1
|
Porreca V, Corbella E, Palmisano B, Peres M, Angelone P, Barbagallo C, Stella M, Mignogna G, Mennini G, Melandro F, Rossi M, Ragusa M, Corsi A, Riminucci M, Maras B, Mancone C. Pigment Epithelium-Derived Factor Inhibits Cell Motility and p-ERK1/2 Signaling in Intrahepatic Cholangiocarcinoma Cell Lines. BIOLOGY 2025; 14:155. [PMID: 40001923 PMCID: PMC11851717 DOI: 10.3390/biology14020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/15/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025]
Abstract
Pigment epithelium-derived factor (PEDF) is a multifunctional soluble glycoprotein, primarily known for its potent anti-angiogenic properties. In recent years, its ability to counteract cell proliferation and motility has generated interest in PEDF as a potential tumor suppressor. In the intrahepatic Cholangiocarcinoma (iCCA), PEDF, Thrombospondin 1 (THBS1), and Thrombospondin 2 (THBS2) are expressed and released into the tumor microenvironment (TME), where they promote lymphangiogenesis at the expense of the neoangiogenic program, aiding the dissemination of cancer cells via lymphatic vessels. Recently, we demonstrated that THBS1 and THBS2 directly affect iCCA cells, exacerbating their malignant behavior, while the direct role of PEDF remains to be elucidated. In this study, through a cell-based assay and molecular analysis, we investigate the direct function of PEDF on two well-established iCCA cell lines. Our results show that PEDF affects cancer cell motility in a paracrine manner, reducing their migratory and invasive capabilities. Notably, our data suggest that the PEDF-induced inhibition of motility in iCCA cells occurs through the MAPK/ERK signaling pathway, as indicated by the reduced phosphorylation of ERK1/2. Overall, this study provides the first evidence of PEDF acting as a tumor suppressor in iCCA.
Collapse
Affiliation(s)
- Veronica Porreca
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.P.); (E.C.); (B.P.); (M.P.); (P.A.); (A.C.); (M.R.)
| | - Eleonora Corbella
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.P.); (E.C.); (B.P.); (M.P.); (P.A.); (A.C.); (M.R.)
| | - Biagio Palmisano
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.P.); (E.C.); (B.P.); (M.P.); (P.A.); (A.C.); (M.R.)
| | - Marco Peres
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.P.); (E.C.); (B.P.); (M.P.); (P.A.); (A.C.); (M.R.)
| | - Pietro Angelone
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.P.); (E.C.); (B.P.); (M.P.); (P.A.); (A.C.); (M.R.)
| | - Cristina Barbagallo
- Department of Biomedical and Biotechnological Sciences-Section of Biology and Genetics, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.R.)
| | - Michele Stella
- Department of Biomedical and Biotechnological Sciences-Section of Biology and Genetics, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.R.)
| | - Giuseppina Mignogna
- Department of Biochemical Science, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (G.M.); (B.M.)
| | - Gianluca Mennini
- General Surgery and Organ Transplantation Unit, Department of General Surgery and Surgical Specialties P. Stefanini, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (G.M.); (F.M.); (M.R.)
| | - Fabio Melandro
- General Surgery and Organ Transplantation Unit, Department of General Surgery and Surgical Specialties P. Stefanini, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (G.M.); (F.M.); (M.R.)
| | - Massimo Rossi
- General Surgery and Organ Transplantation Unit, Department of General Surgery and Surgical Specialties P. Stefanini, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (G.M.); (F.M.); (M.R.)
| | - Marco Ragusa
- Department of Biomedical and Biotechnological Sciences-Section of Biology and Genetics, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.R.)
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.P.); (E.C.); (B.P.); (M.P.); (P.A.); (A.C.); (M.R.)
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.P.); (E.C.); (B.P.); (M.P.); (P.A.); (A.C.); (M.R.)
| | - Bruno Maras
- Department of Biochemical Science, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (G.M.); (B.M.)
| | - Carmine Mancone
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.P.); (E.C.); (B.P.); (M.P.); (P.A.); (A.C.); (M.R.)
| |
Collapse
|
2
|
Jones IC, Dass CR. Roles of pigment epithelium-derived factor in cardiomyocytes: implications for use as a cardioprotective therapeutic. J Pharm Pharmacol 2023:7146108. [PMID: 37104852 DOI: 10.1093/jpp/rgad037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 04/12/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVES Cardiovascular diseases are the leading cause of death worldwide, with patients having limited options for treatment. Pigment epithelium-derived factor (PEDF) is an endogenous multifunctional protein with several mechanisms of action. Recently, PEDF has emerged as a potential cardioprotective agent in response to myocardial infarction. However, PEDF is also associated with pro-apoptotic effects, complicating its role in cardioprotection. This review summarises and compares knowledge of PEDF's activity in cardiomyocytes with other cell types and draws links between them. Following this, the review offers a novel perspective of PEDF's therapeutic potential and recommends future directions to understand the clinical potential of PEDF better. KEY FINDINGS PEDF's mechanisms as a pro-apoptotic and pro-survival protein are not well understood, despite PEDF's implication in several physiological and pathological activities. However, recent evidence suggests that PEDF may have significant cardioprotective properties mediated by key regulators dependent on cell type and context. CONCLUSIONS While PEDF's cardioprotective activity shares some key regulators with its apoptotic activity, cellular context and molecular features likely allow manipulation of PEDF's cellular activity, highlighting the importance of further investigation into its activities and its potential to be applied as a therapeutic to mitigate damage from a range of cardiac pathologies.
Collapse
Affiliation(s)
- Isobel C Jones
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| |
Collapse
|
3
|
Xiang W, Li L, Hong F, Zeng Y, Zhang J, Xie J, Shen G, Wang J, Fang Z, Qi W, Yang X, Gao G, Zhou T. N-cadherin cleavage: A critical function that induces diabetic retinopathy fibrosis via regulation of β-catenin translocation. FASEB J 2023; 37:e22878. [PMID: 36939278 DOI: 10.1096/fj.202201664rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/15/2023] [Accepted: 03/02/2023] [Indexed: 03/21/2023]
Abstract
Retinal fibrosis is a severe pathological change in the late stage of diabetic retinopathy and is also the leading cause of blindness. We have previously revealed that N-cadherin was significantly increased in type 1 and type 2 diabetic mice retinas and the fibrovascular membranes from proliferative diabetic retinopathy (PDR) patients. However, whether N-cadherin directly induces retinal fibrosis in DR and the related mechanism is unknown. Here, we investigated the pathogenic role of N-cadherin in mediating retinal fibrosis and further explored the relevant therapeutic targets. We found that the level of N-cadherin was significantly increased in PDR patients and STZ-induced diabetic mice and positively correlated with the fibrotic molecules Connective Tissue Growth Factor (CTGF) and fibronectin (FN). Moreover, intravitreal injection of N-cadherin adenovirus significantly increased the expression of FN and CTGF in normal mice retinas. Mechanistically, overexpression of N-cadherin promotes N-cadherin cleavage, and N-cadherin cleavage can further induce translocation of non-p-β-catenin in the nucleus and upregulation of fibrotic molecules. Furthermore, we found a novel N-cadherin cleavage inhibitor, pigment epithelial-derived factor (PEDF), which ameliorated the N-cadherin cleavage and subsequent retinal fibrosis in diabetic mice. Thus, our findings provide novel evidence that elevated N-cadherin level not only acts as a classic EMT maker but also plays a causative role in diabetic retinal fibrosis, and targeting N-cadherin cleavage may provide a strategy to inhibit retinal fibrosis in DR patients.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Longhui Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Fuyan Hong
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yongcheng Zeng
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jin Zhang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinye Xie
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Gang Shen
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinhong Wang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhenzhen Fang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Qi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xia Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Guoquan Gao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-Sen University, Guangzhou, China
| | - Ti Zhou
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- China Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| |
Collapse
|
4
|
Wang Y, Liu X, Quan X, Qin X, Zhou Y, Liu Z, Chao Z, Jia C, Qin H, Zhang H. Pigment epithelium-derived factor and its role in microvascular-related diseases. Biochimie 2022; 200:153-171. [DOI: 10.1016/j.biochi.2022.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/19/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023]
|
5
|
Kuriyama S, Tanaka G, Takagane K, Itoh G, Tanaka M. Pigment Epithelium Derived Factor Is Involved in the Late Phase of Osteosarcoma Metastasis by Increasing Extravasation and Cell-Cell Adhesion. Front Oncol 2022; 12:818182. [PMID: 35174090 PMCID: PMC8842676 DOI: 10.3389/fonc.2022.818182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Organ tropism of metastatic cells is not well understood. To determine the key factors involved in the selection of a specific organ upon metastasis, we established metastatic cell lines and analyzed their homing to specific tissues. Toward this, 143B osteosarcoma cells were injected intracardially until the kidney-metastasizing sub-cell line Bkid was established, which significantly differed from the parental 143B cells. The candidate genes responsible for kidney metastasis were validated, and SerpinF1/Pigment epithelium derived factor (PEDF) was identified as the primary target. Bkid cells with PEDF knockdown injected intracardially did not metastasize to the kidneys. In contrast, PEDF overexpressing 143B cells injected into femur metastasized to the lungs and kidneys. PEDF triggered mesenchymal-to-epithelial transition (MET) in vitro as well as in vivo. Based on these results, we hypothesized that the MET might be a potential barrier to extravasation. PEDF overexpression in various osteosarcoma cell lines increased their extravasation to the kidneys and lungs. Moreover, when cultured close to the renal endothelial cell line TKD2, Bkid cells disturbed the TKD2 layer and hindered wound healing via the PEDF-laminin receptor (lamR) axis. Furthermore, novel interactions were observed among PEDF, lamR, lysyl oxidase-like 1 (Loxl1), and SNAI3 (Snail-like transcription factor) during endothelial-to-mesenchymal transition (EndoMT). Collectively, our results show that PEDF induces cancer cell extravasation by increasing the permeability of kidney and lung vasculature acting via lamR and its downstream genes. We also speculate that PEDF promotes extravasation via inhibiting EndoMT, and this warrants investigation in future studies.
Collapse
Affiliation(s)
- Sei Kuriyama
- Department of Molecular Medicine and Biochemistry, Graduate School and Faculty of Medicine, Akita University, Akita City, Japan
| | - Gentaro Tanaka
- Department of Molecular Medicine and Biochemistry, Graduate School and Faculty of Medicine, Akita University, Akita City, Japan.,Department of Lifescience, Faculty and Graduate School of Engineering and Resource Science, Akita University, Akita City, Japan
| | - Kurara Takagane
- Department of Molecular Medicine and Biochemistry, Graduate School and Faculty of Medicine, Akita University, Akita City, Japan
| | - Go Itoh
- Department of Molecular Medicine and Biochemistry, Graduate School and Faculty of Medicine, Akita University, Akita City, Japan
| | - Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Graduate School and Faculty of Medicine, Akita University, Akita City, Japan
| |
Collapse
|
6
|
Lai V, Neshat SY, Rakoski A, Pitingolo J, Doloff JC. Drug delivery strategies in maximizing anti-angiogenesis and anti-tumor immunity. Adv Drug Deliv Rev 2021; 179:113920. [PMID: 34384826 DOI: 10.1016/j.addr.2021.113920] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/15/2022]
Abstract
Metronomic chemotherapy has been shown to elicit anti-tumor immune response and block tumor angiogenesis distinct from that observed with maximal tolerated dose (MTD) therapy. This review delves into the mechanisms behind anti-tumor immunity and seeks to identify the differential effect of dosing regimens, including daily low-dose and medium-dose intermittent chemotherapy (MEDIC), on both innate and adaptive immune populations involved in observed anti-tumor immune response. Given reports of VEGF/VEGFR blockade antagonizing anti-tumor immunity, drug choice, dose, and selective delivery determined by advanced formulations/vehicles are highlighted as potential sources of innovation for identifying anti-angiogenic modalities that may be combined with metronomic regimens without interrupting key immune players in the anti-tumor response. Engineered drug delivery mechanisms that exhibit extended and local release of anti-angiogenic agents both alone and in combination with chemotherapeutic treatments have also been demonstrated to elicit a potent and potentially systemic anti-tumor immune response, favoring tumor regression and stasis over progression. This review examines this interplay between various cancer models, the host immune response, and select anti-cancer agents depending on drug dosing, scheduling/regimen, and delivery modality.
Collapse
Affiliation(s)
- Victoria Lai
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah Y Neshat
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amanda Rakoski
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - James Pitingolo
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joshua C Doloff
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Materials Science and Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, Division of Cancer Immunology, Sidney Kimmel Comprehensive Cancer Center and the Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
7
|
Zhang M, Tombran-Tink J, Yang S, Zhang X, Li X, Barnstable CJ. PEDF is an endogenous inhibitor of VEGF-R2 angiogenesis signaling in endothelial cells. Exp Eye Res 2021; 213:108828. [PMID: 34742690 DOI: 10.1016/j.exer.2021.108828] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/20/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023]
Abstract
Pigment epithelium derived factor (PEDF), an endogenous inhibitor of angiogenesis, targets the growth of aberrant blood vessels in many tissues, including the eye. In this study we show that PEDF prevented early mitogenic signals of vascular endothelial growth factor (VEGF-A) in primate retinal endothelial cells, blocking proliferation, migration and tube formation. PEDF inhibited the phosphorylation and activation of five major downstream VEGF-A signaling partners, namely phosphoinositide-3-OH Kinase (PI3K), AKT, FAK, Src (Y416), and PLC-γ. It did so by binding to the extracellular domain of VEGF-R2, blocking VEGF-A-induced tyrosine phosphorylation (Tyr 951 and Tyr 1175), and inhibiting VEGF-R2 receptor kinase activity. PEDF had no effect on the transcription or translation of VEGF-R2 in cultured HUVECs. PEDF also bound to the extracellular domain of VEGF-R1. We conclude that PEDF blocks the growth of new blood vessels, in part, by reducing VEGF-A activation of its key mitogenic receptor, VEGF-R2, and by preventing its downstream signals in endothelial cells.
Collapse
Affiliation(s)
- Mingliang Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 251 Fukang Road, Tianjin, 300384, China
| | - Joyce Tombran-Tink
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 251 Fukang Road, Tianjin, 300384, China; Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA.
| | - Songyang Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 251 Fukang Road, Tianjin, 300384, China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 251 Fukang Road, Tianjin, 300384, China.
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 251 Fukang Road, Tianjin, 300384, China.
| | - Colin J Barnstable
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 251 Fukang Road, Tianjin, 300384, China; Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
8
|
Riley A, Jones H, England J, Kuvshinov D, Green V, Greenman J. Identification of soluble tissue-derived biomarkers from human thyroid tissue explants maintained on a microfluidic device. Oncol Lett 2021; 22:780. [PMID: 34594421 DOI: 10.3892/ol.2021.13041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/09/2021] [Indexed: 01/08/2023] Open
Abstract
Although a large cohort of potential biomarkers for thyroid cancer aggressiveness have been tested in various formats in recent years, to the best of our knowledge, thyroglobulin and calcitonin remain the only two established biomarkers associated with thyroid cancer management. Our group has recently validated a novel means of maintaining live, human ex vivo thyroid tissue within a tissue-on-chip format. The present pilot study aimed to interrogate the tissue effluent, containing all the soluble markers released by the tissue samples maintained within the devices' tissue chamber, for the presence of markers potentially associated with thyroid cancer aggressiveness. Culture effluent from tissue samples harvested from 19 individual patients who had undergone thyroidectomy for the treatment of suspected thyroid cancer was assessed, first using a proteome profiler™ angiogenesis array kit. Patients were subcategorised as 'aggressive' if they possessed a minimum of N1b level metastases, whilst 'non-aggressive' samples were T3 or lower without evidence of multifocality; and contralateral healthy thyroid tissue was harvested for comparative studies. Levels of Serpin-F1, vascular endothelial growth factor, Thrombospondin-1 and chemokine (C-C motif) ligand were significantly altered and, thus, were further investigated using ELISA to allow for quantitative analysis. The concentration of serpin-F1 was significantly increased in the effluent of aggressive thyroid cancer tissue when compared with levels released by both non-aggressive and benign samples. The present study demonstrated the usability of microfluidic technology for the analysis of the ex vivo tissue secretome in order to identify novel biomarkers.
Collapse
Affiliation(s)
- Andrew Riley
- Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK
| | - Heidi Jones
- Department of ENT, Hull University Teaching Hospitals NHS Trust, Castle Hill Hospital, Hull HU16 5JQ, UK
| | - James England
- Department of ENT, Hull University Teaching Hospitals NHS Trust, Castle Hill Hospital, Hull HU16 5JQ, UK
| | - Dmitriy Kuvshinov
- Faculty of Engineering, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Victoria Green
- Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK
| | - John Greenman
- Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK
| |
Collapse
|
9
|
Khalid M, Paracha RZ, Nisar M, Malik S, Tariq S, Arshad I, Siddiqa A, Hussain Z, Ahmad J, Ali A. Long non-coding RNAs and their targets as potential biomarkers in breast cancer. IET Syst Biol 2021; 15:137-147. [PMID: 33991433 PMCID: PMC8675856 DOI: 10.1049/syb2.12020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/10/2021] [Accepted: 04/27/2021] [Indexed: 01/09/2023] Open
Abstract
Breast cancer is among the lethal types of cancer with a high mortality rate, globally. Its high prevalence can be controlled through improved analysis and identification of disease-specific biomarkers. Recently, long non-coding RNAs (lncRNAs) have been reported as key contributors of carcinogenesis and regulate various cellular pathways through post-transcriptional regulatory mechanisms. The specific aim of this study was to identify the novel interactions of aberrantly expressed genetic components in breast cancer by applying integrative analysis of publicly available expression profiles of both lncRNAs and mRNAs. Differential expression patterns were identified by comparing the breast cancer expression profiles of samples with controls. Significant co-expression networks were identified through WGCNA analysis. WGCNA is a systems biology approach used to elucidate the pattern of correlation between genes across microarray samples. It is also used to identify the highly correlated modules. The results obtained from this study revealed significantly differentially expressed and co-expressed lncRNAs and their cis- and trans-regulating mRNA targets which include RP11-108F13.2 targeting TAF5L, RPL23AP2 targeting CYP4F3, CYP4F8 and AL022324.2 targeting LRP5L, AL022324.3, and Z99916.3, respectively. Moreover, pathway analysis revealed the involvement of identified mRNAs and lncRNAs in major cell signalling pathways, and target mRNAs expression is also validated through cohort data. Thus, the identified lncRNAs and their target mRNAs represent novel biomarkers that could serve as potential therapeutics for breast cancer and their roles could also be further validated through wet labs to employ them as potential therapeutic targets in future.
Collapse
Affiliation(s)
- Maryam Khalid
- Research Centre for Modeling and Simulation - RCMS, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rehan Zafar Paracha
- Research Centre for Modeling and Simulation - RCMS, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Maryum Nisar
- Research Centre for Modeling and Simulation - RCMS, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Sumaira Malik
- Research Centre for Modeling and Simulation - RCMS, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Salma Tariq
- Research Centre for Modeling and Simulation - RCMS, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Iqra Arshad
- Research Centre for Modeling and Simulation - RCMS, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Amnah Siddiqa
- The Jackson Laboratory for Genomic Medicine, Connecticut, USA
| | - Zamir Hussain
- Research Centre for Modeling and Simulation - RCMS, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Jamil Ahmad
- Department of Computer Science and Information Technology, University of Malakand, Chakdara, Pakistan
| | - Amjad Ali
- Atta-ur-Rahman School of Applied Biosciences - ASAB, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| |
Collapse
|
10
|
Chen Z, Che D, Gu X, Lin J, Deng J, Jiang P, Xu K, Xu B, Zhang T. Upregulation of PEDF Predicts a Poor Prognosis and Promotes Esophageal Squamous Cell Carcinoma Progression by Modulating the MAPK/ERK Signaling Pathway. Front Oncol 2021; 11:625612. [PMID: 33718190 PMCID: PMC7953146 DOI: 10.3389/fonc.2021.625612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/18/2021] [Indexed: 02/05/2023] Open
Abstract
Invasion and metastasis represent the primary causes of therapeutic failure in patients diagnosed with esophageal squamous cell carcinoma (ESCC). The lack of effective treatment strategies for metastatic ESCC is the major cause of the low survival rate. Therefore, it is crucial to understand the molecular mechanisms underlying ESCC metastasis and identify potential biomarkers for targeted therapy. Herein, we reported that PEDF is significantly correlated with tumor cell invasion and metastasis in ESCC. The high expression of PEDF is an independent unfavorable prognostic factor for ESCC patients’ overall survival (OS). We successfully developed and verified a nomogram to predict the preoperative OS of ESCC patients, and the actual and nomogram-predicted 1-, 3-, and 5-year survival rates had good consistency. The receiver operating characteristic (ROC) curve showed that the area under the curve (AUC) values for 1-, 3- and 5- survival were 0.764, 0.871, and 0.91, respectively. Overexpression of PEDF significantly promoted the migration and invasion of ESCC cells in vitro, while silencing PEDF yielded the opposite effects. Elevated levels of PEDF altered the expression of proteins involved in epithelial–mesenchymal transition (EMT), as indicated by the upregulation of N-cadherin and the downregulation of α-catenin and E-cadherin in ESCC cells. Mechanistically, PEDF promoted tumor cell motility and EMT by activating the MAPK/ERK signaling pathway. In conclusion, our results reveal that PEDF is involved in ESCC metastasis and could act as a prognostic factor for ESCC. Our research provides a fresh perspective into the mechanism of ESCC metastasis.
Collapse
Affiliation(s)
- Zui Chen
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Di Che
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaoqiong Gu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jiamin Lin
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jing Deng
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ping Jiang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Kaixiong Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ting Zhang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
11
|
Zangouei AS, Hamidi AA, Rahimi HR, Saburi E, Mojarrad M, Moghbeli M. Chemokines as the critical factors during bladder cancer progression: an overview. Int Rev Immunol 2021; 40:344-358. [PMID: 33591855 DOI: 10.1080/08830185.2021.1877287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Bladder cancer (BCa) is one of the most frequent urogenital malignancies which is mainly observed among men. There are various genetic and environmental risk factors associated with BCa progression. Transurethral endoscopic resection and open ablative surgery are the main treatment options for muscle invasive BCa. BCG therapy is also employed following the endoscopic resection to prevent tumor relapse. The tumor microenvironment is the main interaction site of tumor cells and immune system in which the immune cells are recruited via chemokines and chemokine receptors. In present review we summarized the main chemokines and chemokine receptors which have been associated with histopathological features of BCa patients in the world. This review highlights the chemokines and chemokine receptors as critical markers in early detection and therapeutic purposes among BCa patients and clarifies their molecular functions during BCa progression and metastasis.
Collapse
Affiliation(s)
- Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Hamidi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Chen JWE, Lumibao J, Leary S, Sarkaria JN, Steelman AJ, Gaskins HR, Harley BAC. Crosstalk between microglia and patient-derived glioblastoma cells inhibit invasion in a three-dimensional gelatin hydrogel model. J Neuroinflammation 2020; 17:346. [PMID: 33208156 PMCID: PMC7677841 DOI: 10.1186/s12974-020-02026-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Glioblastoma is the most common and deadly form of primary brain cancer, accounting for more than 13,000 new diagnoses annually in the USA alone. Microglia are the innate immune cells within the central nervous system, acting as a front-line defense against injuries and inflammation via a process that involves transformation from a quiescent to an activated phenotype. Crosstalk between GBM cells and microglia represents an important axis to consider in the development of tissue engineering platforms to examine pathophysiological processes underlying GBM progression and therapy. METHODS This work used a brain-mimetic hydrogel system to study patient-derived glioblastoma specimens and their interactions with microglia. Here, glioblastoma cells were either cultured alone in 3D hydrogels or in co-culture with microglia in a manner that allowed secretome-based signaling but prevented direct GBM-microglia contact. Patterns of GBM cell invasion were quantified using a three-dimensional spheroid assay. Secretome and transcriptome (via RNAseq) were used to profile the consequences of GBM-microglia interactions. RESULTS Microglia displayed an activated phenotype as a result of GBM crosstalk. Three-dimensional migration patterns of patient-derived glioblastoma cells showed invasion was significantly decreased in response to microglia paracrine signaling. Potential molecular mechanisms underlying with this phenotype were identified from bioinformatic analysis of secretome and RNAseq data. CONCLUSION The data demonstrate a tissue engineered hydrogel platform can be used to investigate crosstalk between immune cells of the tumor microenvironment related to GBM progression. Such multi-dimensional models may provide valuable insight to inform therapeutic innovations to improve GBM treatment.
Collapse
Affiliation(s)
- Jee-Wei Emily Chen
- Department of Chemical & Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jan Lumibao
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Current Address: Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sarah Leary
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Andrew J Steelman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL, 61801, USA
| | - H Rex Gaskins
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL, 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Department of Chemical & Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL, 61801, USA.
| |
Collapse
|
13
|
Panda S, Sahoo S, Tripathy K, Singh YD, Sarma MK, Babu PJ, Singh MC. Essential oils and their pharmacotherapeutics applications in human diseases. ADVANCES IN TRADITIONAL MEDICINE 2020. [DOI: 10.1007/s13596-020-00477-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
14
|
Baxter-Holland M, Dass CR. Pigment epithelium-derived factor: a key mediator in bone homeostasis and potential for bone regenerative therapy. J Pharm Pharmacol 2018; 70:1127-1138. [DOI: 10.1111/jphp.12942] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 05/19/2018] [Indexed: 01/02/2023]
Abstract
Abstract
Objectives
Pigment epithelium-derived factor (PEDF), a multifunctional endogenous glycoprotein, has a very wide range of biological actions, notably in bone homeostasis. The question has been raised regarding the place of PEDF in the treatment of bone disorders and osteosarcoma, and its potential for tumour growth suppression.
Methods
The PubMed database was used to compile this review.
Key findings
Pigment epithelium-derived factor's actions in osteoid tissues include promoting mesenchymal stem cell commitment to osteoblasts, increasing matrix mineralisation, and promoting osteoblast proliferation. It shows potential to improve therapeutic outcomes in treatment of multiple cancer types and regrowth of bone after trauma or resection in animal studies. PEDF may possibly have a reduced adverse effect profile compared with current osteo-regenerative treatments; however, there is currently very limited evidence regarding the safety or efficacy in human models.
Summary
Pigment epithelium-derived factor is very active within the body, particularly in osseous tissue, and its physiological actions give it potential for treatment of both bone disorders and multiple tumour types. Further research is needed to ascertain the adverse effects and safety profile of PEDF as a therapeutic agent.
Collapse
Affiliation(s)
- Mia Baxter-Holland
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, WA, Australia
| |
Collapse
|
15
|
Cheng Y, Meng Q, Huang L, Shi X, Hou J, Li X, Liang J. iTRAQ-based quantitative proteomic analysis and bioinformatics study of proteins in retinoblastoma. Oncol Lett 2018; 14:8084-8091. [PMID: 29344252 DOI: 10.3892/ol.2017.7221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/16/2017] [Indexed: 01/08/2023] Open
Abstract
The aim of the present study was to analyze proteins in the aqueous humor (AH) of patients' retinoblastoma (RB), and investigate their potential role in RB using the comparative proteomic technique of isobaric tags for relative and absolute quantitation (iTRAQ) coupled with offline two-dimensional liquid chromatography-tandem mass spectrometry. A total of 0.1 ml AH was collected from 10 children with RB (mean age, 3.8 years; range, 2-5 years) and patients with senile cataracts (mean age, 70.4 years; range, 65-79 years), which was used as the control. iTRAQ was used to analyze proteins in the AH of patients and controls. Proteins with a fold change of >1.20 or <0.83 were considered to be significantly differentially expressed (with corrected P<0.05). The identified proteins were subjected to subsequent gene ontology (GO) analysis using the DAVID database. A total of 83 proteins that were expressed differently between the controls and patients' AH samples were identified using iTRAQ analysis. Of these proteins, 44 were upregulated and 39 were downregulated. On the basis of biological processes in GO, the identified proteins were primarily involved in glycoprotein, amyloid acute-inflammatory and defensive responses. Among these proteins, pigment epithelium-derived factor serves a potential role in the treatment of RB, and stimulated by retinoic acid 6 may serve as a potential protein involved in RB development. To the best of our knowledge, the present study is the first to identify 83 proteins associated with RB using iTRAQ technology. The results of the present study will aid in furthering the understanding of RB and developing novel therapy targets in the future.
Collapse
Affiliation(s)
- Yong Cheng
- Ophthalmology Department, Peking University People's Hospital, Beijing 100044, P.R. China.,Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100044, P.R. China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Qingyu Meng
- Ophthalmology Department, Peking University People's Hospital, Beijing 100044, P.R. China.,Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100044, P.R. China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Lvzhen Huang
- Ophthalmology Department, Peking University People's Hospital, Beijing 100044, P.R. China.,Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100044, P.R. China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Xuan Shi
- Ophthalmology Department, Peking University People's Hospital, Beijing 100044, P.R. China.,Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100044, P.R. China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Jing Hou
- Ophthalmology Department, Peking University People's Hospital, Beijing 100044, P.R. China.,Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100044, P.R. China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Xiaoxin Li
- Ophthalmology Department, Peking University People's Hospital, Beijing 100044, P.R. China.,Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100044, P.R. China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Jianhong Liang
- Ophthalmology Department, Peking University People's Hospital, Beijing 100044, P.R. China.,Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100044, P.R. China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Peking University People's Hospital, Beijing 100044, P.R. China
| |
Collapse
|
16
|
Parkin JD, San Antonio JD, Persikov AV, Dagher H, Dalgleish R, Jensen ST, Jeunemaitre X, Savige J. The collαgen III fibril has a "flexi-rod" structure of flexible sequences interspersed with rigid bioactive domains including two with hemostatic roles. PLoS One 2017; 12:e0175582. [PMID: 28704418 PMCID: PMC5509119 DOI: 10.1371/journal.pone.0175582] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/20/2017] [Indexed: 01/18/2023] Open
Abstract
Collagen III is critical to the integrity of blood vessels and distensible organs, and in hemostasis. Examination of the human collagen III interactome reveals a nearly identical structural arrangement and charge distribution pattern as for collagen I, with cell interaction domains, fibrillogenesis and enzyme cleavage domains, several major ligand-binding regions, and intermolecular crosslink sites at the same sites. These similarities allow heterotypic fibril formation with, and substitution by, collagen I in embryonic development and wound healing. The collagen III fibril assumes a "flexi-rod" structure with flexible zones interspersed with rod-like domains, which is consistent with the molecule's prominence in young, pliable tissues and distensible organs. Collagen III has two major hemostasis domains, with binding motifs for von Willebrand factor, α2β1 integrin, platelet binding octapeptide and glycoprotein VI, consistent with the bleeding tendency observed with COL3A1 disease-causing sequence variants.
Collapse
Affiliation(s)
- J. Des Parkin
- From the University of Melbourne Department of Medicine (Northern Health), Melbourne, VIC, Australia
| | - James D. San Antonio
- Operations, Stryker Global Quality and Operations, Malvern, PA, United States of America
| | - Anton V. Persikov
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Carl Icahn Lab, Princeton, NJ, United States of America
| | - Hayat Dagher
- From the University of Melbourne Department of Medicine (Northern Health), Melbourne, VIC, Australia
| | - Raymond Dalgleish
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - Shane T. Jensen
- Wharton Business School, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Xavier Jeunemaitre
- INSERM U970 Paris Cardiovascular Research Centre, Paris France
- University Paris Descartes, Paris Sorbonne Cite, Paris, France
| | - Judy Savige
- From the University of Melbourne Department of Medicine (Northern Health), Melbourne, VIC, Australia
- * E-mail:
| |
Collapse
|
17
|
Belkacemi L, Zhang SX. Anti-tumor effects of pigment epithelium-derived factor (PEDF): implication for cancer therapy. A mini-review. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:4. [PMID: 26746675 PMCID: PMC4706649 DOI: 10.1186/s13046-015-0278-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/22/2015] [Indexed: 12/22/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a secreted glycoprotein and a non-inhibitory member of the serine protease inhibitor (serpin) family. It is widely expressed in human fetal and adult tissues but its expression decreases with age and in malignant tissues. The main anti-cancer activities of PEDF derive from its dual effects, either indirectly on the tumor microenvironment (indirect antitumor action) or directly on the tumor itself (direct antitumor influence). The indirect antitumor activities of PEDF were uncovered from the early findings that it stimulates retinoblastoma cell differentiation and that additionally it possesses anti-angiogenic, anti-tumorigenic and anti-metastatic properties. The mechanisms of its direct antitumor effect, however, have not been fully elucidated. This review highlights recent progress in our understanding of the multifunctional activities of PEDF and, in particular, its anti-cancer signaling mechanisms. Additionally, we discuss the possibility of using novel phosphaplatin compounds that can upregulate PEDF expression as a chemotherapy for cancer treatment.
Collapse
Affiliation(s)
- Louiza Belkacemi
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, 77204, USA.
| | - Shaun Xiaoliu Zhang
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA. .,Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
18
|
Pigment Epithelium-Derived Factor (PEDF) is a Determinant of Stem Cell Fate: Lessons from an Ultra-Rare Disease. J Dev Biol 2015; 3:112-128. [PMID: 27239449 PMCID: PMC4883593 DOI: 10.3390/jdb3040112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
PEDF is a secreted glycoprotein that is widely expressed by multiple organs. Numerous functional contributions have been attributed to PEDF with antiangiogenic, antitumor, anti-inflammatory, and neurotrophic properties among the most prominent. The discovery that null mutations in the PEDF gene results in Osteogenesis Imperfecta Type VI, a rare autosomal recessive bone disease characterized by multiple fractures, highlights a critical developmental function for this protein. This ultra-rare orphan disease has provided biological insights into previous studies that noted PEDF’s effects on various stem cell populations. In addition to bone development, PEDF modulates resident stem cell populations in the brain, muscle, and eye. Functional effects on human embryonic stem cells have also been demonstrated. An overview of recent advances in our understanding by which PEDF regulates stem cells and their potential clinical applications will be evaluated in this review.
Collapse
|
19
|
Hong H, Zhou T, Fang S, Jia M, Xu Z, Dai Z, Li C, Li S, Li L, Zhang T, Qi W, Bardeesi ASA, Yang Z, Cai W, Yang X, Gao G. Pigment epithelium-derived factor (PEDF) inhibits breast cancer metastasis by down-regulating fibronectin. Breast Cancer Res Treat 2014; 148:61-72. [PMID: 25284724 DOI: 10.1007/s10549-014-3154-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 09/24/2014] [Indexed: 11/29/2022]
Abstract
Pigment epithelium-derived factor (PEDF) plays an important role in the tumor growth and metastasis inhibition. It has been reported that PEDF expression is significantly reduced in breast cancer, and associated with disease progression and poor patient outcome. However, the exact mechanism of PEDF on breast cancer metastasis including liver and lung metastasis remains unclear. The present study aims to reveal the impact of PEDF on breast cancer. The orthotopic tumor mice model inoculated by MDA-MB-231 cells stably expressing PEDF or control cells was used to assess liver and lung metastasis of breast cancer. In vitro, migration and invasion experiments were used to detect the metastatic abilities of MDA-MB-231 and SKBR3 breast cancer cells with or without overexpression of PEDF. The metastatic-related molecules including EMT makers, fibronectin, and p-AKT and p-ERK were detected by qRT-PCR, Western blot, and Fluorescent immunocytochemistry. PEDF significantly inhibited breast cancer growth and metastasis in vivo and in vitro. Mechanically, PEDF inhibited breast cancer cell migration and invasion by down-regulating fibronectin and subsequent MMP2/MMP9 reduction via p-ERK and p-AKT signaling pathways. However, PEDF had no effect on EMT conversion in the breast cancer cells which was usually involved in cancer metastasis. Furthermore, the study showed that laminin receptor mediated the down-regulation of fibronectin by PEDF. These results reported for the first time that PEDF inhibited breast cancer metastasis by down-regulating fibronectin via laminin receptor/AKT/ERK pathway. Our findings demonstrated PEDF as a dual effector in limiting breast cancer growth and metastasis and highlighted a new avenue to block breast cancer progression.
Collapse
Affiliation(s)
- Honghai Hong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, 510080, Guangdong, China,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Li L, Yao YC, Fang SH, Ma CQ, Cen Y, Xu ZM, Dai ZY, Li C, Li S, Zhang T, Hong HH, Qi WW, Zhou T, Li CY, Yang X, Gao GQ. Pigment epithelial-derived factor (PEDF)-triggered lung cancer cell apoptosis relies on p53 protein-driven Fas ligand (Fas-L) up-regulation and Fas protein cell surface translocation. J Biol Chem 2014; 289:30785-30799. [PMID: 25225287 DOI: 10.1074/jbc.m114.590000] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF), a potent antiangiogenesis agent, has recently attracted attention for targeting tumor cells in several types of tumors. However, less is known about the apoptosis-inducing effect of PEDF on human lung cancer cells and the underlying molecular events. Here we report that PEDF has a growth-suppressive and proapoptotic effect on lung cancer xenografts. Accordingly, in vitro, PEDF apparently induced apoptosis in A549 and Calu-3 cells, predominantly via the Fas-L/Fas death signaling pathway. Interestingly, A549 and Calu-3 cells are insensitive to the Fas-L/Fas apoptosis pathway because of the low level of cell surface Fas. Our results revealed that, in addition to the enhancement of Fas-L expression, PEDF increased the sensitivity of A549 and Calu-3 cells to Fas-L-mediated apoptosis by triggering the translocation of Fas protein to the plasma membrane in a p53- and FAP-1-dependent manner. Similarly, the up-regulation of Fas-L by PEDF was also mediated by p53. Furthermore, peroxisome proliferator-activated receptor γ was determined to be the upstream regulator of p53. Together, these findings uncover a novel mechanism of tumor cell apoptosis induced by PEDF and provide a potential therapeutic strategy for tumors that are insensitive to Fas-L/Fas-dependent apoptosis because of a low level of cell surface Fas.
Collapse
Affiliation(s)
- Lei Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China,; Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Ya-Chao Yao
- Laboratory Center of Guangdong No. 2 Provincial People's Hospital, Guangzhou 510317, Guangdong Province, China
| | - Shu-Huan Fang
- DME Center, Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Cai-Qi Ma
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yi Cen
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zu-Min Xu
- Cancer Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524000, China
| | - Zhi-Yu Dai
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Cen Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Shuai Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ting Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hong-Hai Hong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei-Wei Qi
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ti Zhou
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Chao-Yang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China,.
| | - Xia Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China,; China Key Laboratory of Tropical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou 510080, China, and.
| | - Guo-Quan Gao
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China,; Key Laboratory of Functional Molecules from Marine Microorganisms, Sun Yat-sen University, Department of Education of Guangdong Province, Guangdong 510080, China.
| |
Collapse
|
21
|
Pigment epithelium-derived factor secreted from retinal pigment epithelium facilitates apoptotic cell death of iPSC. Sci Rep 2014; 3:2334. [PMID: 23903667 PMCID: PMC3730169 DOI: 10.1038/srep02334] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 07/05/2013] [Indexed: 01/10/2023] Open
Abstract
We show that pigment epithelium-derived factor (PEDF), which is secreted from primary or iPSC-derived retinal pigment epithelium (RPE), dramatically inhibits the growth of iPSCs. PEDF is detected abundantly in culture supernatants of primary or iPSC-derived RPE. Apoptotic cell death is induced in iPSC when co-cultured with RPE, a process that is significantly blocked by addition of antibody against PEDF. Indeed, addition of recombinant PEDF to the iPSC cell culture induces apoptotic cell death in iPSCs, but the expression of pluripotency related-genes is maintained, suggesting that PEDF causes cell death, not differentiation, of iPSCs. To recapitulate this event in vivo, we examined tumor formation in NOG mice after subcutaneous injection of iPSCs with or without an iPSC-derived RPE sheet (2.5 × 105 RPE cells). We observed that the tumor forming potential of iPSCs was significantly suppressed by simultaneous transplantation with an iPSC-derived RPE sheet.
Collapse
|
22
|
Yao Y, Li L, Huang X, Gu X, Xu Z, Zhang Y, Huang L, Li S, Dai Z, Li C, Zhou T, Cai W, Yang Z, Gao G, Yang X. SERPINA3K induces apoptosis in human colorectal cancer cells via activating the Fas/FasL/caspase-8 signaling pathway. FEBS J 2013; 280:3244-55. [PMID: 23615374 DOI: 10.1111/febs.12303] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 04/19/2013] [Accepted: 04/22/2013] [Indexed: 01/03/2023]
Abstract
SERPINA3K, also known as kallikrein-binding protein (KBP), is a serine proteinase inhibitor with anti-inflammatory and anti-angiogenic activities. Our previous studies showed that SERPINA3K inhibited proliferation in a dose-dependent manner and induced apoptosis of endothelial cells but had no influence on SGC-7901 gastric carcinoma cells or HepG2 hepatocarcinoma cells. However, it is unknown whether SERPINA3K has a direct impact on other carcinoma cells and which mechanisms are involved. In this study, we report for the first time that SERPINA3K not only decreased cell viability but also induced apoptosis in the colorectal carcinoma cell lines SW480 and HT-29. SERPINA3K-induced apoptosis of SW480 and HT-29 was rescued by interference with Fas ligand (FasL) small hairpin RNA. Moreover, SERPINA3K increased the expression of FasL and activated caspase-8. Peroxisome proliferator-activated receptor γ (PPARγ), a transcription factor of FasL, was also upregulated by SERPINA3K in a dose-dependent manner. The upregulation effect of FasL induced by SERPINA3K was reversed after interference with PPARγ small interfering RNA. These results demonstrated that SERPINA3K-induced SW480 and HT-29 cell apoptosis was mediated by the PPARγ/Fas/FasL signaling pathway. Therefore, our study provides additional insight into the direct anti-tumor function by inducing tumor cell apoptosis of SERPINA3K in colorectal tumors.
Collapse
Affiliation(s)
- Yachao Yao
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Craword SE, Fitchev P, Veliceasa D, Volpert OV. The many facets of PEDF in drug discovery and disease: a diamond in the rough or split personality disorder? Expert Opin Drug Discov 2013; 8:769-92. [PMID: 23642051 DOI: 10.1517/17460441.2013.794781] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Pigment epithelium-derived factor (PEDF) was discovered as a neurotrophic factor secreted by retinal pigment epithelial cells. A decade later, it re-emerged as a powerful angiogenesis inhibitor guarding ocular function. Since then, significant advances were made identifying PEDF's mechanisms, targets and biomedical applications. AREAS COVERED The authors review several methodologies that have generated significant new information about the potential of PEDF as a drug. Furthermore, the authors review and discuss mechanistic and structure-function analyses combined with the functional mapping of active fragments, which have yielded several short bioactive PEDF peptides. Additionally, the authors present functional studies in knockout animals and human correlates that have provided important information about conditions amenable to PEDF-based therapies. EXPERT OPINION Through its four known receptors, PEDF causes a wide range of cellular events vitally important for the organism, which include survival and differentiation, migration and invasion, lipid metabolism and stem cell maintenance. These processes are deregulated in multiple pathological conditions, including cancer, metabolic and cardiovascular disease. PEDF has been successfully used in countless preclinical models of these conditions and human correlates suggest a wide utility of PEDF-based drugs. The most significant clinical application of PEDF, to date, is its potential therapeutic use for age-related macular degeneration. Moreover, PEDF-based gene therapy has advanced to early stage clinical trials. PEDF active fragments have been mapped and used to design short peptide mimetics conferring distinct functions of PEDF, which may address specific clinical problems and become prototype drugs.
Collapse
Affiliation(s)
- Susan E Craword
- St. Louis University School of Medicine, Department of Pathology, St. Louis, Missouri, USA
| | | | | | | |
Collapse
|
24
|
Becerra SP, Notario V. The effects of PEDF on cancer biology: mechanisms of action and therapeutic potential. Nat Rev Cancer 2013; 13:258-71. [PMID: 23486238 PMCID: PMC3707632 DOI: 10.1038/nrc3484] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The potent actions of pigment epithelium-derived factor (PEDF) on tumour-associated cells, and its extracellular localization and secretion, stimulated research on this multifunctional serpin. Such studies have identified several PEDF receptors and downstream signalling pathways. Known cellular PEDF responses have expanded from the initial discovery that PEDF induces retinoblastoma cell differentiation to its anti-angiogenic, antitumorigenic and antimetastatic properties. Although the diversity of PEDF activities seems to be complex, they are consistent with the varied mechanisms that regulate this multimodal factor. If PEDF is to be used for cancer management, a deeper appreciation of its many functions and mechanisms of action is needed.
Collapse
Affiliation(s)
- S Patricia Becerra
- National Eye Institute, US National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|
25
|
Jia L, Waxman DJ. Thrombospondin-1 and pigment epithelium-derived factor enhance responsiveness of KM12 colon tumor to metronomic cyclophosphamide but have disparate effects on tumor metastasis. Cancer Lett 2012; 330:241-9. [PMID: 23228633 DOI: 10.1016/j.canlet.2012.11.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/30/2012] [Accepted: 11/30/2012] [Indexed: 12/18/2022]
Abstract
The anti-tumor activity, metronomic chemotherapy sensitization potential and metastatic effects of the endogenous angiogenesis inhibitors thrombospondin-1 and PEDF were investigated in KM12 colon adenocarcinoma xenografts. Thrombospondin-1 and PEDF decreased KM12 tumor microvessel density, increased macrophage infiltration, and improved responsiveness to metronomic cyclophosphamide (CPA) treatment, but did not activate the anti-tumor innate immunity that metronomic CPA induces in other tumor models. Moreover, thrombospondin-1, but not PEDF, significantly increased KM12 metastasis to the lung, while PEDF augmented the anti-metastatic activity of metronomic CPA. Thus, while thrombospondin-1 and PEDF both increase the KM12 tumor responsiveness to metronomic CPA, they have disparate effects on tumor metastasis.
Collapse
Affiliation(s)
- Li Jia
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, MA 02215, United States
| | | |
Collapse
|
26
|
Li L, Yang J, Wang WW, Yao YC, Fang SH, Dai ZY, Hong HH, Yang X, Shuai XT, Gao GQ. Pigment epithelium-derived factor gene loaded in cRGD–PEG–PEI suppresses colorectal cancer growth by targeting endothelial cells. Int J Pharm 2012; 438:1-10. [DOI: 10.1016/j.ijpharm.2012.08.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 07/30/2012] [Accepted: 08/21/2012] [Indexed: 01/19/2023]
|
27
|
Jang TJ, Kim SW, Lee KS. The expression of pigment epithelium-derived factor in bladder transitional cell carcinoma. KOREAN JOURNAL OF PATHOLOGY 2012; 46:261-5. [PMID: 23110012 PMCID: PMC3479768 DOI: 10.4132/koreanjpathol.2012.46.3.261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 06/05/2012] [Accepted: 06/05/2012] [Indexed: 11/29/2022]
Abstract
Background Pigment epithelium-derived factor (PEDF) is an anti-angiogenic factor. The purpose of this study is to examine the involvement of PEDF in the angiogenesis and biological behavior of bladder transitional cell carcinoma (TCC). Methods We examined the expression of PEDF in 99 bladder TCCs and ten non-neoplastic tissues, and evaluated microvessel density (MVD). Results The positive immunoreactivity for PEDF was seen in normal urothelium in 60% (6/10) and TCC in 13% (13/99). The PEDF expression had a significant correlation with MVD, i.e., a low MVD in 42% (5/12), a middle MVD in 11% (8/76) and a high MVD 0% (0/11) of tumors. The PEDF expression was not significantly correlated with the differentiation and invasion of TCC, but the degree of MVD was significantly higher in both high grade TCC and the pT2 tumors. Conclusions The degree of PEDF expression is significantly higher in normal bladder urothelium than bladder TCC; it is inversely correlated with the angiogenesis; and it is not related to the differentiation and progression of TCC. It can therefore be concluded that bladder TCC would initially occur if there is a lack of the PEDF expression.
Collapse
Affiliation(s)
- Tae Jung Jang
- Department of Pathology, Dongguk University College of Medicine, Gyeongju, Korea
| | | | | |
Collapse
|
28
|
Changes in the gene expression profile of A375 human melanoma cells induced by overexpression of multifunctional pigment epithelium-derived factor. Melanoma Res 2011; 21:285-97. [PMID: 21673604 DOI: 10.1097/cmr.0b013e32834495c3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a broad-spectrum angiogenesis inhibitor that displays potent antimetastatic activity in multiple tumor types. We have previously shown that PEDF prevents primary tumor growth and metastatic spread of human melanoma in mouse experimental models. Consistent with these observations, PEDF expression is lost at the late stages of melanoma progression, allowing melanoma cells to become angiogenic, migratory, and invasive. PEDF's ability to modify the interplay between the host and tumor tissues strongly supports its use as a therapeutic agent for the treatment of metastatic melanoma. However, transition to the clinic requires a more detailed knowledge of the molecular mechanisms underpinning PEDF's activity. In this study, we describe changes in the gene expression profile of A375 human melanoma cells induced by PEDF overexpression. PEDF modulated diverse categories of genes known to be involved in angiogenesis and migration. It downregulated cytokines such as interleukin-8 and extracellular matrix proteins such as collagen IV, while it upregulated fibronectin. Multiple transcripts previously described as contributing to the acquisition of malignant phenotype by melanoma were also diminished by PEDF overexpression, among which we validated galectin 3 and jagged 1. In addition, PEDF downregulated S100β and melanoma inhibitory activity, which are widely used in the pathological diagnosis of melanoma. Interestingly, PEDF increased the expression of melanophilin and decreased rab27A, which are relevant targets for melanosome transport; suggesting that PEDF could directly impinge on melanocytic lineage-specific processes. Our study identifies new molecular targets and signaling pathways that may potentially contribute to determine PEDF's ability to restrict the aggressiveness of A375 human melanoma cells.
Collapse
|
29
|
Broadhead ML, Dass CR, Choong PFM. Systemically administered PEDF against primary and secondary tumours in a clinically relevant osteosarcoma model. Br J Cancer 2011; 105:1503-11. [PMID: 21979423 PMCID: PMC3242526 DOI: 10.1038/bjc.2011.410] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background: Pigment epithelium-derived factor (PEDF) is an endogenous glycoprotein with a potential role as a therapeutic for osteosarcoma. Animal studies have demonstrated the biological effects of PEDF on osteosarcoma; however, these results are difficult to extrapolate for human use due to the chosen study design and drug delivery methods. Methods: In this study we have attempted to replicate the human presentation and treatment of osteosarcoma using a murine orthotopic model of osteosarcoma. The effects of PEDF on osteosarcoma cell lines were evaluated in vitro prior to animal experimentation. Orthotopic tumours were induced by intra-tibial injection of SaOS-2 osteosarcoma cells. Treatment with PEDF was delayed until after the macroscopic appearance of primary tumours. Pigment epithelium-derived factor was administered systemically via an implanted intraperitoneal micro-osmotic pump. Results: In vitro, PEDF inhibited proliferation, induced apoptosis and inhibited cell cycling of osteosarcoma cells. Pigment epithelium-derived factor promoted adhesion to Collagen I and inhibited invasion through Collagen I. In vivo, treatment with PEDF caused a reduction in both primary tumour volume and burden of pulmonary metastases. Systemic administration of PEDF did not cause toxic effects on normal tissues. Conclusion: Systemically delivered PEDF is effective in suppressing the size of primary and secondary tumours in an orthotopic murine model of osteosarcoma.
Collapse
Affiliation(s)
- M L Broadhead
- Department of Orthopaedics, St Vincent's Hospital Melbourne, Level 3, Daly Wing, 35 Victoria Pde, Fitzroy, VIC 3065, Australia
| | | | | |
Collapse
|
30
|
Schmitz JC, Protiva P, Gattu AK, Utsumi T, Iwakiri Y, Neto AG, Quinn M, Cornwell ML, Fitchev P, Lugea A, Crawford SE, Chung C. Pigment epithelium-derived factor regulates early pancreatic fibrotic responses and suppresses the profibrotic cytokine thrombospondin-1. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2990-9. [PMID: 21964188 DOI: 10.1016/j.ajpath.2011.08.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 08/01/2011] [Accepted: 08/10/2011] [Indexed: 01/09/2023]
Abstract
Pigment epithelium-derived factor (PEDF) is important for maintaining the normal extracellular matrix. We hypothesized that the initiation of pancreatic fibrosis is dependent on the loss of PEDF. Pancreatic PEDF expression was assessed in wild-type mice fed either a control or ethanol diet using an intragastric feeding model. Pancreatitis responses were elicited with either a single episode or a repetitive cerulein-induced (50 μg/kg, 6 hourly i.p. injections) protocol in wild-type and PEDF-null mice. Quantitative real-time PCR and immunoblotting were performed to assess fibrogenic responses. In wild-type animals, PEDF expression increased with pancreatitis and was more pronounced in mice fed ethanol. Compared with wild-type mice, α-smooth muscle actin staining and expression levels of fibrogenic markers (eg, transforming growth factor-β1, platelet-derived growth factor, collagen I, and thrombospondin-1) were higher in PEDF-null mice at baseline. Sirius red staining revealed more fibrosis in PEDF-null versus wild-type pancreas 1 week after pancreatitis. Differences in tissue fibrosis resolved with longer recovery periods. PEDF overexpression suppressed thrombospondin-1 levels in vitro. Ethanol feeding and experimental pancreatitis increased PEDF expression in wild-type mice. PEDF-null mice, however, demonstrated enhanced early fibrotic responses compared with wild-type mice with pancreatitis. These findings indicate that PEDF acts as a compensatory antifibrotic cytokine in pancreatitis.
Collapse
Affiliation(s)
- John C Schmitz
- Section of Digestive Diseases, VA Connecticut Healthcare System, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ho TC, Chen SL, Shih SC, Chang SJ, Yang SL, Hsieh JW, Cheng HC, Chen LJ, Tsao YP. Pigment epithelium-derived factor (PEDF) promotes tumor cell death by inducing macrophage membrane tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). J Biol Chem 2011; 286:35943-35954. [PMID: 21846721 DOI: 10.1074/jbc.m111.266064] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF) is an intrinsic anti-angiogenic factor and a potential anti-tumor agent. The tumoricidal mechanism of PEDF, however, has not been fully elucidated. Here we report that PEDF induces the apoptosis of TC-1 and SK-Hep-1 tumor cells when they are cocultured with bone marrow-derived macrophages (BMDMs). This macrophage-mediated tumor killing is prevented by blockage of TNF-related apoptosis-inducing ligand (TRAIL) following treatment with the soluble TRAIL receptor. PEDF also increases the amount of membrane-bound TRAIL on cultured mouse BMDMs and on macrophages surrounding subcutaneous tumors. PEDF-induced tumor killing and TRAIL induction are abrogated by peroxisome proliferator-activated receptor γ (PPARγ) antagonists or small interfering RNAs targeting PPARγ. PEDF also induces PPARγ in BMDMs. Furthermore, the activity of the TRAIL promoter in human macrophages is increased by PEDF stimulation. Chromatin immunoprecipitation and DNA pull-down assays confirmed that endogenous PPARγ binds to a functional PPAR-response element (PPRE) in the TRAIL promoter, and mutation of this PPRE abolishes the binding of the PPARγ-RXRα heterodimer. Also, PPARγ-dependent transactivation and PPARγ-RXRα binding to this PPRE are prevented by PPARγ antagonists. Our results provide a novel mechanism for the tumoricidal activity of PEDF, which involves tumor cell killing via PPARγ-mediated TRAIL induction in macrophages.
Collapse
Affiliation(s)
- Tsung-Chuan Ho
- Department of Medical Research, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Show-Li Chen
- Department of Microbiology, School of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Shou-Chuan Shih
- Department of Gastroenterology, Mackay Memorial Hospital, Taipei 104, Taiwan; Mackay Medicine, Nursing, and Management College, Taipei 112, Taiwan
| | - Shing-Jyh Chang
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei 104, Taiwan; Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Su-Lin Yang
- Centers for Disease Control, Taipei 115, Taiwan; Department of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan
| | - Jui-Wen Hsieh
- Mackay Medicine, Nursing, and Management College, Taipei 112, Taiwan; Department of Ophthalmology, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Huey-Chuan Cheng
- Department of Ophthalmology, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Lee-Jen Chen
- Mackay Medicine, Nursing, and Management College, Taipei 112, Taiwan; Department of Ophthalmology, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Yeou-Ping Tsao
- Department of Medical Research, Mackay Memorial Hospital, Taipei 104, Taiwan; Department of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan; Department of Ophthalmology, Mackay Memorial Hospital, Taipei 104, Taiwan.
| |
Collapse
|
32
|
Hirsch J, Johnson CL, Nelius T, Kennedy R, Riese WD, Filleur S. PEDF inhibits IL8 production in prostate cancer cells through PEDF receptor/phospholipase A2 and regulation of NFκB and PPARγ. Cytokine 2011; 55:202-10. [PMID: 21570865 DOI: 10.1016/j.cyto.2011.04.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 03/02/2011] [Accepted: 04/15/2011] [Indexed: 11/19/2022]
Abstract
Interleukin-8 (IL8/CXCL8) has been described as a key effector in prostate cancer progression and resistance to standard chemotherapeutic drugs. In the present study, we investigated the effect of the natural, angio-inhibitory and anti-tumoral Pigment Epithelium-Derived Factor (PEDF) on the expression of IL8 cytokine by prostate cancer cells. Using a cytokine antibody array and ELISA, in addition to IL8 quantitative RT PCR, we showed that PEDF inhibits the production of IL8 in human hormone-refractory prostate cancer cells, and delays the growth of these cells in vitro. IL8 reduction was mimicked in cancer cells treated with PPARγ agonist and NFκB-specific inhibitors. Accordingly, PPARγ expression increased in response to PEDF, whereas RelA/p65 expression and nuclear localization, and NFκB transcriptional activity decreased. NFκB deactivation was reversed by the PPARγ antagonist GW9662 and PPARγ (Leu(468)/Glu(471)) dominant negative suggesting a PPARγ-dependent process. We also investigated PEDF Receptor/PLA2 as key player in this pathway by small interference RNA. PEDFR knock down in prostate cancer cells reversed PEDF-induced PPARγ up-regulation, and NFκB and IL8 inhibition compared to non-targeting control siRNA. We conclude that by binding to PEDFR, PEDF up-regulates PPARγ, leading subsequently to suppressed NFκB-mediated transcriptional activation, reduced production of IL8 and limited proliferation of prostate cancer cells. These results reinforce PEDF's therapeutic potential and imply that blocking IL8 could represent a novel alternative for prostate cancer treatment.
Collapse
Affiliation(s)
- Jennifer Hirsch
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, TX 79430, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Feng CC, Ding Q, Zhang YF, Jiang HW, Wen H, Wang PH, Wu Z. Pigment epithelium-derived factor expression is down-regulated in bladder tumors and correlates with vascular endothelial growth factor and matrix metalloproteinase-9. Int Urol Nephrol 2010; 43:383-90. [PMID: 20848197 DOI: 10.1007/s11255-010-9834-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2010] [Accepted: 08/24/2010] [Indexed: 01/05/2023]
Abstract
Growth of solid tumor depends on angiogenesis, a process regulated by the balance of pro- and anti-angiogenic factors. We investigated the expression of anti-angiogenic factor pigment epithelium-derived factor (PEDF) and proangiogenic factors vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9) with immunohistochemistry in 64 bladder tumor samples and 23 normal controls. Compared with normal urothelium, we identified decreased PEDF expression (P = 0.000) whereas increased expression of VEGF (P = 0.000) and MMP-9 (P = 0.000) in tumorous tissue as well as in papillary urothelial neoplasm of low malignant potential (PUNLMP) (P = 0.009 and P = 0.000 accordingly) but MMP-9 (P = 0.704). Decreased PEDF expression was revealed with higher tumor grade (P = 0.014) but stage (P = 0.687). There was no age or gender preference in PEDF, VEGF or MMP-9 expression. Negative correlation of expression in tumorous and cancerous tissue regarding PEDF and VEGF (P = 0.000, r = -0.56, and P = 0.000, r = -0.50, respectively), PEDF and MMP-9 (P = 0.002, r = -0.39, and P = 0.032, r = -0.30, respectively) was identified. There was a negative correlation of expression between PEDF and VEGF (P = 0.016, r = -0.677) and no correlation between PEDF and MMP-9 (P = 0.147, r = -0.45) in PUNLMP. Decreased PEDF and increased VEGF and MMP-9 expression may play considerable roles in differentiation and invasion of bladder tumor.
Collapse
Affiliation(s)
- Chen-Chen Feng
- Department of Urology, Huashan Hospital, Fudan University, 12 Central Urumqi Rd, Shanghai 200040, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Pigment epithelium-derived factor (PEDF) is an endogenously produced glycoprotein with a spectrum of biological roles across diverse pathologies. Recent research has focused on the biochemical properties of PEDF and its associated receptors. This review discusses the recent developments in PEDF biochemistry and how this new knowledge will help progress our understanding of PEDF as a molecular mediator for anti-angiogenesis and -tumorigenesis. Additionally, pathophysiological roles for PEDF in healing and tissue homeostasis are being revealed and our enhanced understanding of the interactions between PEDF and its receptors may yet prove useful in propelling PEDF towards clinical application.
Collapse
Affiliation(s)
- Matthew L Broadhead
- Department of Orthopaedics, St Vincent's Hospital, University of Melbourne, Melbourne, Vic., Australia
| | | | | | | |
Collapse
|
35
|
Konson A, Pradeep S, Seger R. Phosphomimetic Mutants of Pigment Epithelium-Derived Factor with Enhanced Antiangiogenic Activity as Potent Anticancer Agents. Cancer Res 2010; 70:6247-57. [DOI: 10.1158/0008-5472.can-10-0434] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
Broadhead ML, Dass CR, Choong PFM. In vitro and in vivo biological activity of PEDF against a range of tumors. Expert Opin Ther Targets 2010; 13:1429-38. [PMID: 19922300 DOI: 10.1517/14728220903307475] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Pigment epithelium-derived factor (PEDF) is an emerging anti-cancer agent that targets both tumor tissue and its supporting vasculature. These direct and indirect effects of PEDF have been examined in vitro and in vivo for a range of malignancies. OBJECTIVE This review seeks to present PEDF as a potential anti-cancer agent with applications across multiple malignancies. We refer closely to experimental methodology whilst still highlighting the clinical significance of PEDF in cancer, drawing on biological findings in vitro and in vivo. METHODS A Pubmed database search was performed limiting the scope of this discussion paper mainly to PEDF's biological role in cancer, specifically lung, breast, prostatic, ovarian and pancreatic carcinomas, melanoma, glioma and osteosarcoma. CONCLUSIONS The biological roles of PEDF are diverse and multidimensional. As an anti-cancer agent, PEDF has great potential as a focused anti-neoplastic therapy against a variety of tumor types.
Collapse
Affiliation(s)
- Matthew L Broadhead
- University of Melbourne, St. Vincent's Hospital, Department of Orthopaedics and Surgery, L3, Daly Wing, 35 Victoria Pde., Fitzroy 3065, VIC, Australia
| | | | | |
Collapse
|
37
|
Cancer cell apoptotic pathways mediated by PEDF: prospects for therapy. Trends Mol Med 2009; 15:461-7. [DOI: 10.1016/j.molmed.2009.08.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 07/28/2009] [Accepted: 08/05/2009] [Indexed: 12/20/2022]
|
38
|
Yang H, Grossniklaus HE. Constitutive overexpression of pigment epithelium-derived factor inhibition of ocular melanoma growth and metastasis. Invest Ophthalmol Vis Sci 2009; 51:28-34. [PMID: 19661223 DOI: 10.1167/iovs.09-4138] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Pigment epithelium-derived factor (PEDF) is known to be an angiogenesis suppressor and to have antitumor effects. This study investigates whether constitutive overexpression of PEDF inhibits the growth and hepatic micrometastasis of ocular melanoma. METHODS Real-time RT-PCR was used to detect endogenous PEDF expression in human uveal melanoma cell lines and mouse melanoma cells. A lentiviral vector containing a mouse PEDF expression sequence was constructed and transduced into mouse melanoma cells in vitro. Transgene expression was assessed by Western blot analysis. Angiogenesis and transendothelial migration assays were performed in constitutively stable PEDF-overexpressing cells and transduced lentiviral vector control cells. The size and microvessel density of the ocular tumor and the number of hepatic micrometastasis were compared between the mice inoculated with PEDF-overexpressing tumor cells and those mice with the control cell line. RESULTS Four human uveal melanoma and three mouse melanoma cell lines were found to express PEDF mRNA. Endogenous overexpressing PEDF melanoma cells lost the ability to migrate and form tubes in vitro. In the animal experiment, the size of the ocular melanoma and the number of hepatic micrometastasis were decreased and microvessel density was also reduced in mice inoculated with constitutively overexpressing PEDF melanoma cells. CONCLUSIONS Lentivirus-mediated gene transfer of PEDF decreased the growth of ocular melanoma and its hepatic micrometastasis in a mouse ocular melanoma model. Dual antitumor/antiangiogenic activities of PEDF suggest that PEDF gene therapy may be considered an approach for the treatment of ocular melanoma.
Collapse
Affiliation(s)
- Hua Yang
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
39
|
Filleur S, Nelius T, de Riese W, Kennedy RC. Characterization of PEDF: a multi-functional serpin family protein. J Cell Biochem 2009; 106:769-75. [PMID: 19180572 DOI: 10.1002/jcb.22072] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a 50 kDa secreted glycoprotein that belongs to the non-inhibitory serpin family group. PEDF has been described as a natural angiogenesis inhibitor with neurotrophic and immune-modulation properties; it balances angiogenesis in the eye and blocks tumor progression. The mechanisms underlying most of these events are not completely clear; however, it appears that PEDF acts via multiple high affinity ligands and cell receptors. In this review article, we will summarize the current knowledge on the biochemical properties of PEDF and its receptors, the multimodal activities of PEDF and finally address the therapeutic potential of PEDF in treating angiogenesis-, neurodegeneration- and inflammation-related diseases.
Collapse
Affiliation(s)
- S Filleur
- Department of Urology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, Texas 79430-6591, USA.
| | | | | | | |
Collapse
|
40
|
Mirochnik Y, Aurora A, Schulze-Hoepfner FT, Deabes A, Shifrin V, Beckmann R, Polsky C, Volpert OV. Short pigment epithelial-derived factor-derived peptide inhibits angiogenesis and tumor growth. Clin Cancer Res 2009; 15:1655-63. [PMID: 19223494 DOI: 10.1158/1078-0432.ccr-08-2113] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Pigment epithelial-derived factor (PEDF) is a potent angiogenesis inhibitor with multiple other functions, some of which enhance tumor growth. Our previous studies mapped PEDF antiangiogenic and prosurvival activities to distinct epitopes. This study was aimed to determine the minimal fragment of PEDF, which maintains antiangiogenic and antitumor efficacy. EXPERIMENTAL DESIGN We analyzed antigenicity, hydrophilicity, and charge distribution of the angioinhibitory epitope (the 34-mer) and designed three peptides covering its COOH terminus, P14, P18, and P23. We analyzed their ability to block endothelial cell chemotaxis and induce apoptosis in vitro and their antiangiogenic activity in vivo. The selected peptide was tested for the antitumor activity against mildly aggressive xenografted prostate carcinoma and highly aggressive renal cell carcinoma. To verify that P18 acts in the same manner as PEDF, we used immunohistochemistry to measure PEDF targets, vascular endothelial growth factor receptor 2, and CD95 ligand expression in P18-treated vasculature. RESULTS P14 and P18 blocked endothelial cell chemotaxis; P18 and P23 induced apoptosis. P18 showed the highest IC50 and blocked angiogenesis in vivo: P23 was inactive and P14 was proangiogenic. P18 increased the production of CD95 ligand and reduced the expression of vascular endothelial growth factor receptor 2 by the endothelial cells in vivo. In tumor studies, P18 was more effective in blocking the angiogenesis and growth of the prostate cancer than parental 34-mer; in the renal cell carcinoma, P18 strongly decreased angiogenesis and halted the progression of established tumors. CONCLUSIONS P18 is a novel and potent antiangiogenic biotherapeutic agent that has potential to be developed for the treatment of prostate and renal cancer.
Collapse
Affiliation(s)
- Yelena Mirochnik
- Urology Department, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Liu W, Wu Z, Guan M, Lu Y. cDNA microarray analysis of pigment epithelium-derived factor-regulated gene expression profile in prostate carcinoma cells. Int J Urol 2009; 16:323-8. [PMID: 19207613 DOI: 10.1111/j.1442-2042.2008.02199.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVES To clarify molecular mechanisms involved in the action of pigment epithelium-derived factor (PEDF) in hormone insensitive prostate cancer cells. METHODS Total ribonucleic acid from untreated and PEDF-treated cells was subjected to microarray analysis using BioStar 8464 microarray. Real-time polymerase chain reaction analysis was conducted to confirm the microarray data. RESULTS Twenty-seven out of 8464 genes were found altered in both cell lines. Common gene responses altered by PEDF were identified and included genes known to alter cell signaling as well as genes involved in catalytic activity, cell proliferation, angiogenesis and apoptosis. Real-time reverse transcription polymerase chain reaction, in accordance with the microarray analysis, indicated that PEDF treatment caused an upregulation in the mRNA expression level of stanniocalcin 2, brain-specific angiogenesis inhibitor 2 and growth arrest, DNA-damage-inducible, alpha, and downregulation in the messenger ribonucleic acid level of fibroblast growth factor 3, teratocarcinoma-derived growth factor, neuropilin1, and endothelial Per/ARNT/Sim domain protein1, respectively. CONCLUSIONS These findings demonstrate that PEDF administration causes significant changes in the gene expression of the prostate, providing insights into the potential role of PEDF in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Weiwei Liu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
42
|
Abstract
Angiogenesis, the sprouting of new blood vessels from preexisting blood vessels, is a hallmark of glioma progression. Malignant gliomas are among the most lethal tumors with a very dismal prognosis, despite advances in standard therapy, including surgery, radiation, and chemotherapy. The median survival of patients with malignant gliomas has changed little in the last few years and is still measured in months. In an attempt to develop new therapeutic strategies and identify the molecular mechanism involved in glioma growth and progression, there has been extraordinary scientific interest in the past 2 decades in angiogenic responses associated with gliomas. This chapter focuses on the molecular mechanism of glioma angiogenesis and summarizes some of the therapeutic approaches based on antiangiogenesis.
Collapse
Affiliation(s)
- Marcia Machein
- Department of Neurosurgery, University of Freiburg Medical School, Breisacher Str. 64, Freiburg 79106, Germany.
| | | |
Collapse
|
43
|
Abstract
BACKGROUND Several studies of drugs that inhibit tumour angiogenesis have shown improvements in the survival of cancer patients, thus validating angiogenesis as a clinically relevant target. Both intracellular and extracellular approaches have shown promising results in clinical situations. OBJECTIVES To compare and contrast oligosaccharide therapies and other anti-angiogenic compounds for their benefits and toxicity. METHODS Analysis of the relevant literature including presentations at recent conferences. RESULTS Receptor tyrosine kinase inhibitors are orally available but have a broad spectrum of activity which is associated with toxicity. Antibodies are associated with different toxicities, however, they are administered parenterally. Oligosaccharides that act as competitive inhibitors of heparan sulfate (HS) are in the early and late phases of clinical development. The advantage of oligosaccharides should be that they can be designed to target several angiogenic molecules, that they are relatively safe and that they can be administered subcutaneously at home. The key questions concerning their development focus on whether compounds with sufficient affinity and relative specificity can be generated, whether they are active at doses that do not perturb the coagulation cascade to a clinically dangerous level, whether the synthetic routes are scalable and, whether the current Phase III trials will yield positive results. CONCLUSIONS Saccharides represent a novel and exciting therapeutic approach that targets a spectrum of angiogenic molecules that cannot be inhibited through established drug development programmes.
Collapse
Affiliation(s)
- Claire Louise Cole
- Translational Angiogenesis Group, Paterson Institute for Cancer Research, Wilmslow Road, Withington, Manchester M20 4BX, UK.
| | | |
Collapse
|
44
|
Ek ETH, Dass CR, Contreras KG, Choong PFM. PEDF-derived synthetic peptides exhibit antitumor activity in an orthotopic model of human osteosarcoma. J Orthop Res 2007; 25:1671-80. [PMID: 17600821 DOI: 10.1002/jor.20434] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Pigment epithelium-derived factor (PEDF) is one of the most potent inhibitors of angiogenesis, and has recently been demonstrated to have an important multifunctional role in tumor growth, invasion, and metastasis. However, relatively little is known of mechanisms through which PEDF exerts its antitumor activity. Therefore, with the aim of identifying potential functional epitopes specifically against osteosarcoma, we evaluated the bioactivity of four 25-mer synthetic PEDF-derived peptides (termed StVOrth-1, -2 -3, and -4) against a human osteosarcoma cell line, SaOS-2. We found that StVOrth-2 (residues 78-102) predominantly inhibited tumor cell proliferation, while StVOrth-3 (residues 90-114) markedly increased cellular adhesion to collagen type-1, with StVOrth-4 (residues 387-411) demonstrating most significant inhibition of Matrigel invasion. Furthermore, we show that StVOrth-1 (residues 40-64), -2 and -3 induce osteoblastic differentiation, evidenced by increased mineralized nodule formation. Interestingly, although no peptide inhibited angiogenesis in the tube formation assay, StVOrth-3 and -4 markedly suppressed VEGF expression. We further tested the activity of StVOrth-2 and StVOrth-3 in vivo, in an orthotopic model of osteosarcoma and found that both peptides significantly inhibited primary tumor growth and the development of pulmonary metastases. Together these results provide greater insight into the potential mechanisms through which PEDF exerts its antitumor function. Furthermore, this raises the possibility of developing short PEDF fragments as lead compounds for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Eugene T H Ek
- Department of Orthopaedics, University of Melbourne, St. Vincent's Hospital, Melbourne, P.O. Box 2900, Fitzroy, 3065, Melbourne, VIC, Australia
| | | | | | | |
Collapse
|
45
|
Pina AL, Kubitza M, Brawanski A, Tombran-Tink J, Kloth S. Expression of pigment-epithelium-derived factor during kidney development and aging. Cell Tissue Res 2007; 329:329-38. [PMID: 17497179 DOI: 10.1007/s00441-007-0420-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Accepted: 03/29/2007] [Indexed: 10/23/2022]
Abstract
Inhibitors and stimulators of endothelial cell growth are essential for the coordination of blood vessel formation during organ growth and development. In the adult kidney, one of the major inhibitors of angiogenesis is pigment-epithelium-derived factor (PEDF). We have analyzed the expression and distribution of PEDF during various stages of renal development and aging with particular emphasis on the formation of functional glomeruli. We show that PEDF gene expression and protein levels in the kidney significantly increase with age. We have detected PEDF in the mesenchyme and endothelial cells at all developmental stages studied, in all regions of the nephrogenic zone in which the formation of new blood vessels is associated with the development of nephrons and collecting ducts, and in mature podocytes in the adult kidney. Our results are the first to suggest that PEDF is important in early renal postnatal development, that it could be relevant to the maturation of glomerular function and the filtration barrier formed by these cells, and that it may serve as an anti-angiogenic modulator during kidney development.
Collapse
Affiliation(s)
- Ana Luisa Pina
- Department of Neurosurgery, University Clinic of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany.
| | | | | | | | | |
Collapse
|
46
|
Ek ETH, Dass CR, Contreras KG, Choong PFM. Pigment epithelium-derived factor overexpression inhibits orthotopic osteosarcoma growth, angiogenesis and metastasis. Cancer Gene Ther 2007; 14:616-26. [PMID: 17479108 DOI: 10.1038/sj.cgt.7701044] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Despite significant improvements, the current management of primary osteosarcoma is still limited by the development of metastatic disease, which occurs in approximately 30% of patients despite aggressive multiagent chemotherapy and tumor-ablative surgery. Therefore, there is a need for the development of novel agents to improve the outcome of these patients. Pigment epithelium-derived factor (PEDF) has been shown to be one of the most potent inhibitors of angiogenesis, and more recently has demonstrated a functional role in tumor growth, invasion and metastasis. In this study we report, for the first time, the multitargeted role of PEDF in the inhibition of growth, angiogenesis and metastasis of two orthotopic models of osteosarcoma (rat UMR 106-01 and human SaOS-2). Through stable plasmid-mediated gene transfer of full-length human PEDF, we show that PEDF overexpression significantly reduced tumor cell proliferation (P<0.05) and Matrigel invasion (UMR(PEDF), P<0.001; SaOS(PEDF), P<0.05) and increased adhesion to collagen type-1 (P<0.01), in vitro. In vivo, PEDF overexpression dramatically suppressed orthotopic osteosarcoma growth (P<0.05) and the development of spontaneous pulmonary metastases (UMR(PEDF), P<0.05; SaOS(PEDF), P<0.001). Furthermore, PEDF-overexpressing tumors exhibited reduced intratumoral angiogenesis, evidenced by a significant decrease in microvessel density (P<0.05). Therefore, together these results suggest that PEDF may be a new and promising approach for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- E T H Ek
- Department of Orthopaedics, University of Melbourne, St Vincent's Hospital, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
47
|
Ek ETH, Dass CR, Contreras KG, Choong PFM. Inhibition of orthotopic osteosarcoma growth and metastasis by multitargeted antitumor activities of pigment epithelium-derived factor. Clin Exp Metastasis 2007; 24:93-106. [PMID: 17458711 DOI: 10.1007/s10585-007-9062-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2006] [Accepted: 02/16/2007] [Indexed: 10/23/2022]
Abstract
Osteosarcoma is major cause of cancer-related death in the pediatric age group, and this is due to the development of pulmonary metastases that fail to be eradicated with current treatment regimes. Although there have been significant improvements in the long-term survival of such patients, 25-50% with initially non-metastatic disease, subsequently develop metastases and this remains the major cause of death for these patients. In this study, we report the multimodal activity of pigment epithelium-derived factor (PEDF) in inhibiting osteosarcoma growth, angiogenesis and metastasis. In vitro, we found that administration of recombinant PEDF (rPEDF) on two osteosarcoma cell lines (rat UMR 106-01 and human SaOS-2) significantly reduced tumor cell proliferation and increased apoptosis, as well as decreased cell invasion, angiogenesis, and increased adhesion to collagen type-1. Administration of rPEDF upregulated the mRNA expression of phenotypic osteoblast differentiation markers (ALP, pro-alpha(1) collagen and osteocalcin) in a pre-osteoblastic cell line, UMR 201, and also increased mineralized nodule formation in both UMR 106-01 and SaOS-2. In vivo, rPEDF dramatically suppressed primary osteosarcoma growth and the development of macroscopic pulmonary metastases in an orthotopic model of human osteosarcoma (SaOS-2). Interestingly, no activity was seen in tumors grown subcutaneously, suggesting a paracrine interaction between PEDF and the bone microenvironment. Preliminary pharmacoevaluation studies demonstrated rPEDF stability within media containing serum and osteosarcoma cells, and no gross systemic toxicity was observed in vivo with rPEDF administration. These results suggest that PEDF is emerging as an attractive and clinically appealing drug candidate for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Eugene T H Ek
- Department of Orthopaedics, University of Melbourne, St. Vincent's Hospital, P.O. Box 2900, Fitzroy, Melbourne, 3065 VIC, Australia
| | | | | | | |
Collapse
|
48
|
Abstract
Since the relationship between angiogenesis and tumor growth was established by Folkman in 1971, scientists have made efforts exploring the possibilities in treating cancer by targeting angiogenesis. Inhibition of angiogenesis growth factors and administration of angiogenesis inhibitors are the basics of anti-angiogenesis therapy. Transfer of anti-angiogenesis genes has received attention recently not only because of the advancement of recombinant vectors, but also because of the localized and sustained expression of therapeutic gene product inside the tumor after gene transfer. This review provides the up-to-date information about the strategies and the vectors studied in the field of anti-angiogenesis cancer gene therapy.
Collapse
Affiliation(s)
- Ching-Chiu Liu
- Institute of Molecular Technology for Drug Discovery and Synthesis, Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | | | | |
Collapse
|
49
|
Fernandez-Garcia NI, Volpert OV, Jimenez B. Pigment epithelium-derived factor as a multifunctional antitumor factor. J Mol Med (Berl) 2006; 85:15-22. [PMID: 17106733 DOI: 10.1007/s00109-006-0111-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 07/14/2006] [Accepted: 08/11/2006] [Indexed: 01/03/2023]
Abstract
The design of new therapeutic strategies for cancer treatment is based on the combination of drugs directed against different tumor compartments, including the tumor cells themselves and components of the stroma, such as the tumor vasculature. Indeed, several antiangiogenic compounds have entered clinical trials for use alone or in combination with conventional cytotoxic drugs. Pigment epithelium-derived factor (PEDF) is a multifunctional natural peptide with complex neurotrophic, neuroprotective, antiangiogenic, and proapoptotic biological activities, any of which could potentially be exploited for therapeutic purposes. This review summarizes recent studies that reveal the antitumor potential of PEDF based on its antiangiogenic properties and its newly discovered direct antitumor effects, which involve the induction of differentiation or apoptosis in tumor cells. We also discuss possible therapeutic applications of PEDF, based on these mechanistic insights and on the identification of functional domains that retain specific biological activities.
Collapse
Affiliation(s)
- N I Fernandez-Garcia
- Departamento de Bioquimica, Facultad de Medicina, Instituto de Investigaciones Biomedicas CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain
| | | | | |
Collapse
|
50
|
Notari L, Baladron V, Aroca-Aguilar JD, Balko N, Heredia R, Meyer C, Notario PM, Saravanamuthu S, Nueda ML, Sanchez-Sanchez F, Escribano J, Laborda J, Becerra SP. Identification of a lipase-linked cell membrane receptor for pigment epithelium-derived factor. J Biol Chem 2006; 281:38022-37. [PMID: 17032652 DOI: 10.1074/jbc.m600353200] [Citation(s) in RCA: 222] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF) is an extracellular multifunctional protein belonging to the serpin superfamily with demonstrable neurotrophic, gliastatic, neuronotrophic, antiangiogenic, and antitumorigenic properties. We have previously provided biochemical evidence for high affinity PEDF-binding sites and proteins in plasma membranes of retina, retinoblastoma, and CNS cells. This study was designed to reveal a receptor involved in the biological activities of PEDF. Using a yeast two-hybrid screening, we identified a novel gene from pigment epithelium of the human retina that codes for a PEDF-binding partner, which we term PEDF-R. The derived polypeptide has putative transmembrane, intracellular and extracellular regions, and a phospholipase domain. Recently, PEDF-R (TTS-2.2/independent phospholipase A(2) (PLA(2))zeta and mouse desnutrin/ATGL) has been described in adipose cells as a member of the new calcium-independent PLA(2)/nutrin/patatin-like phospholipase domain-containing 2 (PNPLA2) family that possesses triglyceride lipase and acylglycerol transacylase activities. Here we describe the PEDF-R gene expression in the retina and its heterologous expression by bacterial and eukaryotic systems, and we demonstrate that its protein product has specific and high binding affinity for PEDF, has a potent phospholipase A(2) activity that liberates fatty acids, and is associated with eukaryotic cell membranes. Most importantly, PEDF binding stimulates the enzymatic phospholipase A(2) activity of PEDF-R. In conclusion, we have identified a novel PEDF-R gene in the retina for a phospholipase-linked membrane protein with high affinity for PEDF, suggesting a molecular pathway by which ligand/receptor interaction on the cell surface could generate a cellular signal.
Collapse
Affiliation(s)
- Luigi Notari
- National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|