensive IGF-1 deficient mice in the present study provides important clues for subsequent studies to elucidate mechanisms by which hypertension may contribute to the pathogenesis and clinical manifestation of VCI. In conclusion, adult-onset, isolated endocrine IGF-1 deficiency exerts deleterious effects on the cerebral microcirculation, leading to a significant decline in cortical and hippocampal capillarity and exacerbating hypertension-induced cerebromicrovascular rarefaction. The morphological impairment of the cerebral microvasculature induced by IGF-1 deficiency and hypertension reported here, in combination with neurovascular uncoupling, increased blood-brain barrier disruption and neuroinflammation reported in previous studies likely contribute to the pathogenesis of vascular cognitive impairment in elderly hypertensive humans.
Collapse
Affiliation(s)
- Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Zsuzsanna Tucsek
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - M Noa Valcarcel-Ares
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Peter Toth
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Physiology, University of Pecs, Pecs, Hungary
| | - Tripti Gautam
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Cory B Giles
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Research Program, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
| | - Praveen Ballabh
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Pediatrics, Regional Neonatal Center, Maria Fareri Children's Hospital at Westchester Medical Center- New York Medical College, Valhalla, NY, USA
| | - Jeanne Y Wei
- Reynolds Institute on Aging and Department of Geriatrics, University of Arkansas for Medical Science, 4301 West Markham Street, No. 748, Little Rock, AR, 72205, USA
- Geriatric Research Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA
| | - Jonathan D Wren
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Research Program, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
| | - Nicole M Ashpole
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Physiology, University of Pecs, Pecs, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Department of Physiology, University of Pecs, Pecs, Hungary.
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
15
|
Sato T, Paquet-Fifield S, Harris NC, Roufail S, Turner DJ, Yuan Y, Zhang YF, Fox SB, Hibbs ML, Wilkinson-Berka JL, Williams RA, Stacker SA, Sly PD, Achen MG. VEGF-D promotes pulmonary oedema in hyperoxic acute lung injury. J Pathol 2016; 239:152-61. [PMID: 26924464 PMCID: PMC5071654 DOI: 10.1002/path.4708] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/07/2016] [Accepted: 02/17/2016] [Indexed: 12/21/2022]
Abstract
Leakage of fluid from blood vessels, leading to oedema, is a key feature of many diseases including hyperoxic acute lung injury (HALI), which can occur when patients are ventilated with high concentrations of oxygen (hyperoxia). The molecular mechanisms driving vascular leak and oedema in HALI are poorly understood. VEGF‐D is a protein that promotes blood vessel leak and oedema when overexpressed in tissues, but the role of endogenous VEGF‐D in pathological oedema was unknown. To address these issues, we exposed Vegfd‐deficient mice to hyperoxia. The resulting pulmonary oedema in Vegfd‐deficient mice was substantially reduced compared to wild‐type, as was the protein content of bronchoalveolar lavage fluid, consistent with reduced vascular leak. Vegf‐d and its receptor Vegfr‐3 were more highly expressed in lungs of hyperoxic, versus normoxic, wild‐type mice, indicating that components of the Vegf‐d signalling pathway are up‐regulated in hyperoxia. Importantly, VEGF‐D and its receptors were co‐localized on blood vessels in clinical samples of human lungs exposed to hyperoxia; hence, VEGF‐D may act directly on blood vessels to promote fluid leak. Our studies show that Vegf‐d promotes oedema in response to hyperoxia in mice and support the hypothesis that VEGF‐D signalling promotes vascular leak in human HALI. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Teruhiko Sato
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.,Ludwig Institute for Cancer Research, Parkville, Victoria, Australia
| | | | - Nicole C Harris
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.,Ludwig Institute for Cancer Research, Parkville, Victoria, Australia
| | - Sally Roufail
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Debra J Turner
- Telethon Institute for Child Health Research and Centre for Child Health Research, University of Western Australia, Nedlands, Australia
| | - Yinan Yuan
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - You-Fang Zhang
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Stephen B Fox
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Margaret L Hibbs
- Ludwig Institute for Cancer Research, Parkville, Victoria, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | | | - Richard A Williams
- Department of Pathology, University of Melbourne, Victoria, Australia.,Department of Anatomical Pathology, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Steven A Stacker
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.,Ludwig Institute for Cancer Research, Parkville, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Peter D Sly
- Telethon Institute for Child Health Research and Centre for Child Health Research, University of Western Australia, Nedlands, Australia
| | - Marc G Achen
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.,Ludwig Institute for Cancer Research, Parkville, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| |
Collapse
|
16
|
Astin JW, Crosier PS. Lymphatics, Cancer and Zebrafish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:199-218. [DOI: 10.1007/978-3-319-30654-4_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
17
|
Xu Y, Lu W, Yang P, Peng W, Wang C, Li M, Li Y, Li G, Meng N, Lin H, Kan L, Wang S, Yang S, Yu L, Zhao Y. A small molecular agent YL529 inhibits VEGF-D-induced lymphangiogenesis and metastasis in preclinical tumor models in addition to its known antitumor activities. BMC Cancer 2015; 15:525. [PMID: 26187637 PMCID: PMC4506598 DOI: 10.1186/s12885-015-1451-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 05/19/2015] [Indexed: 02/05/2023] Open
Abstract
Background The lymph node metastasis is a key early step of the tumor metastatic process. VEGFD-mediated tumor lymphangiogenesis plays a key role, since down-regulation of p-VEGFR-3 could block the lymph node metastasis. YL529 has been reported to possess potent anti-angiogenesis and antitumor activities; however, its roles in tumor-associated lymphangiogenesis and lymphatic metastasis remain unclear. Method We investigated the effect of YL529 on tumor-associated lymphangiogenesis and lymph node metastasis using in vitro lymph node metastasis models and in vivo subcutaneous tumor models in C57 BL/6 mice. Result We found that YL529 inhibited VEGF-D-induced survival, proliferation and tube-formation of Human Lymphatic Endothelial Cells. Furthermore, in established in vitro and in vivo lymph node metastasis models using VEGF-D-LL/2 cells, YL529 significantly inhibited the tumor-associated lymphangiogenesis and metastasis. At molecular level, YL529 down-regulated p-VEGFR-3, p-JNK and Bax while up-regulated Bcl-2. Conclusion YL529 provided the therapeutic benefits by both direct effects on tumor cells and inhibiting lymphangiogenesis and metastasis via the VEGFR-3 signaling pathway, which may have significant direct clinical implications.
Collapse
Affiliation(s)
- Youzhi Xu
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, 81#, Mei Shan Road, Hefei, 230032, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Wenjie Lu
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, 81#, Mei Shan Road, Hefei, 230032, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Peng Yang
- Department of Transfusion, the First Affiliated Hospital of Anhui Medical University, 81#, Mei Shan Road, Hefei, 230032, China.
| | - Wen Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China. .,Department of Oncology, The People's Hospital of Guizhou Province, 83#, Zhong Shan East Road, Guiyang, 550004, China.
| | - Chunting Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Manli Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Yan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Guobo Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Nana Meng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Hongjun Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Lixin Kan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Siying Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - Luoting Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| | - YingLan Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, and Collaborative Innovation Center for Biotherapy, Sichuan University, 17#, 3rd Section, Ren min South Road, Chengdu, 610041, China.
| |
Collapse
|
18
|
Cui Y, Osorio JC, Risquez C, Wang H, Shi Y, Gochuico BR, Morse D, Rosas IO, El-Chemaly S. Transforming growth factor-β1 downregulates vascular endothelial growth factor-D expression in human lung fibroblasts via the Jun NH2-terminal kinase signaling pathway. Mol Med 2014; 20:120-34. [PMID: 24515257 DOI: 10.2119/molmed.2013.00123] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/03/2014] [Indexed: 01/28/2023] Open
Abstract
Vascular endothelial growth factor (VEGF)-D, a member of the VEGF family, induces both angiogenesis and lymphangiogenesis by activating VEGF receptor-2 (VEGFR-2) and VEGFR-3 on the surface of endothelial cells. Transforming growth factor (TGF)-β1 has been shown to stimulate VEGF-A expression in human lung fibroblast via the Smad3 signaling pathway and to induce VEGF-C in human proximal tubular epithelial cells. However, the effects of TGF-β1 on VEGF-D regulation are unknown. To investigate the regulation of VEGF-D, human lung fibroblasts were studied under pro-fibrotic conditions in vitro and in idiopathic pulmonary fibrosis (IPF) lung tissue. We demonstrate that TGF-β1 downregulates VEGF-D expression in a dose- and time-dependent manner in human lung fibroblasts. This TGF-β1 effect can be abolished by inhibitors of TGF-β type I receptor kinase and Jun NH2-terminal kinase (JNK), but not by Smad3 knockdown. In addition, VEGF-D knockdown in human lung fibroblasts induces G1/S transition and promotes cell proliferation. Importantly, VEGF-D protein expression is decreased in lung homogenates from IPF patients compared with control lung. In IPF lung sections, fibroblastic foci show very weak VEGF-D immunoreactivity, whereas VEGF-D is abundantly expressed within alveolar interstitial cells in control lung. Taken together, our data identify a novel mechanism for downstream signal transduction induced by TGF-β1 in lung fibroblasts, through which they may mediate tissue remodeling in IPF.
Collapse
Affiliation(s)
- Ye Cui
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Juan C Osorio
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cristobal Risquez
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hao Wang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ying Shi
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Bernadette R Gochuico
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Danielle Morse
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
19
|
Stacker SA, Williams SP, Karnezis T, Shayan R, Fox SB, Achen MG. Lymphangiogenesis and lymphatic vessel remodelling in cancer. Nat Rev Cancer 2014; 14:159-72. [PMID: 24561443 DOI: 10.1038/nrc3677] [Citation(s) in RCA: 594] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The generation of new lymphatic vessels through lymphangiogenesis and the remodelling of existing lymphatics are thought to be important steps in cancer metastasis. The past decade has been exciting in terms of research into the molecular and cellular biology of lymphatic vessels in cancer, and it has been shown that the molecular control of tumour lymphangiogenesis has similarities to that of tumour angiogenesis. Nevertheless, there are significant mechanistic differences between these biological processes. We are now developing a greater understanding of the specific roles of distinct lymphatic vessel subtypes in cancer, and this provides opportunities to improve diagnostic and therapeutic approaches that aim to restrict the progression of cancer.
Collapse
Affiliation(s)
- Steven A Stacker
- 1] Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia. [3] Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Steven P Williams
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Tara Karnezis
- 1] Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
| | - Ramin Shayan
- 1] Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. [2] Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia. [3] Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia. [4] O'Brien Institute, Australian Catholic University, Fitzroy, Victoria 3065, Australia
| | - Stephen B Fox
- 1] Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia. [2] Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Marc G Achen
- 1] Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia. [3] Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| |
Collapse
|
20
|
Le C, Karnezis T, Achen MG, Stacker S, Sloan E. Lymphovascular and neural regulation of metastasis: shared tumour signalling pathways and novel therapeutic approaches. Best Pract Res Clin Anaesthesiol 2013; 27:409-25. [PMID: 24267548 PMCID: PMC4007214 DOI: 10.1016/j.bpa.2013.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/08/2013] [Indexed: 12/13/2022]
Abstract
The progression of cancer is supported by a wide variety of non-neoplastic cell types which make up the tumour stroma, including immune cells, endothelial cells, cancer-associated fibroblasts and nerve fibres. These host cells contribute molecular signals that enhance primary tumour growth and provide physical avenues for metastatic dissemination. This article provides an overview of the role of blood vessels, lymphatic vessels and nerve fibres in the tumour microenvironment and highlights the interconnected molecular signalling pathways that control their development and activation in cancer. Further, this article highlights the known pharmacological agents which target these pathways and discusses the potential therapeutic uses of drugs that target angiogenesis, lymphangiogenesis and stress-response pathways in the different stages of cancer care.
Collapse
Affiliation(s)
- C.P. Le
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - T. Karnezis
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
| | - M. G. Achen
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - S.A. Stacker
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - E.K. Sloan
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Department of Cancer Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
- Cousins Center for PNI, Semel Institute for Neuroscience and Human Behavior, UCLA AIDS Institute and Jonsson Comprehensive Cancer Center, University of California Los Angeles, USA
| |
Collapse
|
21
|
Vascular Endothelial Growth Factor-d Modulates Caliber and Function of Initial Lymphatics in the Dermis. J Invest Dermatol 2013; 133:2074-84. [DOI: 10.1038/jid.2013.83] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 01/18/2013] [Accepted: 01/23/2013] [Indexed: 12/21/2022]
|
22
|
Signaling for lymphangiogenesis via VEGFR-3 is required for the early events of metastasis. Clin Exp Metastasis 2013; 30:819-32. [PMID: 23591595 DOI: 10.1007/s10585-013-9581-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 04/02/2013] [Indexed: 01/08/2023]
Abstract
Metastasis to regional lymph nodes is an important and early event in many tumors. Vascular endothelial growth factor-C (VEGF-C), VEGF-D and their receptor VEGFR-3, play a role in tumor spread via the lymphatics, although the timing of their involvement is not understood. In contrast, VEGFR-2, activated by VEGF-A, VEGF-C and VEGF-D, is a mediator of angiogenesis and drives primary tumor growth. We demonstrate the critical role for VEGFR-3, but not VEGFR-2, in the early events of metastasis. In a tumor model exhibiting both VEGF-D-dependent angiogenesis and lymphangiogenesis, an antibody to VEGFR-2 (DC101) was capable of inhibiting angiogenesis (79 % reduction in PECAM + blood vessels) and growth (93 % reduction in tumor volume). However, unlike an anti-VEGFR-3 Mab (mF4-31C1), DC101 was not capable of eliminating either tumor lymphangiogenesis or lymphogenous metastasis (60 % reduction of lymph node metastasis by DC101 vs 95 % by mF4-31C1). Early excision of the primary tumors demonstrated that VEGF-D-mediated tumor spread precedes angiogenesis-induced growth. Small but highly metastatic primary human breast cancers had significantly higher lymphatic vessel density (23.1 vessels/mm(2)) than size-matched (11.7) or larger non-metastatic tumors (12.4) thus supporting the importance of lymphatic vessels, as opposed to angiogenesis-mediated primary tumor growth, for nodal metastasis. These results suggest that lymphangiogenesis via VEGF-D is more critical than angiogenesis for nodal metastasis.
Collapse
|
23
|
Harris NC, Davydova N, Roufail S, Paquet-Fifield S, Paavonen K, Karnezis T, Zhang YF, Sato T, Rothacker J, Nice EC, Stacker SA, Achen MG. The propeptides of VEGF-D determine heparin binding, receptor heterodimerization, and effects on tumor biology. J Biol Chem 2013; 288:8176-8186. [PMID: 23404505 DOI: 10.1074/jbc.m112.439299] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
VEGF-D is an angiogenic and lymphangiogenic glycoprotein that can be proteolytically processed generating various forms differing in subunit composition due to the presence or absence of N- and C-terminal propeptides. These propeptides flank the central VEGF homology domain, that contains the binding sites for VEGF receptors (VEGFRs), but their biological functions were unclear. Characterization of propeptide function will be important to clarify which forms of VEGF-D are biologically active and therefore clinically relevant. Here we use VEGF-D mutants deficient in either propeptide, and in the capacity to process the remaining propeptide, to monitor the functions of these domains. We report for the first time that VEGF-D binds heparin, and that the C-terminal propeptide significantly enhances this interaction (removal of this propeptide from full-length VEGF-D completely prevents heparin binding). We also show that removal of either the N- or C-terminal propeptide is required for VEGF-D to drive formation of VEGFR-2/VEGFR-3 heterodimers which have recently been shown to positively regulate angiogenic sprouting. The mature form of VEGF-D, lacking both propeptides, can also promote formation of these receptor heterodimers. In a mouse tumor model, removal of only the C-terminal propeptide from full-length VEGF-D was sufficient to enhance angiogenesis and tumor growth. In contrast, removal of both propeptides is required for high rates of lymph node metastasis. The findings reported here show that the propeptides profoundly influence molecular interactions of VEGF-D with VEGF receptors, co-receptors, and heparin, and its effects on tumor biology.
Collapse
Affiliation(s)
- Nicole C Harris
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia; Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Natalia Davydova
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Sally Roufail
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Sophie Paquet-Fifield
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Karri Paavonen
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Tara Karnezis
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - You-Fang Zhang
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Teruhiko Sato
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Julie Rothacker
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Edouard C Nice
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Steven A Stacker
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia; Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3050, Australia
| | - Marc G Achen
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia; Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3050, Australia.
| |
Collapse
|
24
|
Tumor location and nature of lymphatic vessels are key determinants of cancer metastasis. Clin Exp Metastasis 2012; 30:345-56. [PMID: 23124573 DOI: 10.1007/s10585-012-9541-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 09/28/2012] [Indexed: 10/27/2022]
Abstract
Tumor metastasis to lymph nodes is a key indicator of patient survival, and is enhanced by the neo-lymphatics induced by tumor-secreted VEGF-C or VEGF-D, acting via VEGFR-3 signalling. These targets constitute important avenues for anti-metastatic treatment. Despite this new understanding, clinical observations linking metastasis with tumor depth or location suggest that lymphangiogenic growth factors are not the sole determinants of metastasis. Here we explored the influence of tumor proximity to lymphatics capable of responding to growth factors on nodal metastasis in a murine VEGF-D over-expression tumor model. We found that primary tumor location profoundly influenced VEGF-D-mediated lymph node metastasis: 89 % of tumors associated with the flank skin metastasised, in contrast with only 19 % of tumors located more deeply on the body wall (p < 0.01). Lymphatics in metastatic tumors arose from small lymphatics, and displayed distinct molecular and morphological profiles compared with those found in normal lymphatics. Smaller lymphatic subtypes were more abundant in skin (2.5-fold, p < 0.01) than in body wall, providing a richer source of lymphatics for VEGF-D(+) skin tumors, a phenomenon also confirmed in human samples. This study shows that the proximity of a VEGF-D(+) primary tumor to small lymphatics is an important determinant of metastasis. These observations may explain why tumor location relative to the lymphatic network is prognostically important for some human cancers.
Collapse
|
25
|
Abstract
Vascular endothelial growth factor-D (VEGF-D) is a secreted glycoprotein that promotes growth of blood vessels (angiogenesis) and lymphatic vessels (lymphangiogenesis), and can induce remodeling of large lymphatics. VEGF-D enhances solid tumor growth and metastatic spread in animal models of cancer, and in some human cancers VEGF-D correlates with metastatic spread, poor patient outcome, and, potentially, with resistance to anti-angiogenic drugs. Hence, VEGF-D signaling is a potential target for novel anti-cancer therapeutics designed to enhance anti-angiogenic approaches and to restrict metastasis. In the cardiovascular system, delivery of VEGF-D in animal models enhanced angiogenesis and tissue perfusion, findings which have led to a range of clinical trials testing this protein for therapeutic angiogenesis in cardiovascular diseases. Despite these experimental and clinical developments, our knowledge of the signaling mechanisms driven by VEGF-D is still evolving--here we explore the biology of VEGF-D, its signaling mechanisms, and the clinical relevance of this growth factor.
Collapse
Affiliation(s)
- Marc G Achen
- Peter MacCallum Cancer Centre, 1 Saint Andrews Place, Locked Bag 1, A'Beckett Street, East Melbourne, Victoria 3002, Australia.
| | | |
Collapse
|
26
|
Karnezis T, Shayan R, Fox S, Achen MG, Stacker SA. The connection between lymphangiogenic signalling and prostaglandin biology: a missing link in the metastatic pathway. Oncotarget 2012; 3:893-906. [PMID: 23097685 PMCID: PMC3478465 DOI: 10.18632/oncotarget.593] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 08/17/2012] [Indexed: 12/21/2022] Open
Abstract
Substantial evidence supports important independent roles for lymphangiogenic growth factor signaling and prostaglandins in the metastatic spread of cancer. The significance of the lymphangiogenic growth factors, vascular endothelial growth factor (VEGF)-C and VEGF-D, is well established in animal models of metastasis, and a strong correlation exits between an increase in expression of VEGF-C and VEGF-D, and metastatic spread in various solid human cancers. Similarly, key enzymes that control the production of prostaglandins, cyclooxygenases (COX-1 and COX-2, prototypic targets of Non-steroidal anti-inflammatory drugs (NSAIDs)), are frequently over-expressed or de-regulated in the progression of cancer. Recent data have suggested an intersection of lymphangiogenic growth factor signaling and the prostaglandin pathways in the control of metastatic spread via the lymphatic vasculature. Furthermore, this correlates with current clinical data showing that some NSAIDs enhance the survival of cancer patients through reducing metastasis. Here, we discuss the potential biochemical and cellular basis for such anti-cancer effects of NSAIDs through the prostaglandin and VEGF signaling pathways.
Collapse
Affiliation(s)
- Tara Karnezis
- Peter MacCallum Cancer Centre, Locked Bag 1, A'Beckett Street, East Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
27
|
Preparation of human vascular endothelial growth factor-D for structural and preclinical therapeutic studies. Protein Expr Purif 2012; 82:232-9. [DOI: 10.1016/j.pep.2012.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 12/21/2011] [Accepted: 01/03/2012] [Indexed: 12/31/2022]
|
28
|
Modulation of tumor tolerance in primary central nervous system malignancies. Clin Dev Immunol 2012; 2012:937253. [PMID: 22312408 PMCID: PMC3270544 DOI: 10.1155/2012/937253] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Revised: 09/29/2011] [Accepted: 10/03/2011] [Indexed: 12/14/2022]
Abstract
Central nervous system tumors take advantage of the unique immunology of the CNS and develop exquisitely complex stromal networks that promote growth despite the presence of antigen-presenting cells and tumor-infiltrating lymphocytes. It is precisely this immunological paradox that is essential to the survival of the tumor. We review the evidence for functional CNS immune privilege and the impact it has on tumor tolerance. In this paper, we place an emphasis on the role of tumor-infiltrating myeloid cells in maintaining stromal and vascular quiescence, and we underscore the importance of indoleamine 2,3-dioxygenase activity as a myeloid-driven tumor tolerance mechanism. Much remains to be discovered regarding the tolerogenic mechanisms by which CNS tumors avoid immune clearance. Thus, it is an open question whether tumor tolerance in the brain is fundamentally different from that of peripheral sites of tumorigenesis or whether it simply stands as a particularly strong example of such tolerance.
Collapse
|
29
|
Nersita R, Matrone A, Klain M, Scavuzzo F, Vitolo G, Abbondanza C, Carlino MV, Giacco V, Amato G, Carella C. Decreased serum vascular endothelial growth factor-D levels in metastatic patients with differentiated thyroid carcinoma. Clin Endocrinol (Oxf) 2012; 76:142-6. [PMID: 21781145 DOI: 10.1111/j.1365-2265.2011.04183.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Vascular endothelial growth factor-D (VEGF-D) has been identified as one of the lymphangiogenic growth factors involved in metastatic diffusion. The aim of this study is to evaluate the serum VEGF-D levels in patients with differentiated thyroid cancer at different conditions of disease. DESIGN AND PATIENTS We studied prospectively the VEGF-D plasma levels in 96 subjects affected by differentiated thyroid cancer. The patients were divided into three groups according to the clinical and biochemical findings: patients with no evidence of disease (Cured), patients with pathological (>1 ng/ml) stimulated thyroglobulin (Tg) (Path-Tg/rhTSH) levels only after rhTSH and patients with elevated basal Tg levels (Path-Tg/LT4). RESULTS The serum VEGF-D concentrations in patients of group Cured were not different from the controls, while group Path-Tg/rhTSH showed baseline serum VEGF-D levels significantly lower than group Cured and controls (P < 0·001 and P < 0·01, respectively). Moreover, the patients of group Path-Tg/LT4 showed median serum cytokine concentrations at baseline not significantly different from the patients of group Path-Tg/rhTSH. The rhTSH stimulation did not modify the difference in serum VEGF-D levels in patients of group Cured and group Path-Tg/rhTSH. CONCLUSIONS Our data demonstrate that the VEGF-D serum levels are reduced in patients with metastases of differentiated thyroid cancer, regardless of the degree of metastatic spread. It is possible that some other molecule produced by the tumoral tissue could affect the VEGF-D physiologically produced of from different tissues, thus conducting to a decrease in the VEGF-D found in blood of patients with evidence of metastatic differentiated thyroid cancer.
Collapse
Affiliation(s)
- Roberto Nersita
- Department of Clinical and Experimental Medicine F. Magrassi and A. Lanzara, Second University of Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Farnsworth RH, Karnezis T, Shayan R, Matsumoto M, Nowell CJ, Achen MG, Stacker SA. A role for bone morphogenetic protein-4 in lymph node vascular remodeling and primary tumor growth. Cancer Res 2011; 71:6547-57. [PMID: 21868759 DOI: 10.1158/0008-5472.can-11-0200] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lymph node metastasis, an early and prognostically important event in the progression of many human cancers, is associated with expression of VEGF-D. Changes to lymph node vasculature that occur during malignant progression may create a metastatic niche capable of attracting and supporting tumor cells. In this study, we sought to characterize molecules expressed in lymph node endothelium that could represent therapeutic or prognostic targets. Differential mRNA expression profiling of endothelial cells from lymph nodes that drained metastatic or nonmetastatic primary tumors revealed genes associated with tumor progression, in particular bone morphogenetic protein-4 (BMP-4). Metastasis driven by VEGF-D was associated with reduced BMP-4 expression in high endothelial venules, where BMP-4 loss could remodel the typical high-walled phenotype to thin-walled vessels. VEGF-D expression was sufficient to suppress proliferation of the more typical BMP-4-expressing high endothelial venules in favor of remodeled vessels, and mechanistic studies indicated that VEGF receptor-2 contributed to high endothelial venule proliferation and remodeling. BMP-4 could regulate high endothelial venule phenotype and cellular function, thereby determining morphology and proliferation responses. Notably, therapeutic administration of BMP-4 suppressed primary tumor growth, acting both at the level of tumor cells and tumor stromal cells. Together, our results show that VEGF-D-driven metastasis induces vascular remodeling in lymph nodes. Furthermore, they implicate BMP-4 as a negative regulator of this process, suggesting its potential utility as a prognostic marker or antitumor agent.
Collapse
Affiliation(s)
- Rae H Farnsworth
- Ludwig Institute for Cancer Research, University of Melbourne, Parkville, Australia
| | | | | | | | | | | | | |
Collapse
|
31
|
McLelland BT, Gravano D, Castillo J, Montoy S, Manilay JO. Enhanced isolation of adult thymic epithelial cell subsets for multiparameter flow cytometry and gene expression analysis. J Immunol Methods 2011; 367:85-94. [PMID: 21354161 DOI: 10.1016/j.jim.2011.02.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 12/13/2010] [Accepted: 02/18/2011] [Indexed: 12/17/2022]
Abstract
The epithelial cells (TECs) are microenvironmental niche cells which support T lymphocyte development in the thymus. Most studies of TEC biology have focused on TEC at the fetal stage of development, whereas the biology of adult-stage TECs is not as well-understood. Delineating the molecular mechanisms that control adult TEC differentiation has implications for the success of T-lymphocyte based therapies for autoimmune diseases and induction of immunological tolerance to stem cell-derived tissues. Detailed analysis of adult TECs is technically challenging due to their rarity, their diminishing numbers with age, and the limited number of markers to distinguish between unique TEC subpopulations. Here, we have devised an improved isolation protocol for adult mouse TECs and combined it with six-color multiparameter flow cytometry. Using these techniques, we have identified four distinct subsets of CD45- EpCAM+ TECs in adult mice: a) UEA1(low) CDR1(low) (UC(low)); b) UEA1(high) CDR1(high)(UC(high)); c) UEA1(low) CDR1(high) MHC(high) (cTEC); and d) UEA1(high)CDR1(low) MHC(int/high) (mTEC). PCR analysis verified that these TEC subsets differentially expressed known TEC genes. TEC subsets were further analyzed using high-throughput quantitative PCR arrays to reveal novel genes that could be important for TEC subset maintenance. Intracellular staining for keratin-5 and keratin-8 can also be added, but our results suggest that keratin expression alone cannot be used to distinguish adult TEC subsets. Our enhanced isolation allows for detailed analysis of rare TEC subpopulations in the adult mouse at the cellular and molecular levels.
Collapse
Affiliation(s)
- Bryce T McLelland
- School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, CA 95340, USA
| | | | | | | | | |
Collapse
|
32
|
Hu J, Ye H, Fu A, Chen X, Wang Y, Chen X, Ye X, Xiao W, Duan X, Wei Y, Chen L. Deguelin--an inhibitor to tumor lymphangiogenesis and lymphatic metastasis by downregulation of vascular endothelial cell growth factor-D in lung tumor model. Int J Cancer 2010; 127:2455-66. [PMID: 20162567 DOI: 10.1002/ijc.25253] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Deguelin, a rotenoid of the flavonoid family, has been reported to possess antiproliferative and anticarcinogenic activities in several cell lines and tumor models. However, it is still unclear whether deguelin effectively inhibits tumor-associated lymphangiogenesis and lymphatic metastasis. Since tumor production of vascular endothelial cell growth factor (VEGF)-D was associated with tumor lymphangiogenesis and lymphatic metastasis, we established the mouse lymphatic metastasis model by transfecting high expression VEGF-D into LL/2 Lewis lung cells (VEGF-D-LL/2) and explored the effects of deguelin on lymphatic metastasis in the immunocompetent C57BL/6 mice. Our results indicated that deguelin inhibited proliferation, migration of VEGF-D-LL/2 cells via downregulating AKT and mitogen-activated protein kinase pathway and interfered tube formation of lymphatic vascular endothelial cells on matrigel at nanomolar concentrations. Deguelin significantly downregulated the expression of VEGF-D both at mRNA and protein levels in VEGF-D-LL/2 cells in a dose-dependent manner. In the in vivo study, intraperitoneal administration of deguelin (4 mg/kg) remarkably inhibited the tumor-associated lymphangiogenesis and lymphatic metastasis. The rates of lymph node and lung metastasis in deguelin-treated mice were 0 and 16.7% compared with 58.3 and 83.3% in control group mice, respectively. Deguelin also resulted in a remarkable delay of tumor growth and prolongation of life span. Immunohistochemical staining with antibodies against VEGF-D, LYVE-1 and VEGFR-3 revealed fewer positive vessel-like structures in deguelin-treated mice compared with control group mice. Taken together, we demonstrate for the first time that deguelin suppresses tumor-associated lymphangiogenesis and lymphatic metastasis by downregulation of VEGF-D both in vitro and in vivo.
Collapse
Affiliation(s)
- Jia Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Song W, Ruder AM, Hu L, Li Y, Ni R, Shao W, Kaslow RA, Butler M, Tang J. Genetic epidemiology of glioblastoma multiforme: confirmatory and new findings from analyses of human leukocyte antigen alleles and motifs. PLoS One 2009; 4:e7157. [PMID: 19774073 PMCID: PMC2742900 DOI: 10.1371/journal.pone.0007157] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 09/01/2009] [Indexed: 11/19/2022] Open
Abstract
Background Human leukocyte antigen (HLA) class I genes mediate cytotoxic T-lymphocyte responses and natural killer cell function. In a previous study, several HLA-B and HLA-C alleles and haplotypes were positively or negatively associated with the occurrence and prognosis of glioblastoma multiforme (GBM). Methodology/Principal Findings As an extension of the Upper Midwest Health Study, we have performed HLA genotyping for 149 GBM patients and 149 healthy control subjects from a non-metropolitan population consisting almost exclusively of European Americans. Conditional logistic regression models did not reproduce the association of HLA-B*07 or the B*07-Cw*07 haplotype with GBM. Nonetheless, HLA-A*32, which has previously been shown to predispose GBM patients to a favorable prognosis, was negatively associated with occurrence of GBM (odds ratio = 0.41, p = 0.04 by univariate analysis). Other alleles (A*29, A*30, A*31 and A*33) within the A19 serology group to which A*32 belongs showed inconsistent trends. Sequencing-based HLA-A genotyping established that A*3201 was the single A*32 allele underlying the observed association. Additional evaluation of HLA-A promoter and exon 1 sequences did not detect any unexpected single nucleotide polymorphisms that could suggest differential allelic expression. Further analyses restricted to female GBM cases and controls revealed a second association with a specific HLA-B sequence motif corresponding to Bw4-80Ile (odds ratio = 2.71, p = 0.02). Conclusions/Significance HLA-A allelic product encoded by A*3201 is likely to be functionally important to GBM. The novel, sex-specific association will require further confirmation in other representative study populations.
Collapse
Affiliation(s)
- Wei Song
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Avima M. Ruder
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Cincinnati, Ohio, United States of America
| | - Liangyuan Hu
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Yufeng Li
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Rong Ni
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Wenshuo Shao
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Richard A. Kaslow
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - MaryAnn Butler
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Cincinnati, Ohio, United States of America
| | - Jianming Tang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
34
|
Lee HS, Han J, Bai HJ, Kim KW. Brain angiogenesis in developmental and pathological processes: regulation, molecular and cellular communication at the neurovascular interface. FEBS J 2009; 276:4622-35. [PMID: 19664072 DOI: 10.1111/j.1742-4658.2009.07174.x] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The vascular network of the brain is formed by the invasion of vascular sprouts from the pia mater toward the ventricles. Following angiogenesis of the primary vascular network, brain vessels experience a maturation process known as barriergenesis, in which the blood-brain barrier is formed. In this minireview, we discuss the processes of brain angiogenesis and barriergenesis, as well as the molecular and cellular mechanisms underlying brain vessel formation. At the molecular level, angiogenesis and barriergenesis occur via the coordinated action of oxygen-responsive molecules (e.g. hypoxia-inducible factor and Src-suppressed C kinase substrate/AKAP12) and soluble factors (e.g. vascular endothelial growth factor and angiopoietin-1), as well as axon guidance molecules and neurotrophic factors. At the cellular level, we focus on neurovascular cells, such as pericytes, astrocytes, vascular smooth muscle cells, neurons and brain macrophages. Each cell type plays a unique role, and works with other types to maintain environmental homeostasis and to respond to certain stimuli. Taken together, this minireview emphasizes the importance of the coordinated action of molecules and cells at the neurovascular interface, with regards to the regulation of angiogenesis and barriergenesis.
Collapse
Affiliation(s)
- Hye Shin Lee
- Neurovascular Coordination Research Center, College of Pharmacy, Seoul National University, Korea
| | | | | | | |
Collapse
|
35
|
Kranich S, Hattermann K, Specht A, Lucius R, Mentlein R. VEGFR-3/Flt-4 mediates proliferation and chemotaxis in glial precursor cells. Neurochem Int 2009; 55:747-53. [PMID: 19646499 DOI: 10.1016/j.neuint.2009.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 07/16/2009] [Accepted: 07/21/2009] [Indexed: 01/19/2023]
Abstract
Neuronal and vascular cells share common chemical signals. Vascular endothelial growth factor (VEGF)-C and -D and their receptor VEGFR-3/Flt-4 mediate lymphangiogenesis, but they occur also in the brain. Quantitative RT-PCR of mouse brain tissues and cultivated cells showed that the VEGFR-3 gene is highest transcribed in postnatal brain and in glial precursor cells whereas VEGF-C and -D are variably produced by different neuronal and glial cells. In neurospheres (neural stem cells) VEGFR-3 was induced by differentiation with platelet-derived growth factor (PDGF). In functional studies with an A2B5- and nestin-positive, O4-negative murine glial precursor cell line, VEGF-C and -D stimulated phosphorylation of the kinases Erk1/2; this signal transduction was inhibited by UO126. Both peptides induced the proliferation of glial precursor cells which could be inhibited by UO126. Furthermore, VEGF-D considerably enhanced their migration into an open space in a wound-healing assay. These results show that VEGF-C/-D together with its receptor VEGFR-3 provides an auto-/paracrine growth and chemotactic system for glial precursors in the developing brain.
Collapse
Affiliation(s)
- Sandra Kranich
- Department of Anatomy, University of Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
| | | | | | | | | |
Collapse
|
36
|
Sie M, Wagemakers M, Molema G, Mooij JJA, de Bont ESJM, den Dunnen WFA. The angiopoietin 1/angiopoietin 2 balance as a prognostic marker in primary glioblastoma multiforme. J Neurosurg 2009; 110:147-55. [PMID: 18991494 DOI: 10.3171/2008.6.17612] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT In the present study, the authors analyzed the ANGPT1/ANGPT2 balance in the context of therapeutic outcome in 62 patients with primary glioblastomas multiforme (GBMs). METHODS The tumor tissue used was obtained in adult patients who underwent neurosurgical debulking. Microvessel density was assessed by morphometric analysis. Double immunostaining for Ki 67/CD34 and cleaved caspase-3/CD34 was used to investigate the proliferation and apoptotic fraction of both endothelial and tumor cells. The expression of VEGFs (A-D) was evaluated on immunohistochemistry. To measure tumor vascular stabilization, the ANGPT1/ANGPT2 mRNA balance was determined using real-time reverse transcriptase polymerase chain reaction. RESULTS Within the hypoxic perinecrotic tumor area, the apoptotic fraction of endothelial cells was positively correlated with VEGFA expression (p < 0.001). Higher levels of VEGFA correlated with greater proliferation of endothelial cells in the intermediate tumor area (p = 0.031). Vascular endothelial growth factor D was significantly more highly expressed within the perinecrotic tumor area compared with the intermediate tumor area (p < 0.001). Multivariate analysis showed a significant association between the ANGPT1/ANGPT2 balance and the survival time of patients with GBMs (p = 0.035). CONCLUSIONS The results of the present study suggest that the ANGPT1/ANGPT2 balance has prognostic value in patients with primary GBMs. The authors' findings support the need for further studies of the feasibility of antiangiogenic therapy in primary GBMs, with a special focus on the normalization of tumor vasculature.
Collapse
Affiliation(s)
- Mariska Sie
- Department of Pediatrics, Pediatric Oncology Division, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
The metastatic spread of tumor cells is the most lethal aspect of cancer and often occurs via the lymphatic vasculature. Both experimental tumor models and human clinicopathologic data indicate that growth of lymphatic vessels (lymphangiogenesis) near solid tumors is often associated with lymph node metastasis. Changes in the adhesive properties of lymphatic endothelium near tumors may also facilitate metastatic spread via the lymphatics. Lymphangiogenic growth factors have been identified that promote formation of tumor lymphatics and metastatic spread of tumor cells to lymph nodes. These include the secreted glycoproteins vascular endothelial growth factor-C (VEGF-C) and VEGF-D, which act via their cognate receptor tyrosine kinase VEGF receptor-3 (VEGFR-3) located on lymphatic endothelial cells. Other signaling molecules that have been reported to promote lymphangiogenesis and/or lymphatic metastasis in cancer include VEGF-A, platelet-derived growth factor-BB, and hepatocyte growth factor. However, the quantitative contribution of these proteins to tumor lymphangiogenesis and lymphatic metastasis in different tumor types requires further investigation. In addition, chemokines are thought to play a role in attracting tumor cells and lymphatic vessels to each other. Moreover, it has recently been shown that lymphangiogenic growth factors secreted from a primary tumor can induce lymphangiogenesis in nearby lymph nodes, even before arrival of tumor cells, which may facilitate further metastasis. This article provides an overview of the molecular mechanisms that control lymphatic metastasis and discusses potential therapeutic approaches for inhibiting this process in human cancer.
Collapse
Affiliation(s)
- Marc G Achen
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Victoria 3050, Melbourne, Australia.
| | | |
Collapse
|
38
|
|
39
|
Wykosky J, Gibo DM, Stanton C, Debinski W. Interleukin-13 receptor alpha 2, EphA2, and Fos-related antigen 1 as molecular denominators of high-grade astrocytomas and specific targets for combinatorial therapy. Clin Cancer Res 2008; 14:199-208. [PMID: 18172271 DOI: 10.1158/1078-0432.ccr-07-1990] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We investigated the expression of interleukin-13 receptor alpha2 (IL-13R alpha 2), EphA2, and Fos-related antigen 1 (Fra-1) in astrocytomas and normal brain. We sought to document whether the expression of the three factors changed with progression to higher grade malignancy and whether two or three targets in combination might be sufficient to target all patients with high-grade astrocytomas. EXPERIMENTAL DESIGN Immunohistochemistry was done for IL-13R alpha 2, EphA2, and Fra-1 using human brain tumor tissue microarrays containing 30 specimens of WHO grades II and III astrocytomas, 46 glioblastoma multiformes (GBM), and 9 normal brain samples. Sections were scored based on frequency and intensity of expression. Western blotting was done for all three markers using GBM tumor specimens and xenograft cell lines. Two cytotoxins, IL-13.E13K.PE38QQR and ephrinA1-PE38QQR, which target IL-13R alpha 2 or EphA2, respectively, were tested for cytotoxicity against human GBM primary explant cells and established cells. RESULTS Expression of all three proteins was significantly higher in GBM compared with normal brain, low-grade, and anaplastic astrocytomas. Greater than 95% of GBM overexpressed at least two of the three markers. Importantly, every GBM overexpressed at least one marker. Human GBM primary explant cells and cell lines were potently killed by IL-13.E13K.PE38QQR and ephrinA1-PE38QQR, in accordance with their level of expression of IL-13R alpha 2 and EphA2, respectively. CONCLUSIONS IL-13R alpha 2, EphA2, and Fra-1 are attractive therapeutic targets representing molecular denominators of high-grade astrocytomas. One hundred percent of GBM tumors overexpress at least one of these proteins, providing the basis for rational combinatorial targeted therapies/diagnostics suitable for all patients with this disease.
Collapse
Affiliation(s)
- Jill Wykosky
- Brain Tumor Center of Excellence, Comprehensive Cancer Center, Department of Neurosurgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | |
Collapse
|
40
|
Roskoski R. Vascular endothelial growth factor (VEGF) signaling in tumor progression. Crit Rev Oncol Hematol 2007; 62:179-213. [PMID: 17324579 DOI: 10.1016/j.critrevonc.2007.01.006] [Citation(s) in RCA: 434] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2006] [Revised: 01/01/2007] [Accepted: 01/29/2007] [Indexed: 12/17/2022] Open
Abstract
Vascular endothelial cells are ordinarily quiescent in adult humans and divide less than once per decade. When tumors reach a size of about 0.2-2.0mm in diameter, they become hypoxic and limited in size in the absence of angiogenesis. There are about 30 endogenous pro-angiogenic factors and about 30 endogenous anti-angiogenic factors. In order to increase in size, tumors undergo an angiogenic switch where the action of pro-angiogenic factors predominates, resulting in angiogenesis and tumor progression. One mechanism for driving angiogenesis results from the increased production of vascular endothelial growth factor (VEGF) following up-regulation of the hypoxia-inducible transcription factor. The human VEGF family consists of VEGF (VEGF-A), VEGF-B, VEGF-C, VEGF-D, and placental growth factor (PlGF). The VEGF family of receptors consists of three protein-tyrosine kinases and two non-protein kinase receptors (neuropilin-1 and -2). Owing to the importance of angiogenesis in tumor progression, inhibition of VEGF signaling represents an attractive cancer treatment.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 116A, Box 19, Horse Shoe, NC 28742, USA.
| |
Collapse
|
41
|
Grau SJ, Trillsch F, Herms J, Thon N, Nelson PJ, Tonn JC, Goldbrunner R. Expression of VEGFR3 in glioma endothelium correlates with tumor grade. J Neurooncol 2006; 82:141-50. [PMID: 17115285 DOI: 10.1007/s11060-006-9272-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2006] [Accepted: 09/13/2006] [Indexed: 11/27/2022]
Abstract
Angiogenic processes are regulated by vascular endothelial growth factors (VEGFs) and their receptors VEGFR1 (Flt-1), 2 (Flk-1) and 3 (Flt-4). While VEGFR2 is thought to play a central role in tumor angiogenesis, anti-angiogenic therapies targeting VEGFR2 in glioma models can show escape phenomena with secondary onset of angiogenesis. The purpose of this study was to find explanations for these processes by searching for alternative pathways regulating glioma angiogenesis and reveal a correlation with tumor grade. Thus, VEGFR3, which is not expressed in normal brain, and its ligands VEGF-C and -D, were assessed in high grade (WHO degrees IV, glioblastomas, GBM) and low grade gliomas [WHO degrees II astrocytomas (AII)]. In all GBM, a strong protein expression of VEGFR3 was found on tumor endothelium, VEGF-C and -D expression was found on numerous cells in areas of high vascularization. On RNA level, a significant up-regulation of VEGFR3 was detected in GBM compared to AII and non-neoplastic brain. In AII, only very moderate VEGFR3, VEGF-C and -D expression was found on protein and RNA level indicating a correlation of VEGFR3 expression with tumor grade. VEGFR3 signal in both grades was found predominantly on endothelial cells, confirmed by VEGFR3 expression on isolated CD31 positive cells and the expression of various endothelial markers on VEGFR3-positive cells isolated from GBM. The demonstration of a complete angiogenic signaling system that is dependent on tumor grade may influence the traditional paradigm of glioma angiogenesis and may provide a basis for more effective anti-angiogenic treatment strategies.
Collapse
Affiliation(s)
- S J Grau
- Department of Neurosurgery, Klinikum Grosshadern, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81373 , Munich, Germany,
| | | | | | | | | | | | | |
Collapse
|
42
|
Young MR, Colburn NH. Fra-1 a target for cancer prevention or intervention. Gene 2006; 379:1-11. [PMID: 16784822 DOI: 10.1016/j.gene.2006.05.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Revised: 04/27/2006] [Accepted: 05/02/2006] [Indexed: 01/20/2023]
Abstract
The transcription factor activator protein-1 (AP-1) has been implicated as a driver of carcinogenesis since its original characterization. Oncogenic transcription factors like AP-1 are becoming new targets for cancer intervention. Inhibitors of AP-1 have been shown to block tumor promotion, transformation, progression and invasion. The Fos related antigen-1 (Fra-1) is activated in multiple cancers and gene ablation can suppress the invasive phenotypes of many tumor cell lines. This review focuses on the regulation of fosl1 expression, stabilization and activation of the Fra-1 polypeptide and on Fra-1-mediated tumorigenesis.
Collapse
Affiliation(s)
- Matthew R Young
- Laboratory of Cancer Prevention, National Cancer Institute-Frederick, Frederick, MD 21702, USA.
| | | |
Collapse
|
43
|
Jenny B, Harrison JA, Baetens D, Tille JC, Burkhardt K, Mottaz H, Kiss JZ, Dietrich PY, De Tribolet N, Pizzolato GP, Pepper MS. Expression and localization of VEGF-C and VEGFR-3 in glioblastomas and haemangioblastomas. J Pathol 2006; 209:34-43. [PMID: 16523449 DOI: 10.1002/path.1943] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Primary human brain tumours account for approximately 2% of all cancers. High levels of expression of vascular endothelial growth factor-A (VEGF-A), a potent angiogenic factor, are linked to poor prognosis. In contrast, the potential role in human brain tumour biology of newer VEGF family members, VEGF-C and VEGF-D, both of which are lymphangiogenic factors, is poorly understood. In the present study, the expression of all VEGFs (VEGF-A, -B, -C, and -D) and their receptors (VEGFR-1, -2, and -3) has been assessed in 39 primary human brain tumours. The well-established findings were confirmed with VEGF-A. Surprisingly, however, VEGF-C and VEGF-D, as well as VEGFR-3, were expressed in some tumour types such as haemangioblastomas and glioblastomas, despite their lack of lymphatic vessels. VEGF-C and VEGFR-3 transcripts were localized to the tumour palisade around necrotic areas in glioblastomas and were evenly distributed throughout haemangioblastomas. VEGF-C protein was localized by immunohistochemistry to the palisade layer in glioblastomas. More than 50% of VEGF-C-positive cells also expressed the intermediate-stage inflammatory macrophage marker CD163; however, a significant proportion of VEGF-C-positive cells were CD163-negative. These data demonstrate the presence of molecules, primarily described as regulators of lymphangiogenesis, in normal human brain and brain tumours that are devoid of lymphatics. Their localization in macrophages points to a role in tumour-associated inflammation.
Collapse
Affiliation(s)
- B Jenny
- Department of Morphology, University Medical Centre, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Moffat BA, Chen M, Kariaapper MST, Hamstra DA, Hall DE, Stojanovska J, Johnson TD, Blaivas M, Kumar M, Chenevert TL, Rehemtulla A, Ross BD. Inhibition of Vascular Endothelial Growth Factor (VEGF)-A Causes a Paradoxical Increase in Tumor Blood Flow and Up-Regulation of VEGF-D. Clin Cancer Res 2006; 12:1525-32. [PMID: 16533777 DOI: 10.1158/1078-0432.ccr-05-1408] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Vascular endothelial growth factor (VEGF)-A is an important mediator of angiogenesis in almost all solid tumors. The aim of this study was to evaluate the effect of VEGF-A expression on tumor growth, perfusion, and chemotherapeutic efficacy in orthotopic 9L gliosarcomas. EXPERIMENTAL DESIGN Stable 9L cell lines underexpressing and overexpressing VEGF-A were generated. Anatomic, susceptibility contrast, and continuous arterial spin-labeling magnetic resonance imaging were used to quantify the volume, blood volume, and blood flow of tumors orthotopically grown from these and wild-type 9L cells. Histologic, immunohistochemical, and quantitative reverse transcription-PCR analyses were also done on excised tumors. Finally, the effects of carmustine chemotherapy were also evaluated. RESULTS Orthotopic tumors underexpressing VEGF-A had slower growth rates (increased median survival), greater blood flow, vessel density, and VEGF-D expression, but no statistical difference in blood volume and chemotherapeutic sensitivity, compared with tumors with wild-type levels of VEGF-A. Tumors overexpressing VEGF-A had faster growth rates, greater blood volume, vessel density, and blood flow but no statistical difference in VEGF-D expression and chemotherapeutic sensitivity compared with wild-type VEGF-A-expressing tumors. CONCLUSION Blood volume and blood flow are independent and different biomarkers of tumor perfusion. Therefore, both should be measured when characterizing the efficacy of antiangiogenic therapies. Underexpression of VEGF-A does not result in complete inhibition of angiogenesis. Moreover, these tumors have a different perfusion phenotype, suggesting that angiogenesis is mediated by an alternative pathway. The results indicate that VEGF-D is a plausible alternative mediator of this angiogenesis.
Collapse
Affiliation(s)
- Bradford A Moffat
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan 48109-0503, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wykosky J, Gibo DM, Stanton C, Debinski W. EphA2 as a novel molecular marker and target in glioblastoma multiforme. Mol Cancer Res 2006; 3:541-51. [PMID: 16254188 DOI: 10.1158/1541-7786.mcr-05-0056] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated the presence of EphA2, and its ligand, ephrinA1, in glioblastoma multiforme (GBM), a malignant neoplasm of glial cells, and normal brain. We also initially examined the functional importance of the interaction between EphA2 and ephrinA1 in glioma cells. Expression and localization of EphA2 and ephrinA1 in human GBM and normal brain were examined using Western blotting, immunofluorescence, and immunohistochemistry. A functional role for EphA2 was investigated by assessing the activation status of the receptor and the effect of ephrinA1 on the anchorage-independent growth and invasiveness of GBM cells. We found EphA2 to be elevated in approximately 90% of GBM specimens and cell lines but not in normal brain, whereas ephrinA1 was present at consistently low levels in both GBM and normal brain. EphA2 was activated and phosphorylated by ephrinA1 in GBM cells. Furthermore, ephrinA1 induced a prominent, dose-dependent inhibitory effect on the anchorage-independent growth and invasiveness of GBM cells highly overexpressing EphA2, which was not seen in cells expressing low levels of the receptor. Thus, EphA2 is both specifically overexpressed in GBM and expressed differentially with respect to its ligand, ephrinA1, which may reflect on the oncogenic processes of malignant glioma cells. EphA2 seems to be functionally important in GBM cells and thus may play an important role in GBM pathogenesis. Hence, EphA2 represents a new marker and novel target for the development of molecular therapeutics against GBM.
Collapse
Affiliation(s)
- Jill Wykosky
- Brain Tumor Center of Excellence, Comprehensive Cancer Center, Department of Neurosurgery, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, USA
| | | | | | | |
Collapse
|
46
|
Sipos B, Kojima M, Tiemann K, Klapper W, Kruse ML, Kalthoff H, Schniewind B, Tepel J, Weich H, Kerjaschki D, Klöppel G. Lymphatic spread of ductal pancreatic adenocarcinoma is independent of lymphangiogenesis. J Pathol 2005; 207:301-12. [PMID: 16161179 DOI: 10.1002/path.1840] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Early lymph node metastasis is common in pancreatic ductal adenocarcinoma (PDAC). The present study has examined the relationship of lymphatic spread to lymph vessel development and the expression of lymphangiogenic cytokines in a series of well-characterized PDACs. The hot spot method revealed the intratumoural and peritumoural lymphatic vessel density (LVD) to be slightly higher in PDACs than in the normal pancreas. The average intratumoural LVD, however, was strikingly decreased. There was no overexpression of vascular endothelial growth factor (VEGF)-C and VEGF-D in PDACs compared with the normal pancreas. LVD and expression of lymphangiogenic cytokines were not related to any of the biological tumour features or to patient survival. Three orthotopic nude mouse PDAC models did not reveal any increase in tumour-associated LVD, despite a high rate of lymph node metastasis. Lymph vessel proliferation was comparable in PDAC and chronic pancreatitis, in both humans and mice. In conclusion, increased lymphangiogenic activity is not required for and does not significantly affect the lymphatic spread of PDAC. The reduced number of human and murine intratumoural lymph vessels indicates that lymphatic metastasis takes place predominantly via peritumoural lymphatic vessels. The weak expression of lymphangiogenic cytokines in neoplastic cells and lymphatic vessel proliferation in peritumoural regions and chronic pancreatitis indicate that inflammation may be the reason for the low rate of lymphangiogenesis.
Collapse
Affiliation(s)
- Bence Sipos
- Department of Pathology, University of Kiel, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Malignant gliomas, and high-grade gliomas (HGG) in particular, are nonmetastasizing but locally infiltrating, hypervascularized brain tumors of poor prognosis. We found previously that a c-fos-inducible vascular endothelial growth factor D is ubiquitously up-regulated in HGG grade IV, glioblastoma multiforme, and that glioblastoma multiforme overexpress Fos-related antigen 1 (Fra-1) rather than the c-Fos. We have thus become interested in the role Fra-1 may play in malignant glioma progression/maintenance, because Fra-1 has the capacity to modulate transcription of a variety of target genes. In this work, we have analyzed the biological effects of ectopic Fra-1 expression or Fra-1 knockdown in malignant glioma cells. Ectopic Fra-1 induced prominent phenotypic changes in all three malignant glioma cell lines examined: H4, U-87 MG, and A-172 MG. These changes were reflected in cells becoming more elongated with larger number of cellular processes. Furthermore, Fra-1 transgene caused H4 cells, which do not form tumor xenografts, to regain tumorigenic capacity. The genotype of these cells changed too, because 50 of 1,056 genes examined became either up-regulated or down-regulated. Conversely, Fra-1 knockdown altered prominently the morphology, anchorage-independent growth, tumorigenic potential, and Fra-1 effector expression, such as vascular endothelial growth factor D, in HGG cells. For example, cells transfected with antisense fra-1 showed shorter cellular processes than the control cells that did not grow in agar, and their tumorigenic potential was significantly diminished. Thus, Fra-1 may likely play an important role in the maintenance/progression of malignant gliomas and potentially represents a new target for therapeutic interventions.
Collapse
Affiliation(s)
- Waldemar Debinski
- Brain Tumor Center of Excellence, Department of Neurosurgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | |
Collapse
|
48
|
Kaushal V, Mukunyadzi P, Dennis RA, Siegel ER, Johnson DE, Kohli M. Stage-Specific Characterization of the Vascular Endothelial Growth Factor Axis in Prostate Cancer: Expression of Lymphangiogenic Markers Is Associated with Advanced-Stage Disease. Clin Cancer Res 2005. [DOI: 10.1158/1078-0432.584.11.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: The vascular endothelial growth factor (VEGF) family plays a critical role in tumor angiogenesis and lymphangiogenesis. We characterized, at the mRNA and protein levels, the expression of VEGF-A and VEGF-D and their cognate receptors, VEGFR-1, VEGFR-2, and VEGFR-3 in early- and advanced-stage prostate cancer specimens.
Experimental Design: The levels of VEGF-A and VEGF-D mRNA in early- and advanced-stage specimens were compared using an angiogenic gene array and were confirmed by quantitative real-time PCR. Receptor protein levels and activation status were determined by immunoblotting. Spatial expression of the proteins was evaluated using immunohistochemistry with fresh and archival tissues from benign prostatic hypertrophy specimens, early-stage prostate specimens, and advanced-stage metastatic specimens. Circulating plasma levels of these growth factors were measured using ELISAs.
Results: We observed that expression patterns of VEGF isotypes corresponded to the prostate cancer stage: high expression of angiogenic growth factor VEGF-A was observed in early-stage prostate specimens, whereas high expression of lymphangiogenic growth factor VEGF-D was associated with advanced-stage metastatic disease. All VEGF receptors were present at variable levels in all specimens, but their activation states varied in a stage-specific manner. VEGFR-1 and, to a limited extent, VEGFR-2 were activated in early-stage specimens, whereas VEGFR-2 and VEGFR-3 were activated in advanced-stage specimens.
Conclusions: Our results suggest that lymphangiogenic markers, such as VEGF-D and VEGFR-2 and VEGFR-3, may be better than angiogenic markers as targets of therapeutic intervention in advanced-stage prostate disease.
Collapse
Affiliation(s)
| | | | | | - Eric R. Siegel
- 4Biostatistics, University of Arkansas for Medical Sciences and Departments of
| | | | - Manish Kohli
- 1Internal Medicine, Departments of
- 6Hematology/Oncology, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| |
Collapse
|
49
|
Sipos B, Klapper W, Kruse ML, Kalthoff H, Kerjaschki D, Klöppel G. Expression of lymphangiogenic factors and evidence of intratumoral lymphangiogenesis in pancreatic endocrine tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:1187-97. [PMID: 15466385 PMCID: PMC1618637 DOI: 10.1016/s0002-9440(10)63379-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Lymphangiogenesis is thought to promote the progression of malignant tumors. Because the lymphangiogenic factors vascular endothelial factor (VEGF)-C and -D are expressed in endocrine cells, we investigated their expression in pancreatic endocrine tumors (PETs) and correlated these data and intratumoral lymph vessel density (iLVD) with clinicopathological features. Lymph vessels were identified with anti-podoplanin antiserum and with podoplanin/proliferating cell nuclear antigen double labeling. PETs (n = 104) were investigated by immunohistochemical staining for VEGF, basic fibroblast growth factor, and VEGF-C expression. VEGF-C and VEGF-D mRNA were quantified by real-time reverse transcriptase-polymerase chain reaction. PETs showed higher iLVD than normal pancreata, but iLVD did not discriminate between benign and malignant PETs. In PETs proliferating lymph vessels were identified. High iLVD was associated with lymph vessel invasion and it was more frequent in angioinvasive/metastatic tumors than in grossly invasive tumors. VEGF-C expression correlated with iLVD as well as with glucagon and pancreatic polypeptide expression. PETs show intratumoral lymphangiogenesis, which is associated with VEGF-C expression in tumor cells. The association between iLVD and lymph vessel invasion and angioinvasive/metastatic features in PETs suggests that lymphangiogenesis may promote malignant progression of PETs. PET is the first human tumor entity in which VEGF-C-related intratumoral lymphangiogenesis has been demonstrated.
Collapse
Affiliation(s)
- Bence Sipos
- Department of Pathology, University of Kiel, Michaelisstr. 11, 24105 Kiel, Germany.
| | | | | | | | | | | |
Collapse
|
50
|
McColl BK, Stacker SA, Achen MG. Molecular regulation of the VEGF family â inducers of angiogenesis and lymphangiogenesis. APMIS 2004; 112:463-80. [PMID: 15563310 DOI: 10.1111/j.1600-0463.2004.apm11207-0807.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The vascular endothelial growth factor (VEGF) family of secreted glycoproteins are critical inducers of angiogenesis (growth of blood vessels) and lymphangiogenesis (growth of lymphatic vessels). These proteins are attractive therapeutic targets for blocking growth of blood vessels and lymphatics in tumors and thereby inhibiting the growth and spread of cancer -- in fact, the first VEGF inhibitor has recently entered the clinic for treatment of cancer. In addition, the VEGFs are being considered for stimulation of angiogenesis in the context of ischemic disease and lymphangiogenesis for treatment of lymphedema. These therapeutic possibilities have focused great interest on the molecular regulation of VEGF family members. Much has been learned in the past five years about the mechanisms controlling the action of the VEGFs, including the importance of hypoxia, proteolysis, transcription factors and RNA splicing. An understanding of these mechanisms offers broader opportunities to manipulate expression and activity of the VEGFs for treatment of disease.
Collapse
Affiliation(s)
- Bradley K McColl
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Victoria, Australia
| | | | | |
Collapse
|