1
|
Sun R, Chen Y, Pei Y, Wang W, Zhu Z, Zheng Z, Yang L, Sun L. The drug release of PLGA-based nanoparticles and their application in treatment of gastrointestinal cancers. Heliyon 2024; 10:e38165. [PMID: 39364250 PMCID: PMC11447355 DOI: 10.1016/j.heliyon.2024.e38165] [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: 06/03/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
The poly (lactic-co-glycolic acid) (PLGA) based nanoparticles have been applied for drug delivery due to their simple preparation, biodegradability, and ideal biocompatibility. In this study, the factors affecting the degradation of PLGA-based nanoparticles are reviewed, encompassing the ratio of PLA to PGA, relative molecular weight, crystallinity, and preparation process of PLGA nanoparticles. The drug release behavior of PLGA-based nanoparticles, such as the degradation environment, encapsulated drug properties of polymers, and drug loading rates, are also discussed. Since gastrointestinal cancer is one of the major global threats to human health, this paper comprehensively summarizes the application of PLGA nanoparticles in gastrointestinal cancers from diagnosis, chemotherapy, radiotherapy, and novel tumor treatment methods (immunotherapy, gene therapy, and photothermal therapy). Finally, the future application of PLGA-based drug delivery systems in treating gastrointestinal cancers is discussed. The bottleneck of application status and the prospect of PLGA-nanoparticles in gastrointestinal tumor application are presented. To truly realize the great and wide application of PLGA-based nanoparticles, collaborative progress in the field of nanomaterials and life sciences is needed.
Collapse
Affiliation(s)
- Rui Sun
- Department of Digestive Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, PR China
| | - Yanfei Chen
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, PR China
| | - Yanjiang Pei
- Department of Digestive Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, PR China
| | - Wenbin Wang
- Department of Digestive Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, PR China
| | - Zhi Zhu
- Department of Digestive Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, PR China
| | - Zhaohua Zheng
- Department of Digestive Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, PR China
| | - Limeng Yang
- School of Textile Science & Engineering, Xi'an Polytechnic University, Xi'an, 710048, PR China
| | - Li Sun
- Department of Digestive Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, PR China
| |
Collapse
|
2
|
Kim G, Chen Z, Li J, Luo J, Castro-Martinez F, Wisniewski J, Cui K, Wang Y, Sun J, Ren X, Crawford SE, Becerra SP, Zhu J, Liu T, Wang S, Zhao K, Wu C. Gut-liver axis calibrates intestinal stem cell fitness. Cell 2024; 187:914-930.e20. [PMID: 38280375 PMCID: PMC10923069 DOI: 10.1016/j.cell.2024.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 10/25/2023] [Accepted: 01/02/2024] [Indexed: 01/29/2024]
Abstract
The gut and liver are recognized to mutually communicate through the biliary tract, portal vein, and systemic circulation. However, it remains unclear how this gut-liver axis regulates intestinal physiology. Through hepatectomy and transcriptomic and proteomic profiling, we identified pigment epithelium-derived factor (PEDF), a liver-derived soluble Wnt inhibitor, which restrains intestinal stem cell (ISC) hyperproliferation to maintain gut homeostasis by suppressing the Wnt/β-catenin signaling pathway. Furthermore, we found that microbial danger signals resulting from intestinal inflammation can be sensed by the liver, leading to the repression of PEDF production through peroxisome proliferator-activated receptor-α (PPARα). This repression liberates ISC proliferation to accelerate tissue repair in the gut. Additionally, treating mice with fenofibrate, a clinical PPARα agonist used for hypolipidemia, enhances colitis susceptibility due to PEDF activity. Therefore, we have identified a distinct role for PEDF in calibrating ISC expansion for intestinal homeostasis through reciprocal interactions between the gut and liver.
Collapse
Affiliation(s)
- Girak Kim
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jian Li
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialie Luo
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Felipe Castro-Martinez
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jan Wisniewski
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kairong Cui
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yan Wang
- Mass Spectrometry Facility, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialei Sun
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaobai Ren
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Susan E Crawford
- Department of Surgery, North Shore University Research Institute, University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - S Patricia Becerra
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jimin Zhu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Sui Wang
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Keji Zhao
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Elmi M, Dass JH, Dass CR. The Various Roles of PEDF in Cancer. Cancers (Basel) 2024; 16:510. [PMID: 38339261 PMCID: PMC10854708 DOI: 10.3390/cancers16030510] [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: 12/05/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Pigment epithelium-derived factor (PEDF) is a natural immunomodulator, anti-inflammatory, anti-angiogenic, anti-tumour growth and anti-metastasis factor, which can enhance tumour response to PEDF but can also conversely have pro-cancerous effects. Inflammation is a major cause of cancer, and it has been proven that PEDF has anti-inflammatory properties. PEDF's functional activity can be investigated through measuring metastatic and metabolic biomarkers that will be discussed in this review.
Collapse
Affiliation(s)
- Mitra Elmi
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| | - Joshua H. Dass
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| | - Crispin R. Dass
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| |
Collapse
|
4
|
Jones IC, Carnagarin R, Armstrong J, Lin DPL, Baxter-Holland M, Elahy M, Dass CR. Pigment Epithelium-Derived Factor: Inhibition of Phosphorylation of Insulin Receptor (IR)/IR Substrate (IRS), Osteogeneration from Adipocytes, and Increased Levels Due to Doxorubicin Exposure. Pharmaceutics 2023; 15:1960. [PMID: 37514146 PMCID: PMC10384968 DOI: 10.3390/pharmaceutics15071960] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVES Pigment epithelium-derived factor (PEDF) has been recently linked to insulin resistance and is capable of differentiating myocytes to bone. We examined in more detail the intricate signalling of the insulin pathway influenced by PEDF in skeletal myocytes. We tested whether this serpin is also capable of generating de novo bone from adipocytes in vitro and in vivo, and how the anticancer drug doxorubicin links with PEDF and cellular metabolism. METHODS AND KEY FINDINGS We demonstrate that PEDF can inhibit phosphorylation of insulin receptor (IR) and insulin receptor substrate (IRS) in skeletal myocytes. PEDF constitutively activates p42/44 MAPK/Erk, but paradoxically does not affect mitogenic signalling. PEDF did not perturb either mitochondrial activity or proliferation in cells representing mesenchymal stem cells, cardiomyocytes, and skeletal myocytes and adipocytes. PEDF induced transdifferentiation of adipocytes to osteoblasts, promoting bone formation in cultured adipocytes in vitro and gelfoam fatpad implants in vivo. Bone formation in white adipose tissue (WAT) was better than in brown adipose tissue (BAT). The frontline anticancer drug doxorubicin increased levels of PEDF in a human breast cancer cell line, mirroring the in vivo finding where cardiac muscle tissue was stained increasingly for PEDF as the dose of doxorubicin increased in mice. PEDF also increased levels of reactive oxygen species (ROS) and glutathione (GSH) in the breast cancer cell line. CONCLUSIONS PEDF may be used to regenerate bone from adipose tissue in cases of trauma such as fractures or bone cancers. The increased presence of PEDF in doxorubicin-treated tumour cells need further exploration, and could be useful therapeutically in future. The safety of PEDF administration in vivo was further demonstrated in this study.
Collapse
Affiliation(s)
- Isobel C Jones
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
- School of Medicine, University of Notre Dame, Fremantle, WA 6160, Australia
| | - Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine-Royal Perth Hospital Unit, Faculty of Medicine, Dentistry & Health Sciences, University of Western Australia, Perth, WA 6009, Australia
- School of Pharmacy, Curtin University, Bentley, WA 6102, Australia
| | - Jo Armstrong
- School of Pharmacy, Curtin University, Bentley, WA 6102, Australia
| | - Daphne P L Lin
- School of Pharmacy, Curtin University, Bentley, WA 6102, Australia
| | - Mia Baxter-Holland
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Mina Elahy
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, WA 6102, Australia
- School of Medical Sciences, University of New South Wales, Kensington, NSW 2052, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
- School of Pharmacy, Curtin University, Bentley, WA 6102, Australia
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, WA 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
| |
Collapse
|
5
|
Brook N, Gill J, Chih H, Francis K, Dharmarajan A, Chan A, Dass CR. Pigment epithelium-derived factor downregulation in oestrogen receptor positive breast cancer bone metastases is associated with menopause. Mol Cell Endocrinol 2023; 559:111792. [PMID: 36309204 DOI: 10.1016/j.mce.2022.111792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/14/2022] [Accepted: 10/03/2022] [Indexed: 11/11/2022]
Abstract
Pigment epithelium-derived factor (PEDF) has a critical role in bone development and anti-tumour function in breast cancer (BC). As the expression and role of PEDF in BC bone metastases is unknown, we aimed to characterise PEDF in primary and metastatic BC. Subcellular PEDF localisation was semi-quantitatively analysed via immunohistochemistry in patient-matched, archived formalin-fixed paraffin-embedded primary BC and liver, lung, and decalcified bone metastases specimens. PEDF localisation was evaluated in 23 metastatic BC patients diagnosed with ER+, human epidermal growth factor receptor-2 (HER2) negative BC or TNBC. Cytoplasmic (p = 0.019) and membrane (p = 0.048) PEDF was lower in bone metastases compared to primary ER+/HER2- BC. In contrast, nuclear PEDF scores were higher in metastases compared to primary TNBC (p = 0.027), and increased membrane PEDF in metastatic tissue had improved disease-free interval (p = 0.016). Nuclear PEDF was decreased in bone metastases compared to primary ER+//HER2- BC in post-menopausal patients (p = 0.029). These novel findings indicate PEDF plays a role in clinical BC metastasis. Significantly lower PEDF levels in the post-menopausal compared to pre-menopausal setting suggests future PEDF research may have greater clinical importance in the post-menopausal ER+/HER2- BC population.
Collapse
Affiliation(s)
- Naomi Brook
- Curtin Medical School, Curtin University, Bentley, 6102, Australia; Curtin Health Innovation Research Institute, Bentley, 6102, Australia
| | - Jespal Gill
- Pathwest, Fiona Stanley Hospital, Murdoch, Australia
| | - HuiJun Chih
- Curtin School of Population Health, Curtin University, Bentley, 6102, Australia
| | - Kate Francis
- Western Diagnostic Pathology, Jandakot, 6164, Australia
| | - Arun Dharmarajan
- Curtin Medical School, Curtin University, Bentley, 6102, Australia; Curtin Health Innovation Research Institute, Bentley, 6102, Australia; Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, 600116, India
| | - Arlene Chan
- Curtin Medical School, Curtin University, Bentley, 6102, Australia; Breast Cancer Research Centre-Western Australia, Hollywood Private Hospital, Nedlands, 6009, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley, 6102, Australia; Curtin Health Innovation Research Institute, Bentley, 6102, Australia.
| |
Collapse
|
6
|
Lv F, Cai X, Ji L. An Update on Animal Models of Osteogenesis Imperfecta. Calcif Tissue Int 2022; 111:345-366. [PMID: 35767009 DOI: 10.1007/s00223-022-00998-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 06/01/2022] [Indexed: 11/02/2022]
Abstract
Osteogenesis imperfecta (OI) is a heterogeneous disorder characterized by bone fragility, multiple fractures, bone deformity, and short stature. In recent years, the application of next generation sequencing has triggered the discovery of many new genetic causes for OI. Until now, more than 25 genetic causes of OI and closely related disorders have been identified. However, the mechanisms of many genes on skeletal fragility in OI are not entirely clear. Animal models of OI could help to understand the cellular, signaling, and metabolic mechanisms contributing to the disease, and how targeting these pathways can provide therapeutic targets. To date, a lot of animal models, mainly mice and zebrafish, have been described with defects in 19 OI-associated genes. In this review, we summarize the known genetic causes and animal models that recapitulate OI with a main focus on engineered mouse and zebrafish models. Additionally, we briefly discuss domestic animals with naturally occurring OI phenotypes. Knowledge of the specific molecular basis of OI will advance clinical diagnosis and potentially stimulate targeted therapeutic approaches.
Collapse
Affiliation(s)
- Fang Lv
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Xizhimen South Street No.11, Beijing, 100044, China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Xizhimen South Street No.11, Beijing, 100044, China.
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Xizhimen South Street No.11, Beijing, 100044, China.
| |
Collapse
|
7
|
Lee WJ, Lin KH, Wang JS, Sheu WHH, Shen CC, Yang CN, Wu SM, Shen LW, Lee SH, Lai DW, Lan KL, Tung CW, Liu SH, Sheu ML. Aryl hydrocarbon receptor deficiency augments dysregulated microangiogenesis and diabetic retinopathy. Biomed Pharmacother 2022; 155:113725. [PMID: 36152407 DOI: 10.1016/j.biopha.2022.113725] [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: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022] Open
Abstract
Diabetic retinopathy (DR) is a pathophysiologic vasculopathic process with obscure mechanisms and limited effective therapeutic strategies. Aryl hydrocarbon receptor (AhR) is an important regulator of xenobiotic metabolism and an environmental sensor. The aim of the present study was to investigate the role of AhR in the development of DR and elucidate the molecular mechanism of its downregulation. DR was evaluated in diabetes-induced retinal injury in wild type and AhR knockout (AhR-/-) mice. Retinal expression of AhR was determined in human donor and mice eyes by immunofluorescence since AhR activity was examined in diabetes. AhR knockout (AhRKO) mice were used to induce diabetes with streptozotocin, high-fat diet, or genetic double knockout with diabetes spontaneous mutation (Leprdb) (DKO; AhR-/-×Leprdb/db) for investigating structural, functional, and metabolic abnormalities in vascular and epithelial retina. Structural molecular docking simulation was used to survey the pharmacologic AhR agonists targeting phosphorylated AhR (Tyr245). Compared to diabetic control mice, diabetic AhRKO mice had aggravated alterations in retinal vasculature that amplified hallmark features of DR like vasopermeability, vascular leakage, inflammation, blood-retinal barrier breakdown, capillary degeneration, and neovascularization. AhR agonists effectively inhibited inflammasome formation and promoted AhR activity in human retinal microvascular endothelial cells and pigment epithelial cells. AhR activity and protein expression was downregulated, resulting in a decrease in DNA promoter binding site of pigment epithelium-derived factor (PEDF) by gene regulation in transcriptional cascade. This was reversed by AhR agonists. Our study identified a novel of DR model that target the protective AhR/PEDF axis can potentially maintain retinal vascular homeostasis, providing opportunities to delay the development of DR.
Collapse
Affiliation(s)
- Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taiwan
| | - Keng-Hung Lin
- Department of Ophthalmology, Taichung Veterans General Hospital, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Jun-Sing Wang
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan; Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taiwan; Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Wayne Huey-Herng Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taipei Veterans General Hospital, Taiwan
| | - Chin-Chang Shen
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Sheng-Mao Wu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Li-Wei Shen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Hua Lee
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - De-Wei Lai
- Experimental Animal Center, Department of Molecular Biology and Cell Research, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Keng-Li Lan
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Meei-Ling Sheu
- Department of Medical Research, Taichung Veterans General Hospital, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan; Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
8
|
Ueno S, Sudo T, Saya H, Sugihara E. Pigment epithelium-derived factor promotes peritoneal dissemination of ovarian cancer through induction of immunosuppressive macrophages. Commun Biol 2022; 5:904. [PMID: 36056141 PMCID: PMC9440245 DOI: 10.1038/s42003-022-03837-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Peritoneal dissemination of ovarian cancer (OC) correlates with poor prognosis, but the mechanisms underlying the escape of OC cells from the intraperitoneal immune system have remained unknown. We here identify pigment epithelium–derived factor (PEDF) as a promoting factor of OC dissemination, which functions through induction of CD206+ Interleukin-10 (IL-10)–producing macrophages. High PEDF gene expression in tumors is associated with poor prognosis in OC patients. Concentrations of PEDF in ascites and serum are significantly higher in OC patients than those with more benign tumors and correlated with early recurrence of OC patients, suggesting that PEDF might serve as a prognostic biomarker. Bromodomain and extraterminal (BET) inhibitors reduce PEDF expression and limit both OC cell survival and CD206+ macrophage induction in the peritoneal cavity. Our results thus implicate PEDF as a driver of OC dissemination and identify a BET protein–PEDF–IL-10 axis as a promising therapeutic target for OC. Endogenously expressed pigment epithelium–derived factor (PEDF) promotes increased survival of ovarian cancer cells in the peritoneal cavity by inducing IL-10 expression in CD206 + peritoneal macrophages.
Collapse
Affiliation(s)
- Sayaka Ueno
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan.,Section of Translational Research, Hyogo Cancer Center, Hyogo, Japan
| | - Tamotsu Sudo
- Section of Translational Research, Hyogo Cancer Center, Hyogo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan. .,Division of Gene Regulation, Cancer Center, Research Promotion Headquarters, Fujita Health University School of Medicine, Aichi, Japan.
| | - Eiji Sugihara
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan. .,Division of Gene Regulation, Cancer Center, Research Promotion Headquarters, Fujita Health University School of Medicine, Aichi, Japan.
| |
Collapse
|
9
|
Teal CJ, Hettiaratchi MH, Ho MT, Ortin-Martinez A, Ganesh AN, Pickering AJ, Golinski AW, Hackel BJ, Wallace VA, Shoichet MS. Directed Evolution Enables Simultaneous Controlled Release of Multiple Therapeutic Proteins from Biopolymer-Based Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202612. [PMID: 35790035 DOI: 10.1002/adma.202202612] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/28/2022] [Indexed: 06/15/2023]
Abstract
With the advent of increasingly complex combination strategies of biologics, independent control over their delivery is the key to their efficacy; however, current approaches are hindered by the limited independent tunability of their release rates. To overcome these limitations, directed evolution is used to engineer highly specific, low affinity affibody binding partners to multiple therapeutic proteins to independently control protein release rates. As a proof-of-concept, specific affibody binding partners for two proteins with broad therapeutic utility: insulin-like growth factor-1 (IGF-1) and pigment epithelium-derived factor (PEDF) are identified. Protein-affibody binding interactions specific to these target proteins with equilibrium dissociation constants (KD ) between 10-7 and 10-8 m are discovered. The affibodies are covalently bound to the backbone of crosslinked hydrogels using click chemistry, enabling sustained, independent, and simultaneous release of bioactive IGF-1 and PEDF over 7 days. The system is tested with C57BL/6J mice in vivo, and the affibody-controlled release of IGF-1 results in sustained activity when compared to bolus IGF-1 delivery. This work demonstrates a new, broadly applicable approach to tune the release of therapeutic proteins simultaneously and independently and thus the way for precise control over the delivery of multicomponent therapies is paved.
Collapse
Affiliation(s)
- Carter J Teal
- Institute of Biomedical Engineering, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Marian H Hettiaratchi
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
| | - Margaret T Ho
- Institute of Biomedical Engineering, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Arturo Ortin-Martinez
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
| | - Ahil N Ganesh
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
| | - Andrew J Pickering
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
| | - Alex W Golinski
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, 421 Washington Avenue Southeast, 356 Amundson Hall, Minneapolis, MN, 55455, USA
| | - Benjamin J Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, 421 Washington Avenue Southeast, 356 Amundson Hall, Minneapolis, MN, 55455, USA
| | - Valerie A Wallace
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 27 King's College Circle, Toronto, ON, M5S 1A1, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, 340 College Street, Toronto, ON, M5T 3A9, Canada
| | - Molly S Shoichet
- Institute of Biomedical Engineering, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, 340 College Street, Toronto, ON, M5T 3A9, Canada
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada
| |
Collapse
|
10
|
Kang H, Aryal AC S, Barnes AM, Martin A, David V, Crawford SE, Marini JC. Antagonism Between PEDF and TGF-β Contributes to Type VI Osteogenesis Imperfecta Bone and Vascular Pathogenesis. J Bone Miner Res 2022; 37:925-937. [PMID: 35258129 PMCID: PMC11152058 DOI: 10.1002/jbmr.4540] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 11/08/2022]
Abstract
Osteogenesis imperfecta (OI) is a heterogeneous genetic disorder of bone and connective tissue, also known as brittle bone disease. Null mutations in SERPINF1, which encodes pigment epithelium-derived factor (PEDF), cause severe type VI OI, characterized by accumulation of unmineralized osteoid and a fish-scale pattern of bone lamellae. Although the potent anti-angiogenic activity of PEDF has been extensively studied, the disease mechanism of type VI OI is not well understood. Using Serpinf1(-/-) mice and primary osteoblasts, we demonstrate that loss of PEDF delays osteoblast maturation as well as extracellular matrix (ECM) mineralization. Barium sulfate perfusion reveals significantly increased vessel density in the tibial periosteum of Serpinf1(-/-) mouse compared with wild-type littermates. The increased bone vascularization in Serpinf1(-/-) mice correlated with increased number of CD31(+)/Endomucin(+) endothelial cells, which are involved in the coupling angiogenesis and osteogenesis. Global transcriptome analysis by RNA-Seq of Serpinf1(-/-) mouse osteoblasts reveals osteogenesis and angiogenesis as the biological processes most impacted by loss of PEDF. Intriguingly, TGF-β signaling is activated in type VI OI cells, and Serpinf1(-/-) osteoblasts are more sensitive to TGF-β stimulation than wild-type osteoblasts. TGF-β stimulation and PEDF deficiency showed additive effects on transcription suppression of osteogenic markers and stimulation of pro-angiogenic factors. Furthermore, PEDF attenuated TGF-β-induced expression of pro-angiogenic factors. These data suggest that functional antagonism between PEDF and TGF-β pathways controls osteogenesis and bone vascularization and is implicated in type VI OI pathogenesis. This antagonism may be exploited in developing therapeutics for type VI OI utilizing PEDF and TGF-β antibody. © 2022 American Society for Bone and Mineral Research (ASBMR). This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Heeseog Kang
- Section on Heritable Disorders of Bone and Extracellular Matrix, NICHD, NIH, Bethesda, MD, USA
| | - Smriti Aryal AC
- Section on Heritable Disorders of Bone and Extracellular Matrix, NICHD, NIH, Bethesda, MD, USA
| | - Aileen M Barnes
- Section on Heritable Disorders of Bone and Extracellular Matrix, NICHD, NIH, Bethesda, MD, USA
| | - Aline Martin
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Valentin David
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Susan E Crawford
- Department of Surgery, NorthShore University HealthSystem Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, USA
| | - Joan C Marini
- Section on Heritable Disorders of Bone and Extracellular Matrix, NICHD, NIH, Bethesda, MD, USA
| |
Collapse
|
11
|
Zinc-binding proteins in stallion seminal plasma as potential sperm function regulators. ANNALS OF ANIMAL SCIENCE 2022. [DOI: 10.2478/aoas-2022-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Sperm functions may be influenced by seminal plasma (SP) proteins with affinity to zinc ions. The aim of the study was to isolate and characterise zinc-binding proteins (ZnBPs) from stallion SP using proteomic tools. Zinc-binding proteins were isolated from the SP of eight stallions by zinc-affinity chromatography. They were analysed in the SDS-PAGE system, and peptide extracts were prepared. Samples of ZnBPs isolated from stallion SP were injected onto the nLC-1000 nanoflow HPLC system coupled via a nano-electrospray ion source to the Orbitrap Elite FTMS mass spectrometer (Thermo Fisher Scientific). Raw MS data were analysed using MaxQuant software with label-free quantification (LFQ). Protein sequences were obtained from the UniProt database. Forty-seven proteins in ZnBPs were annotated in a gene ontology (GO) analysis. The LFQ intensity analysis of individual proteins revealed that ZnBPs comprised mainly clusterin (CLU, 27%), serin protease inhibitor F1 (SERPINF1, 13%), actin, cytoplasmic 1 (ACTB, 9%), nucleobindin 2 (NUCB2, 8%) and polymeric immunoglobulin receptor (PIGR, 6%). This is the first proteomic study to analyse ZnBPs in stallion SP. The present findings show that ZnBPs in stallion SP could play an important role in the regulation of sperm function.
Collapse
|
12
|
Alijaj N, Pavlovic B, Martel P, Rakauskas A, Cesson V, Saba K, Hermanns T, Oechslin P, Veit M, Provenzano M, Rüschoff JH, Brada MD, Rupp NJ, Poyet C, Derré L, Valerio M, Banzola I, Eberli D. Identification of Urine Biomarkers to Improve Eligibility for Prostate Biopsy and Detect High-Grade Prostate Cancer. Cancers (Basel) 2022; 14:1135. [PMID: 35267445 PMCID: PMC8909910 DOI: 10.3390/cancers14051135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 11/30/2022] Open
Abstract
PCa screening is based on the measurements of the serum prostate specific antigen (PSA) to select men with higher risks for tumors and, thus, eligible for prostate biopsy. However, PSA testing has a low specificity, leading to unnecessary biopsies in 50-75% of cases. Therefore, more specific screening opportunities are needed to reduce the number of biopsies performed on healthy men and patients with indolent tumors. Urine samples from 45 patients with elevated PSA were collected prior to prostate biopsy, a mass spectrometry (MS) screening was performed to identify novel biomarkers and the best candidates were validated by ELISA. The urine quantification of PEDF, HPX, CD99, CANX, FCER2, HRNR, and KRT13 showed superior performance compared to PSA. Additionally, the combination of two biomarkers and patient age resulted in an AUC of 0.8196 (PSA = 0.6020) and 0.7801 (PSA = 0.5690) in detecting healthy men and high-grade PCa, respectively. In this study, we identified and validated novel urine biomarkers for the screening of PCa, showing that an upfront urine test, based on quantitative biomarkers and patient age, is a feasible method to reduce the number of unnecessary prostate biopsies and detect both healthy men and clinically significant PCa.
Collapse
Affiliation(s)
- Nagjie Alijaj
- Department of Urology, University Hospital of Zürich and University of Zürich, 8006 Zürich, Switzerland; (N.A.); (B.P.)
| | - Blaz Pavlovic
- Department of Urology, University Hospital of Zürich and University of Zürich, 8006 Zürich, Switzerland; (N.A.); (B.P.)
| | - Paul Martel
- Department of Urology, Urology Research Unit and Urology Biobank, University Hospital of Lausanne, 1011 Lausanne, Switzerland; (P.M.); (A.R.); (V.C.); (L.D.); (M.V.)
| | - Arnas Rakauskas
- Department of Urology, Urology Research Unit and Urology Biobank, University Hospital of Lausanne, 1011 Lausanne, Switzerland; (P.M.); (A.R.); (V.C.); (L.D.); (M.V.)
| | - Valérie Cesson
- Department of Urology, Urology Research Unit and Urology Biobank, University Hospital of Lausanne, 1011 Lausanne, Switzerland; (P.M.); (A.R.); (V.C.); (L.D.); (M.V.)
| | - Karim Saba
- Department of Urology, University Hospital of Zürich, 8091 Zürich, Switzerland; (K.S.); (T.H.); (P.O.); (M.V.); (M.P.); (C.P.); (D.E.)
| | - Thomas Hermanns
- Department of Urology, University Hospital of Zürich, 8091 Zürich, Switzerland; (K.S.); (T.H.); (P.O.); (M.V.); (M.P.); (C.P.); (D.E.)
| | - Pascal Oechslin
- Department of Urology, University Hospital of Zürich, 8091 Zürich, Switzerland; (K.S.); (T.H.); (P.O.); (M.V.); (M.P.); (C.P.); (D.E.)
| | - Markus Veit
- Department of Urology, University Hospital of Zürich, 8091 Zürich, Switzerland; (K.S.); (T.H.); (P.O.); (M.V.); (M.P.); (C.P.); (D.E.)
| | - Maurizio Provenzano
- Department of Urology, University Hospital of Zürich, 8091 Zürich, Switzerland; (K.S.); (T.H.); (P.O.); (M.V.); (M.P.); (C.P.); (D.E.)
| | - Jan H. Rüschoff
- Department of Pathology and Molecular Pathology, University Hospital of Zürich, 8091 Zürich, Switzerland; (J.H.R.); (M.D.B.); (N.J.R.)
| | - Muriel D. Brada
- Department of Pathology and Molecular Pathology, University Hospital of Zürich, 8091 Zürich, Switzerland; (J.H.R.); (M.D.B.); (N.J.R.)
| | - Niels J. Rupp
- Department of Pathology and Molecular Pathology, University Hospital of Zürich, 8091 Zürich, Switzerland; (J.H.R.); (M.D.B.); (N.J.R.)
- Faculty of Medicine, University of Zürich, 8032 Zürich, Switzerland
| | - Cédric Poyet
- Department of Urology, University Hospital of Zürich, 8091 Zürich, Switzerland; (K.S.); (T.H.); (P.O.); (M.V.); (M.P.); (C.P.); (D.E.)
| | - Laurent Derré
- Department of Urology, Urology Research Unit and Urology Biobank, University Hospital of Lausanne, 1011 Lausanne, Switzerland; (P.M.); (A.R.); (V.C.); (L.D.); (M.V.)
| | - Massimo Valerio
- Department of Urology, Urology Research Unit and Urology Biobank, University Hospital of Lausanne, 1011 Lausanne, Switzerland; (P.M.); (A.R.); (V.C.); (L.D.); (M.V.)
| | - Irina Banzola
- Department of Urology, University Hospital of Zürich and University of Zürich, 8006 Zürich, Switzerland; (N.A.); (B.P.)
| | - Daniel Eberli
- Department of Urology, University Hospital of Zürich, 8091 Zürich, Switzerland; (K.S.); (T.H.); (P.O.); (M.V.); (M.P.); (C.P.); (D.E.)
| |
Collapse
|
13
|
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
|
14
|
Ko VH, Yu LJ, Secor JD, Pan A, Mitchell PD, Kishikawa H, Puder M. Deficiency in pigment epithelium-derived factor accelerates pulmonary growth and development in a compensatory lung growth model. FASEB J 2021; 35:e21850. [PMID: 34569654 DOI: 10.1096/fj.202002661rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 01/05/2023]
Abstract
Children with hypoplastic lung disease associated with congenital diaphragmatic hernia (CDH) continue to suffer significant morbidity and mortality secondary to progressive pulmonary disease. Recently published work from our lab demonstrated the potential of Roxadustat (FG-4592), a prolyl hydroxylase inhibitor, as a treatment for CDH-associated pulmonary hypoplasia. Treatment with Roxadustat led to significantly accelerated compensatory lung growth (CLG) through downregulation of pigment epithelium-derived factor (PEDF), an anti-angiogenic factor, rather than upregulation of vascular endothelial growth factor (VEGF). PEDF and its role in pulmonary development is a largely unexplored field. In this study, we sought to further evaluate the role of PEDF in accelerating CLG. PEDF-deficient mice demonstrated significantly increased lung volume, total lung capacity, and alveolarization compared to wild type controls following left pneumonectomy without increased VEGF expression. Furthermore, Roxadustat administration in PEDF-deficient mice did not further accelerate CLG. Human microvascular endothelial lung cells (HMVEC-L) and human pulmonary alveolar epithelial cells (HPAEC) similarly demonstrated decreased PEDF expression with Roxadustat administration. Additionally, downregulation of PEDF in Roxadustat-treated HMVEC-L and HPAEC, a previously unreported finding, speaks to the potential translatability of Roxadustat from small animal studies. Taken together, these findings further suggest that PEDF downregulation is the primary mechanism by which Roxadustat accelerates CLG. More importantly, these data highlight the critical role PEDF may have in pulmonary growth and development, a previously unexplored field.
Collapse
Affiliation(s)
- Victoria H Ko
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lumeng J Yu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jordan D Secor
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amy Pan
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul D Mitchell
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Hiroko Kishikawa
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark Puder
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Moghaddam S, Jalali A, O’Neill A, Murphy L, Gorman L, Reilly AM, Heffernan Á, Lynch T, Power R, O’Malley KJ, Taskèn KA, Berge V, Solhaug VA, Klocker H, Murphy TB, Watson RW. Integrating Serum Biomarkers into Prediction Models for Biochemical Recurrence Following Radical Prostatectomy. Cancers (Basel) 2021; 13:4162. [PMID: 34439316 PMCID: PMC8391749 DOI: 10.3390/cancers13164162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/10/2021] [Accepted: 08/14/2021] [Indexed: 12/13/2022] Open
Abstract
This study undertook to predict biochemical recurrence (BCR) in prostate cancer patients after radical prostatectomy using serum biomarkers and clinical features. Three radical prostatectomy cohorts were used to build and validate a model of clinical variables and serum biomarkers to predict BCR. The Cox proportional hazard model with stepwise selection technique was used to develop the model. Model evaluation was quantified by the AUC, calibration, and decision curve analysis. Cross-validation techniques were used to prevent overfitting in the Irish training cohort, and the Austrian and Norwegian independent cohorts were used as validation cohorts. The integration of serum biomarkers with the clinical variables (AUC = 0.695) improved significantly the predictive ability of BCR compared to the clinical variables (AUC = 0.604) or biomarkers alone (AUC = 0.573). This model was well calibrated and demonstrated a significant improvement in the predictive ability in the Austrian and Norwegian validation cohorts (AUC of 0.724 and 0.606), compared to the clinical model (AUC of 0.665 and 0.511). This study shows that the pre-operative biomarker PEDF can improve the accuracy of the clinical factors to predict BCR. This model can be employed prior to treatment and could improve clinical decision making, impacting on patients' outcomes and quality of life.
Collapse
Affiliation(s)
- Shirin Moghaddam
- School of Mathematical Sciences, University College Cork, T12XF62 Cork, Ireland
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Amirhossein Jalali
- School of Mathematical Sciences, University College Cork, T12XF62 Cork, Ireland
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Amanda O’Neill
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Lisa Murphy
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Laura Gorman
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Anne-Marie Reilly
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Áine Heffernan
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Thomas Lynch
- Department of Urology, Trinity College, St James Hospital, D08 W9RT Dublin 8, Ireland;
| | - Richard Power
- Department of Urology, Royal College of Surgeons in Ireland, Beaumont Hospital, D09V2N0 Dublin 9, Ireland;
| | - Kieran J. O’Malley
- Department of Urology, University College Dublin, Mater Misericordiae University Hospital, D07YH5R Dublin 7, Ireland;
| | - Kristin A. Taskèn
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (K.A.T.); (V.B.)
- Department of Tumor Biology, Oslo University Hospital, 0379 Oslo, Norway
| | - Viktor Berge
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (K.A.T.); (V.B.)
- Department of Urology, Oslo University Hospital, 0379 Oslo, Norway;
| | - Vivi-Ann Solhaug
- Department of Urology, Oslo University Hospital, 0379 Oslo, Norway;
| | - Helmut Klocker
- Department of Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - T. Brendan Murphy
- UCD School of Mathematics and Statistics, University College Dublin, D04V1W8 Dublin 4, Ireland;
| | - R. William Watson
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| |
Collapse
|
16
|
The Effect of Mesenchymal Stem Cell Secretome on Corneal Endothelial Cell Preservation in an Oxidative Injury Model. Cornea 2021; 39:1426-1430. [PMID: 32732699 DOI: 10.1097/ico.0000000000002442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE To describe a reproducible oxidative injury model in ex vivo porcine corneas and to investigate the effects of corneal mesenchymal stem cell (Co-MSC) secretome and specific factors on the preservation of corneal endothelium after oxidative injury. METHODS Porcine corneas underwent vital staining with trypan blue and alizarin red with different concentration and time points. Ex vivo porcine corneas were exposed (endothelial side) to varied concentrations of hydrogen peroxide. After injury, 3 groups of 5 corneas underwent treatment with secretome from either a wild-type (WT) murine Co-MSC, a pigment epithelium derived factor (PEDF) knock out (K/O) murine Co-MSC, or basal media for 4 hours at 37°C. The viability of the endothelium was evaluated using the optimized vital staining protocol. RESULTS The optimal vital staining was achieved with 0.4% trypan blue for 60 seconds and 0.5% alizarin red for 90 seconds. The optimal oxidative injury (for consistency and level of damage) was obtained with 1% hydrogen peroxide for 15 seconds. Treatment with both WT Co-MSC and PEDF K/O Co-MSC secretome significantly reduced the endothelial damage compared with control (17.2% ± 10.0%, 33.5% ± 11.6%, and 68% ± 17%, respectively, P < 0.01). The WT Co-MSC secretome was significantly more effective compared with PEDF K/O Co-MSC secretome (P < 0.05). CONCLUSIONS A reproducible model of vital staining and oxidative injury is described for studying porcine corneal endothelial survival. Our results demonstrate a beneficial role of a corneal MSC secretome in reducing oxidative damage to the corneal endothelium. In addition, it suggests a potential role for PEDF in this process.
Collapse
|
17
|
Tsuruhisa S, Matsui T, Koga Y, Sotokawauchi A, Yagi M, Yamagishi SI. Pigment epithelium-derived factor inhibits advanced glycation end product-induced proliferation, VEGF and MMP-9 expression in breast cancer cells via interaction with laminin receptor. Oncol Lett 2021; 22:629. [PMID: 34267821 DOI: 10.3892/ol.2021.12890] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/14/2021] [Indexed: 12/27/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF) is one of the adipocytokines with multifaceted functions, which may serve a role in the development of various types of cardiometabolic disorders. Advanced glycation end products (AGEs) have been shown to contribute to numerous aging-associated disorders, such as cancer. However, it remains unclear whether and how PEDF exerts antitumor effects in AGE-exposed human breast cancer MCF-7 cells, and therefore this was explored in the present study. NADPH oxidase activity was measured with luciferase assay, while gene and protein expression levels were evaluated with quantitative PCR and western blot analysis, respectively. AGEs significantly increased NADPH oxidase-driven superoxide generation, cytochrome b-245 β chain (gp91phox) and receptor for AGE (RAGE) mRNA expression, proliferation, mRNA and protein expression levels of vascular endothelial growth factor (VEGF), and matrix metalloproteinase (MMP)-9 mRNA expression in MCF-7 cells, all of which were dose-dependently inhibited by PEDF. Neutralizing antibody against laminin receptor (LR-Ab) significantly blocked these beneficial effects of PEDF in AGE-exposed MCF-7 cells. Furthermore, as in AGE-treated cells, PEDF dose-dependently inhibited the NADPH oxidase-driven superoxide generation, gp91phox, RAGE and MMP-9 mRNA expression, proliferation, mRNA and protein expression levels of VEGF in non-treated control MCF-7 cells, and these effects were also reversed by LR-Ab. LR levels were not affected by the treatment with AGEs, PEDF or LR-Ab. The present study suggested that PEDF may exert antitumor effects in AGE-exposed breast cancer cells by suppressing NADPH oxidase-induced ROS generation and VEGF and MMP-9 expression via interaction with LR. Since PEDF expression is decreased in breast cancer tissues, pharmacological upregulation or restoration of PEDF may inhibit the growth and metastasis of breast cancer.
Collapse
Affiliation(s)
- Shiori Tsuruhisa
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Yoshinori Koga
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Ami Sotokawauchi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Minoru Yagi
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Sho-Ichi Yamagishi
- Division of Diabetes, Metabolism and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan
| |
Collapse
|
18
|
Teuwen LA, De Rooij LPMH, Cuypers A, Rohlenova K, Dumas SJ, García-Caballero M, Meta E, Amersfoort J, Taverna F, Becker LM, Veiga N, Cantelmo AR, Geldhof V, Conchinha NV, Kalucka J, Treps L, Conradi LC, Khan S, Karakach TK, Soenen S, Vinckier S, Schoonjans L, Eelen G, Van Laere S, Dewerchin M, Dirix L, Mazzone M, Luo Y, Vermeulen P, Carmeliet P. Tumor vessel co-option probed by single-cell analysis. Cell Rep 2021; 35:109253. [PMID: 34133923 DOI: 10.1016/j.celrep.2021.109253] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 05/16/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
Tumor vessel co-option is poorly understood, yet it is a resistance mechanism against anti-angiogenic therapy (AAT). The heterogeneity of co-opted endothelial cells (ECs) and pericytes, co-opting cancer and myeloid cells in tumors growing via vessel co-option, has not been investigated at the single-cell level. Here, we use a murine AAT-resistant lung tumor model, in which VEGF-targeting induces vessel co-option for continued growth. Single-cell RNA sequencing (scRNA-seq) of 31,964 cells reveals, unexpectedly, a largely similar transcriptome of co-opted tumor ECs (TECs) and pericytes as their healthy counterparts. Notably, we identify cell types that might contribute to vessel co-option, i.e., an invasive cancer-cell subtype, possibly assisted by a matrix-remodeling macrophage population, and another M1-like macrophage subtype, possibly involved in keeping or rendering vascular cells quiescent.
Collapse
Affiliation(s)
- Laure-Anne Teuwen
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Laura P M H De Rooij
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Anne Cuypers
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Katerina Rohlenova
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Sébastien J Dumas
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Melissa García-Caballero
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Elda Meta
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Jacob Amersfoort
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Federico Taverna
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Lisa M Becker
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Nuphar Veiga
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Anna Rita Cantelmo
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Vincent Geldhof
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Nadine V Conchinha
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Lena-Christin Conradi
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Shawez Khan
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Tobias K Karakach
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Stefaan Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Luc Schoonjans
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 510275, Guangzhou, Guangdong, P.R. China
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Steven Van Laere
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, CCB, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, Qingdao 266555, P.R. China; BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, BGI-Shenzhen, Shenzhen 518120, P.R. China.
| | - Peter Vermeulen
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 510275, Guangzhou, Guangdong, P.R. China; Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark.
| |
Collapse
|
19
|
Tomiyasu T, Sato A, Mori H, Okazaki K. L233P mutation in the bovine leukemia virus Tax protein has impact on annexin A3 and type I collagen secretion by host cells. Vet Microbiol 2021; 256:109042. [PMID: 33819840 DOI: 10.1016/j.vetmic.2021.109042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/19/2021] [Indexed: 01/02/2023]
Abstract
Bovine leukemia virus (BLV) is the causative agent of enzootic bovine leukosis (EBL) and can be classified into two types based on the amino acid at position 233 in Tax protein, which probably plays crucial roles in leukemogenesis. We previously revealed that L233-Tax-expressing cells secreted chemoattractants for endothelial cells and formed significantly larger tumors accompanying neovascularization than P233-Tax-expressing cells in athymic mice. In the present study, comparative proteomic analysis of the culture medium of Tax-expressing cells revealed that annexin A3 and probably extracellular matrix protein 1 served as chemoattractants. Conversely, L233-Tax-expressing cells were impaired in the secretion of collagen alpha-1 (I) chain precursor, which participates in tissue tension homeostasis, leading to tumor mass development. The analysis also demonstrated that both L233-Tax- and P233-Tax-expressing cells had deficits in the secretion of potentially antiangiogenic molecules, including pigment epithelium-derived factor and collagen alpha-1 (VIII) chain, and they produced complement component 3, which might participate in tumor cell proliferation, metastasis, and immune evasion. These findings provided novel insights into prognostication of EBL and the function of Tax in leukemogenesis induced by BLV.
Collapse
Affiliation(s)
- Takafumi Tomiyasu
- Laboratory of Microbiology and Immunology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| | - Ayuki Sato
- Laboratory of Microbiology and Immunology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| | - Hiroshi Mori
- Laboratory of Microbiology and Immunology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| | - Katsunori Okazaki
- Laboratory of Microbiology and Immunology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan.
| |
Collapse
|
20
|
Brook N, Brook E, Dass CR, Chan A, Dharmarajan A. Pigment Epithelium-Derived Factor and Sex Hormone-Responsive Cancers. Cancers (Basel) 2020; 12:cancers12113483. [PMID: 33238558 PMCID: PMC7700359 DOI: 10.3390/cancers12113483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Oestrogens and androgens play important roles in normal and cancerous tissue and have been shown to negatively regulate pigment epithelium-derived factor (PEDF) expression in sex hormone-responsive tumours. PEDF suppresses tumour growth and its downregulation by oestrogen is implicated in tumorigenesis, metastasis, and progression. PEDF expression is reduced in cancerous tissue of the prostate, breast, ovary, and endometrium compared to their normal tissue counterparts, with a link between PEDF downregulation and sex hormone signalling observed in pre-clinical studies. PEDF reduces growth and metastasis of tumour cells by promoting apoptosis, inhibiting angiogenesis, increasing adhesion, and reducing migration. PEDF may also prevent treatment resistance in some cancers by downregulating oestrogen receptor signalling. By interacting with components of the tumour microenvironment, PEDF counteracts the proliferative and immunosuppressive effects of oestrogens, to ultimately reduce tumorigenesis and metastasis. In this review, we focus on sex hormone regulation of PEDF's anti-tumour action in sex hormone-responsive tumours.
Collapse
Affiliation(s)
- Naomi Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA 6102, Australia; (N.B.); (E.B.)
- Curtin Health Innovation Research Institute, Bentley, WA 6102, Australia
| | - Emily Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA 6102, Australia; (N.B.); (E.B.)
- Curtin Health Innovation Research Institute, Bentley, WA 6102, Australia
| | - Crispin R. Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA 6102, Australia; (N.B.); (E.B.)
- Curtin Health Innovation Research Institute, Bentley, WA 6102, Australia
- Correspondence: (C.R.D.); (A.D.); Tel.: +61-8-9266-1489 (C.R.D.)
| | - Arlene Chan
- School of Medicine, Curtin University, Bentley, WA 6102, Australia;
- Breast Cancer Research Centre-Western Australia, Hollywood Private Hospital, Nedlands, WA 6009, Australia
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India
- Correspondence: (C.R.D.); (A.D.); Tel.: +61-8-9266-1489 (C.R.D.)
| |
Collapse
|
21
|
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: 4.3] [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
|
22
|
Bou-Dargham MJ, Sang QXA. Secretome analysis reveals upregulated granzyme B in human androgen-repressed prostate cancer cells with mesenchymal and invasive phenotype. PLoS One 2020; 15:e0237222. [PMID: 32764784 PMCID: PMC7413421 DOI: 10.1371/journal.pone.0237222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 07/22/2020] [Indexed: 11/18/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a critical early step in cancer metastasis and a complex process that involves multiple factors. In this study, we used proteomics approaches to investigate the secreted proteins (secretome) of paired human androgen-repressed prostate cancer (ARCaP) cell lines, representing the epithelial (ARCaP-E) and mesenchymal (ARCaP-M) phenotypes. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analyses showed high levels of proteins involved in bone remodeling and extracellular matrix degradation in the ARCaP-M cells, consistent with the bone metastasis phenotype. Furthermore, LC-MS/MS showed a significantly higher level of the serine protease granzyme B (GZMB) in ARCaP-M conditioned media (CM) compared to that of ARCaP-E. Using quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) to detect mRNA and Western blot to detect protein expression, we further demonstrated that the GZMB gene was expressed by ARCaP-M and the protein was secreted extracellularly. ARCaP-M cells with GZMB gene knockdown using small interfering RNA (siRNA) have markedly reduced invasiveness as demonstrated by the Matrigel invasion assay in comparison with the scrambled siRNA negative control. This study reports that GZMB secretion by mesenchymal-like androgen-repressed human prostate cancer cells promotes invasion, suggesting a possible extracellular role for GZMB in addition to its classic role in immune cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Mayassa J. Bou-Dargham
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States of America
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States of America
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, United States of America
| |
Collapse
|
23
|
Dixit S, Polato F, Samardzija M, Abu-Asab M, Grimm C, Crawford SE, Becerra SP. PEDF deficiency increases the susceptibility of rd10 mice to retinal degeneration. Exp Eye Res 2020; 198:108121. [PMID: 32721425 DOI: 10.1016/j.exer.2020.108121] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/29/2022]
Abstract
The SERPINF1 gene encodes pigment epithelium-derived factor (PEDF), a member of the serpin superfamily with neurotrophic and antiangiogenic properties in the retina. We hypothesized that absence of PEDF would lead to increased stress-associated retinal degeneration in Serpinf1 null mice. Accordingly, using a Serpinf1 null mouse model, we investigated the impact of PEDF absence on retinal morphology, and susceptibility to induced and inherited retinal degeneration. We studied the pattern of Serpinf1 expression in the mouse retina layers. PEDF protein was detected by western blotting. Transmission electron microscopy was performed on mouse retina. Serpinf1 null mice and wild type littermates were injected with NaIO3 (30 mg/kg body weight) intraperitonially. At post-injection day 1, 3, 4, 6 and 8 mice were euthanized, and eyes were enucleated. Serpinf1 null and rd10 double mutant mice were generated and their eyes enucleated at different time points from post-natal day 15 to post-natal day 28. Enucleated eyes were processed for hematoxylin and eosin staining and histopathological evaluations. We found that Serpinf1 was expressed in the retinal pigment epithelium, in the inner nuclear layer and in the ganglion cell layer, but undetectable in the outer nuclear layer of wild type mice. Plasma PEDF protein levels were undetectable in Serpinf1 null animals. RPE atrophy and retinal thinning were observed in NaIO3-treated wild type mice that progressed with time post-injection. NaIO3-treated Serpinf1 null mice showed comparatively better retinal morphology than wild type mice at day 4 post-injection. However, the absence of PEDF in Serpinf1 null x rd10 mice increased the susceptibility to retinal degeneration relative to that of rd10 mice. We concluded that histopathological evaluation of retinas lacking PEDF showed that removal of the Serpinf1 gene may activate PEDF-independent compensatory mechanisms to protect the retina against oxidative stress, while it increases the susceptibility to degenerate the retina in inherited retinal degeneration models.
Collapse
Affiliation(s)
- Shivani Dixit
- Section of Protein Structure and Function, LRCMB-NEI-NIH, Bethesda, MD, USA
| | - Federica Polato
- Section of Protein Structure and Function, LRCMB-NEI-NIH, Bethesda, MD, USA
| | - Marijana Samardzija
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Zurich, Switzerland
| | | | - Christian Grimm
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Zurich, Switzerland
| | - Susan E Crawford
- Department of Surgery, NorthShore University Health System Research Institute, Evanston, IL, USA
| | - S Patricia Becerra
- Section of Protein Structure and Function, LRCMB-NEI-NIH, Bethesda, MD, USA.
| |
Collapse
|
24
|
de Diego-Otero Y, Giráldez-Pérez RM, Lima-Cabello E, Heredia-Farfan R, Calvo Medina R, Sanchez-Salido L, Pérez Costillas L. Pigment epithelium-derived factor (PEDF) and PEDF-receptor in the adult mouse brain: Differential spatial/temporal localization pattern. J Comp Neurol 2020; 529:141-158. [PMID: 32427349 DOI: 10.1002/cne.24940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a multifunctional protein which was initially described in the retina, although it is also present in other tissues. It functions as an antioxidant agent promoting neuronal survival. Recently, a PEDF receptor has shown an elevated binding affinity for PEDF. There are no relevant data regarding the distribution of both proteins in the brain, therefore the main goal of this work was to investigate the spatiotemporal presence of PEDF and PEDFR in the adult mouse brain, and to determine the PEDF blood level in mouse and human. The localization of both proteins was analyzed by different experimental methods such as immunohistochemistry, western-blotting, and also by enzyme-linked immunosorbent assay. Differential expression was found in some telencephalic structures and positive signals for both proteins were detected in the cerebellum. The magnitude of the PEDFR labeling pattern was higher than PEDF and included some cortical and subventricular areas. Age-dependent changes in intensity of both protein immunoreactions were found in the cortical and hippocampal areas with greater reactivity between 4 and 8 months of age, whilst others, like the subventricular zones, these differences were more evident for PEDFR. Although ubiquitous presence was not found in the brain for these two proteins, their relevant functions must not be underestimated. It has been described that PEDF plays an important role in neuroprotection and data provided in the present work represents the first extensive study to understand the relevance of these two proteins in specific brain areas.
Collapse
Affiliation(s)
- Yolanda de Diego-Otero
- Research Laboratory, Hospital Civil, Institute of Biomedical Research in Malaga (IBIMA), Málaga, Spain.,Mental Health Clinic Unit, .Regional University Hospital, Hospital Civil, Málaga, Spain.,Research Unit, International Institute of Innovation and Attention to Neurodevelopment and Language, Málaga, Spain
| | - Rosa María Giráldez-Pérez
- Cellular Biology, Physiology and Immunology Department, University of Cordoba, Edificio Charles Darwin, Córdoba, Spain
| | - Elena Lima-Cabello
- Research Laboratory, Hospital Civil, Institute of Biomedical Research in Malaga (IBIMA), Málaga, Spain
| | - Raúl Heredia-Farfan
- Research Laboratory, Hospital Civil, Institute of Biomedical Research in Malaga (IBIMA), Málaga, Spain
| | - Rocío Calvo Medina
- Pediatric Clinic Unit. Regional University Hospital, Hospital Materno-Infantil Avd, Arroyo de los Angeles, Málaga, Spain
| | - Lourdes Sanchez-Salido
- Research Laboratory, Hospital Civil, Institute of Biomedical Research in Malaga (IBIMA), Málaga, Spain
| | - Lucía Pérez Costillas
- Mental Health Clinic Unit, .Regional University Hospital, Hospital Civil, Málaga, Spain.,Psychiatry and Physiotherapy Department, University of Malaga. Medical School, Málaga, Spain
| |
Collapse
|
25
|
Bar-Joseph H, Hikri E, Chuderland D, Ben-Ami I, Shalgi R. Pigment epithelium derived factor as a novel multi-target treatment for uterine fibroids. Reprod Biomed Online 2020; 41:335-342. [PMID: 32532667 DOI: 10.1016/j.rbmo.2020.03.024] [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] [Received: 12/05/2019] [Revised: 03/12/2020] [Accepted: 03/31/2020] [Indexed: 01/11/2023]
Abstract
RESEARCH QUESTION Does recombinant pigment epithelium derived factor (PEDF) have potential in treating uterine fibroids? DESIGN In-vitro models that used human leiomyoma and Eker rat uterine leiomyoma (ELT-3) cell lines. The ELT-3 cell line was used to examine cellular targets after adding recombinant PEDF to the culture media. Athymic nude female mice were used as an in-vivo model. They were injected with ELT-3 cells to induce ectopic fibroid lesions, then treated with recombinant PEDF. RESULTS RNA expression of PEDF and its receptors was found in both leiomyoma cell lines, as well as the expression of PEDF receptors. Addition of recombinant PEDF to the culture medium of leiomyoma cell lines activated ERK in a time-dependent manner, induced down-regulation of vascular endothelial growth factor mRNA and protein, as well as the mRNAs of oestrogen receptors alpha and beta and inhibited cellular proliferation. Treatment of mice-bearing fibroids with recombinant PEDF reduced fibroid growth rate and resulted in smaller tumours. CONCLUSIONS This study suggests that recombinant PEDF is a putative novel potent physiological treatment for uterine fibroids. It targets several cornerstones of fibroid pathobiology in parallel, including vascular endothelial growth factor and oestrogen receptors, which are needed for vascularization, and restricts fibroid growth and final size in an animal model.
Collapse
Affiliation(s)
- Hadas Bar-Joseph
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Ramat-Aviv, Israel
| | - Elad Hikri
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Ramat-Aviv, Israel
| | - Dana Chuderland
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Ramat-Aviv, Israel
| | - Ido Ben-Ami
- IVF and Infertility Unit, Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, affiliated with the Hebrew University Medical School of Jerusalem Jerusalem, Israel.
| | - Ruth Shalgi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Ramat-Aviv, Israel
| |
Collapse
|
26
|
Brook N, Brook E, Dharmarajan A, Chan A, Dass CR. Pigment epithelium-derived factor regulation of neuronal and stem cell fate. Exp Cell Res 2020; 389:111891. [DOI: 10.1016/j.yexcr.2020.111891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/02/2020] [Accepted: 02/04/2020] [Indexed: 01/25/2023]
|
27
|
Yamagishi SI, Koga Y, Sotokawauchi A, Hashizume N, Fukahori S, Matsui T, Yagi M. Therapeutic Potential of Pigment Epithelium-derived Factor in Cancer. Curr Pharm Des 2020; 25:313-324. [PMID: 30892156 DOI: 10.2174/1381612825666190319112106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/13/2019] [Indexed: 12/11/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is one of the serine protease inhibitors with multifunctional properties, which is produced by various types of organs and tissues. There is an accumulating body of evidence that PEDF plays an important role in the maintenance of tissue homeostasis. Indeed, PEDF not only works as an endogenous inhibitor of angiogenesis, but also suppresses oxidative stress, inflammatory and thrombotic reactions in cell culture systems, animal models, and humans. Furthermore, we, along with others, have found that PEDF inhibits proliferation of, and induces apoptotic cell death in, numerous kinds of tumors. In addition, circulating as well as tumor expression levels of PEDF have been inversely associated with tumor growth and metastasis. These observations suggest that supplementation of PEDF proteins and/or enhancement of endogenous PEDF expression could be a novel therapeutic strategy for the treatment of cancer. Therefore, in this paper, we review the effects of PEDF on diverse types of cancer, and discuss its therapeutic perspectives.
Collapse
Affiliation(s)
- Sho-Ichi Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Yoshinori Koga
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan.,Department of Pediatric Surgery, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Ami Sotokawauchi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Naoki Hashizume
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Suguru Fukahori
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Minoru Yagi
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume 830-0011, Japan
| |
Collapse
|
28
|
Nardi F, Fitchev P, Brooks KM, Franco OE, Cheng K, Hayward SW, Welte MA, Crawford SE. Lipid droplet velocity is a microenvironmental sensor of aggressive tumors regulated by V-ATPase and PEDF. J Transl Med 2019; 99:1822-1834. [PMID: 31409893 PMCID: PMC7289525 DOI: 10.1038/s41374-019-0296-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/19/2022] Open
Abstract
Lipid droplets (LDs) utilize microtubules (MTs) to participate in intracellular trafficking of cargo proteins. Cancer cells accumulate LDs and acidify their tumor microenvironment (TME) by increasing the proton pump V-ATPase. However, it is not known whether these two metabolic changes are mechanistically related or influence LD movement. We postulated that LD density and velocity are progressively increased with tumor aggressiveness and are dependent on V-ATPase and the lipolysis regulator pigment epithelium-derived factor (PEDF). LD density was assessed in human prostate cancer (PCa) specimens across Gleason scores (GS) 6-8. LD distribution and velocity were analyzed in low and highly aggressive tumors using live-cell imaging and in cells exposed to low pH and/or treated with V-ATPase inhibitors. The MT network was disrupted and analyzed by α-tubulin staining. LD density positively correlated with advancing GS in human tumors. Acidification promoted peripheral localization and clustering of LDs. Highly aggressive prostate, breast, and pancreatic cell lines had significantly higher maximum LD velocity (LDVmax) than less aggressive and benign cells. LDVmax was MT-dependent and suppressed by blocking V-ATPase directly or indirectly with PEDF. Upon lowering pH, LDs moved to the cell periphery and carried metalloproteinases. These results suggest that acidification of the TME can alter intracellular LD movement and augment velocity in cancer. Restoration of PEDF or blockade of V-ATPase can normalize LD distribution and decrease velocity. This study identifies V-ATPase and PEDF as new modulators of LD trafficking in the cancer microenvironment.
Collapse
Affiliation(s)
- Francesca Nardi
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201
| | - Philip Fitchev
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201
| | - Kyrsten M. Brooks
- Department of Pathology, Saint Louis University School of Medicine, 1402 South Grand Blvd., Saint Louis, MO 63104
| | - Omar E. Franco
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201
| | - Kevin Cheng
- Department of Pathology, Saint Louis University School of Medicine, 1402 South Grand Blvd., Saint Louis, MO 63104
| | - Simon W. Hayward
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201
| | - Michael A. Welte
- Department of Biology, University of Rochester, RC Box 270211, Rochester, NY 14627
| | - Susan E. Crawford
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201,Department of Pathology, Saint Louis University School of Medicine, 1402 South Grand Blvd., Saint Louis, MO 63104
| |
Collapse
|
29
|
Wei Y, Elahy M, Friedhuber AM, Wong JY, Hughes JD, Doschak MR, Dass CR. Triple-threat activity of PEDF in bone tumors: Tumor inhibition, tissue preservation and cardioprotection against doxorubicin. Bone 2019; 124:103-117. [PMID: 31028961 DOI: 10.1016/j.bone.2019.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/20/2019] [Accepted: 04/23/2019] [Indexed: 12/31/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is known for its osteogenic properties, but its effects against primary and secondary bone tumors have not comprehensively been demonstrated. We show the ubiquitous expression of PEDF in murine embryonic tissue. Continuous administration of PEDF in pregnant mice for five days did not adversely affect foetal health, despite PEDF's known potent antiangiogenic properties. In the case of the devastating childhood bone cancer osteosarcoma, PEDF has direct anticancer activity per se, and protects against the toxicity of doxorubicin in the heart, small intestine and testes. PEDF demonstrated anti-proliferative and pro-apoptotic effects against human prostate and breast cancer cells, tumors which are known to metastasize to bone as the preferred secondary site. Caspase-2 was activated in both tumor cell types by PEDF. In models of prostate and breast cancer in bone, PEDF significantly reduced tumor volumes. When combined with zoledronic acid, continuously-administered PEDF significantly reduced breast tumor volume at the bone, and was able to preserve the quality of bone better than the combination therapy. These multiple positive findings make PEDF an ideal endogenous and safe biological for possible future clinical testing.
Collapse
Affiliation(s)
- Yongzhong Wei
- Department of Orthopaedics, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Mina Elahy
- School of Medical Sciences, University of New South Wales, Kensington, Sydney, NSW 2052, Australia
| | - Anna M Friedhuber
- Department of Pathology, University of Melbourne, Parkville, Melbourne, VIC 3050, Australia
| | - Jia Y Wong
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Jeffery D Hughes
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Michael R Doschak
- Department of Biomedical Engineering, University of Alberta, Alberta T6G 2E1, Canada; Department of Dentistry, University of Alberta, Alberta T6G 2E1, Canada
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Perth, WA 6102, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; College of Health and Biomedicine, Victoria University, St Albans, Melbourne, VIC 3021, Australia.
| |
Collapse
|
30
|
Jones NM, Yang H, Zhang Q, Morales-Tirado VM, Grossniklaus HE. Natural killer cells and pigment epithelial-derived factor control the infiltrative and nodular growth of hepatic metastases in an Orthotopic murine model of ocular melanoma. BMC Cancer 2019; 19:484. [PMID: 31117965 PMCID: PMC6532210 DOI: 10.1186/s12885-019-5712-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/14/2019] [Indexed: 12/13/2022] Open
Abstract
Background Metastases account for 90% of all cancer-related deaths, becoming a therapeutic problem. Approximately 50% of all uveal melanoma (UM) patients will develop metastases, mainly in the liver. Post-mortem analyses of livers from metastatic UM patients showed two different metastatic growth patterns: infiltrative and nodular. The infiltrative pattern exhibits tumor infiltration directly to the hepatic lobule and minimal angiogenesis. The nodular pattern shows clusters of tumor cells around the portal venules that efface the liver parenchyma. We recently demonstrated Natural Killer (NK) cells play a pivotal role in the control of hepatic metastases and the pigment epithelial-derived factor (PEDF) controls angiogenesis in the liver using our established ocular melanoma animal model. In this study we investigated the role of NK cells and PEDF in the development of metastatic growth patterns, as this can contribute to the development of novel therapeutics specific towards each growth pattern. Methods We utilize our established ocular melanoma animal model by inoculation of B16-LS9 melanoma cells into C57BL/6 J mice (WT), anti-asialo GM1-treated C57BL/6 J mice (NK-depleted), and PEDF−/− C57BL/6 J mice. Three weeks after inoculation we evaluated the metastatic growth patterns and stratified them based of the numbers of tumor cells. To evaluate angiogenesis the mean vascular density (MVD) was calculated. The immune compartment of the liver was analyzed by flow cytometry. Results Our in vivo work showed two distinct metastatic growth patterns, the infiltrative and nodular, recapitulating the post-mortem analyses on human liver tissue. We discovered NK cells control the infiltrative growth. In contrast, PEDF controlled anti-angiogenic responses, showing higher MVD values compared to NK-depleted and WT animals. The myeloid lineage, comprised of monocytes, macrophages, and myeloid-derived suppressor cells, was reduced in the absence of NK cells or PEDF. Conclusions Our animal model recapitulates the metastatic growth patterns observed in the human disease. We demonstrated a role for NK cells in the development of the infiltrative growth pattern, and a role for PEDF in the development of the nodular pattern. The understanding of the complexity associated with the metastatic progression has profound clinical implications in the diagnostic and disease-management as we can develop and direct more effective therapies.
Collapse
Affiliation(s)
- Nyasia M Jones
- Graduate Division of Biological and Biomedical Sciences Cancer Biology and Translational Oncology, Emory University, Atlanta, GA, 30322, USA
| | - Hua Yang
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Qing Zhang
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Vanessa M Morales-Tirado
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Hans E Grossniklaus
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Winship Cancer Institute at Emory University, 1365 Clifton Road NE, BT428, Atlanta, GA, 30322, USA.
| |
Collapse
|
31
|
Niyogi S, Ghosh M, Adak M, Chakrabarti P. PEDF promotes nuclear degradation of ATGL through COP1. Biochem Biophys Res Commun 2019; 512:806-811. [PMID: 30926171 DOI: 10.1016/j.bbrc.2019.03.111] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022]
Abstract
Adipose triglyceride lipase (ATGL) plays a compelling role in hepatic lipid turnover and in the pathophysiology of non-alcoholic fatty liver disease. Hepatic ATGL is post-transcriptionally regulated by E3 ubiquitin ligase constitutive photomorphogenic1 (COP1) through polyubiquitylation and proteasomal degradation. However the physiological cue for COP1-mediated hepatocellular degradation of ATGL remained unknown. Here we checked for the role of pigment epithelium-derived factor (PEDF), a moonlighting hepatokine and the so-called ligand of ATGL for its stability in hepatocytes. We show that PEDF diminishes ATGL protein stability by promoting its proteasomal degradation in COP1-dependent manner. Despite being a secretory glycoprotein, PEDF is also sequestered in the nuclear compartment so as COP1. Interestingly, PEDF enhances nuclear import of predominantly cytosolic ATGL protein for its subsequent proteasomal degradation in the nucleus. PEDF also controls cell autonomous hepatocyte lipid accumulation and mobilization through COP1-ATGL axis, thereby unraveling a novel pathway for hepatic lipid metabolism.
Collapse
Affiliation(s)
- Sougata Niyogi
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Mainak Ghosh
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Moumita Adak
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Partha Chakrabarti
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India; Academy of Innovative and Scientific Research, India.
| |
Collapse
|
32
|
Nardi F, Franco OE, Fitchev P, Morales A, Vickman RE, Hayward SW, Crawford SE. DGAT1 Inhibitor Suppresses Prostate Tumor Growth and Migration by Regulating Intracellular Lipids and Non-Centrosomal MTOC Protein GM130. Sci Rep 2019; 9:3035. [PMID: 30816200 PMCID: PMC6395665 DOI: 10.1038/s41598-019-39537-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 12/31/2018] [Indexed: 11/10/2022] Open
Abstract
Acyl-CoA:diacylglycerol acyltransferase I (DGAT1) is a key enzyme in lipogenesis which is increased in metabolically active cells to meet nutrient requirements. DGAT1 has been recognized as an anti-obesity target; however, its role in the tumor microenvironment remains unclear. We postulated that, in prostate cancer (PCa) cells, augmented lipogenesis and growth are due to increased DGAT1 expression leading to microtubule-organizing center (MTOC) amplification. Thus, therapeutic targeting of DGAT1 potentially has tumor suppressive activity. We tested whether blocking DGAT1 in PCa cells altered MTOC and lipid signaling. Western blot and immunofluorescence were performed for MTOC and triglyceride mediators. Treatment with a DGAT1 inhibitor was evaluated. We found a stepwise increase in DGAT1 protein levels when comparing normal prostate epithelial cells to PCa cells, LNCaP and PC-3. Lipid droplets, MTOCs, and microtubule-regulating proteins were reduced in tumor cells treated with a DGAT1 inhibitor. Depletion of the non-centrosomal MTOC protein GM130 reduced PCa cell proliferation and migration. Inhibition of DGAT1 reduced tumor growth both in vitro and in vivo, and a negative feedback loop was discovered between DGAT1, PEDF, and GM130. These data identify DGAT1 as a promising new target for suppressing PCa growth by regulating GM130, MTOC number and disrupting microtubule integrity.
Collapse
Affiliation(s)
- Francesca Nardi
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Omar E Franco
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Philip Fitchev
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Alejandro Morales
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Renee E Vickman
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Simon W Hayward
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Susan E Crawford
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States.
| |
Collapse
|
33
|
Léveillard T, Klipfel L. Mechanisms Underlying the Visual Benefit of Cell Transplantation for the Treatment of Retinal Degenerations. Int J Mol Sci 2019; 20:ijms20030557. [PMID: 30696106 PMCID: PMC6387096 DOI: 10.3390/ijms20030557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/13/2022] Open
Abstract
The transplantation of retinal cells has been studied in animals to establish proof of its potential benefit for the treatment of blinding diseases. Photoreceptor precursors have been grafted in animal models of Mendelian-inherited retinal degenerations, and retinal pigmented epithelial cells have been used to restore visual function in animal models of age-related macular degeneration (AMD) and recently in patients. Cell therapy over corrective gene therapy in inherited retinal degeneration can overcome the genetic heterogeneity by providing one treatment for all genetic forms of the diseases. In AMD, the existence of multiple risk alleles precludes a priori the use of corrective gene therapy. Mechanistically, the experiments of photoreceptor precursor transplantation reveal the importance of cytoplasmic material exchange between the grafted cells and the host cells for functional rescue, an unsuspected mechanism and novel concept. For transplantation of retinal pigmented epithelial cells, the mechanisms behind the therapeutic benefit are only partially understood, and clinical trials are ongoing. The fascinating studies that describe the development of methodologies to produce cells to be grafted and demonstrate the functional benefit for vision are reviewed.
Collapse
Affiliation(s)
- Thierry Léveillard
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| | - Laurence Klipfel
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
34
|
Huang KT, Hsu LW, Chen KD, Kung CP, Goto S, Chen CL. Decreased PEDF Expression Promotes Adipogenic Differentiation through the Up-Regulation of CD36. Int J Mol Sci 2018; 19:ijms19123992. [PMID: 30544997 PMCID: PMC6321369 DOI: 10.3390/ijms19123992] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 01/10/2023] Open
Abstract
Adipogenesis is a tightly regulated cellular process that involves the action of multiple signaling pathways. Characterization of regulators that are associated with adipose development is crucial to understanding the mechanisms underlying obesity and other metabolic disorders. Pigment epithelium-derived factor (PEDF) is a secreted glycoprotein that was first described as a neurotrophic factor. The role of PEDF in lipid metabolism was established when adipose triglyceride lipase (ATGL), a major triglyceride hydrolase, was characterized as its binding partner. In this study, we investigated the downstream effects of PEDF on adipogenic differentiation using rat adipose-derived stem cells (AdSCs) and the mouse pre-adipocyte cell line 3T3-L1. Knocking down PEDF in differentiating cells resulted in elevated levels of ATGL and CD36, as well as other adipogenic markers, with a concomitant increase in adipocyte number. CD36, a scavenger receptor for a variety of ligands, regulated proliferation and lipogenic gene expression during adipogenesis. The CD36 increase due to PEDF down-regulation might be a result of elevated PPARγ. We further demonstrated that PEDF expression was regulated by dexamethasone, a synthetic glucocorticoid that is widely used for adipogenesis at the transcriptional level. Taken together, our findings highlight that PEDF negatively regulates adipogenesis through the regulation of various signaling intermediates, and it may play a crucial role in lipid metabolic disorders.
Collapse
Affiliation(s)
- Kuang-Tzu Huang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan.
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan.
| | - Li-Wen Hsu
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan.
| | - Kuang-Den Chen
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan.
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan.
| | - Chao-Pin Kung
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan.
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan.
| | - Shigeru Goto
- Fukuoka Institute of Occupational Health, Fukuoka 815-0081, Japan.
| | - Chao-Long Chen
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan.
| |
Collapse
|
35
|
Ma S, Wang S, Li M, Zhang Y, Zhu P. The effects of pigment epithelium-derived factor on atherosclerosis: putative mechanisms of the process. Lipids Health Dis 2018; 17:240. [PMID: 30326915 PMCID: PMC6192115 DOI: 10.1186/s12944-018-0889-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 10/03/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death worldwide. Atherosclerosis is believed to be the major cause of CVD, characterized by atherosclerotic lesion formation and plaque disruption. Although remarkable advances in understanding the mechanisms of atherosclerosis have been made, the application of these theories is still limited in the prevention and treatment of atherosclerosis. Therefore, novel and effective strategies to treat high-risk patients with atherosclerosis require further development. Pigment epithelium-derived factor (PEDF), a glycoprotein with anti-inflammatory, anti-oxidant, anti-angiogenic, anti-thrombotic and anti-tumorigenic properties, is of considerable interest in the prevention of atherosclerosis. Accumulating research has suggested that PEDF exerts beneficial effects on atherosclerotic lesions and CVD patients. Our group, along with colleagues, has demonstrated that PEDF may be associated with acute coronary syndrome (ACS), and that the polymorphisms of rs8075977 of PEDF are correlated with coronary artery disease (CAD). Moreover, we have explored the anti-atherosclerosis mechanisms of PEDF, showing that oxidized-low density lipoprotein (ox-LDL) reduced PEDF concentrations through the upregulation of reactive oxygen species (ROS), and that D-4F can protect endothelial cells against ox-LDL-induced injury by preventing the downregulation of PEDF. Additionally, PEDF might alleviate endothelial injury by inhibiting the Wnt/β-catenin pathway. These data suggest that PEDF may be a novel therapeutic target for the treatment of atherosclerosis. In this review, we will summarize the role of PEDF in the development of atherosclerosis, focusing on endothelial dysfunction, inflammation, oxidative stress, angiogenesis and cell proliferation. We will also discuss its promising therapeutic implications for atherosclerosis.
Collapse
Affiliation(s)
- Shouyuan Ma
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Shuxia Wang
- Department of Cadre Clinic, Chinese PLA General Hospital, Beijing, 100853, China
| | - Man Li
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yan Zhang
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Ping Zhu
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
36
|
Jarvis C, Nelius T, Martinez-Marin D, Sennoune SR, Filleur S. Cabazitaxel regimens inhibit the growth of prostate cancer cells and enhances the anti-tumor properties of PEDF with various efficacy and toxicity. Prostate 2018; 78:905-914. [PMID: 29749077 DOI: 10.1002/pros.23647] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/16/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Taxanes chemotherapies represent the major therapeutic alternative for symptomatic mCRPC. While docetaxel is the most commonly prescribed Taxane for mCRPC; cabazitaxel has been approved for patients unresponsive to docetaxel. Still mCRPC remains incurable and patients often experience severe side effects. Recently, the FIRSTANA trial first demonstrated the absence of superiority in overall survival between cabazitaxel and docetaxel in mCRPC patients. Inversely, different toxicity were reported suggesting that cabazitaxel may provide a first line treatment option for some patients urging for a deeper characterization of cabazitaxel mechanisms of action as well as a re-evaluation of cabazitaxel conventional dose and schedule. In this study, our goal was therefore to evaluate the anti-tumor efficacy of various cabazitaxel regimens delivered as monotherapy or in combination with PEDF, a known anti-angiogenic and anti-neoplastic agent. METHODS CRPC cells undergoing Taxane treatment were evaluated for cell proliferation, migration and death, and apoptosis using crystal violet staining, chemotaxis, cell cycle, and TUNEL assays. In vitro data were corroborated in CL1 CRPC xenografts where mice received intermittent or metronomic low-doses cabazitaxel ± PEDF. RESULTS We found that cabazitaxel inhibits the proliferation of CRPC cells with a higher efficacy than docetaxel in vitro. As expected, high-doses of Taxanes blocked the cells in mitosis. Surprisingly, low-doses of cabazitaxel induced more cell death than docetaxel mainly through apoptosis. In vivo, intermittent cabazitaxel lead to disease stabilization when combined with PEDF. Unexpectedly, low-doses of cabazitaxel delayed tumor growth with severe toxicity for some of the doses tested. Other results showed that PEDF and low-doses of cabazitaxel combination inhibited the migration of tumor cell and increased the tumoricidal activity of macrophages toward prostate tumor cells. CONCLUSIONS Our findings highlight the great promise of cabazitaxel drug and predict a possible move of cabazitaxel forward within the therapeutic sequence of prostate cancer.
Collapse
Affiliation(s)
- Courtney Jarvis
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas
- Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, Lubbock, Texas
| | - Thomas Nelius
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas
| | - Dalia Martinez-Marin
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas
| | - Souad R Sennoune
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University-Health Sciences Center, Lubbock, Texas
| | - Stéphanie Filleur
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas
- Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, Lubbock, Texas
| |
Collapse
|
37
|
Michalczyk ER, Chen L, Fine D, Zhao Y, Mascarinas E, Grippo PJ, DiPietro LA. Pigment Epithelium-Derived Factor (PEDF) as a Regulator of Wound Angiogenesis. Sci Rep 2018; 8:11142. [PMID: 30042381 PMCID: PMC6057962 DOI: 10.1038/s41598-018-29465-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/10/2018] [Indexed: 12/15/2022] Open
Abstract
Although the inflammatory and proliferative phases of wound healing have been well described, much less is known about how healing resolves. During the resolution phase, pruning of the capillary bed and maturation of capillaries occurs and influences the final strength and fidelity of the wound. PEDF, an endogenous anti-angiogenic factor, is produced in wounds and may contribute to the removal of capillaries during wound resolution. This study utilized PEDF-/- mice to examine how PEDF influences wound angiogenesis, particularly capillary density and permeability. The absence of PEDF led to transient changes in dermal wound closure and collagen content, but caused substantial changes in wound angiogenesis. Compared to wild type (WT) mice, wounds from PEDF-/- mice exhibited a significant increase in capillaries during the proangiogenic phase of repair, and a delay in capillary pruning. Conversely, the addition of rPEDF caused a reduction in capillary density within skin wounds in WT mice. In vitro studies showed that PEDF inhibited migration and tube formation by dermal microvascular endothelial cells, and caused a decrease in the expression of VEGFR2, VCAM-1, and other surface receptors. The results demonstrate that loss of PEDF causes a distinctive wound healing phenotype that is characterized by increased angiogenesis and delayed resolution. The findings suggest that PEDF most likely acts through multiple mechanisms to regulate proper capillary refinement in wounds.
Collapse
Affiliation(s)
- Elizabeth R Michalczyk
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, USA
| | - David Fine
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Yan Zhao
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Emman Mascarinas
- Division of Gastroenterology and Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Paul J Grippo
- Division of Gastroenterology and Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Luisa A DiPietro
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
38
|
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: 1.7] [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
|
39
|
Sadri H, Saremi B, Dänicke S, Rehage J, Mielenz M, Hosseini A, Sauerwein H. Lactation-related changes in tissue expression of PEDF in dairy cows. Domest Anim Endocrinol 2018; 64:93-101. [PMID: 29758402 DOI: 10.1016/j.domaniend.2018.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 12/21/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is evolving as metabolic regulatory protein. Albeit mostly considered in only pathological conditions related to excess energy intake resulting in obesity and insulin resistance, PEDF is likely to be involved in other physiological processes such as the homeorhetic adaptation of metabolism to lactation. We aimed to characterize the expression of PEDF and its association to the concomitant mobilization of body reserves during lactation in nonobese subjects. This mobilization is particularly distinct in dairy cows, and we therefore assessed the mRNA expression of PEDF and its putative receptors in different tissues in 2 trials with dairy cows fed with or without conjugated linoleic acids (CLAs). Conjugated linoleic acids depress milk fat synthesis and may thus reduce the drain of energy via milk. In pluriparous cows, the serum PEDF concentrations and the mRNA abundance in subcutaneous adipose tissue (scAT), as well as the hepatic and scAT mRNA abundance of the putative receptors, adipose triglyceride lipase, and laminin receptor 1, changed over time of sampling (day -21 until day 252 relative to calving). Conjugated linoleic acid treatment was associated with reduced PEDF concentrations in serum and lower PEDF mRNA abundance in scAT on day 21 postpartum. Comparing different tissues from primiparous cows, PEDF mRNA was highest in the liver, followed by scAT, visceral adipose tissue (AT), and mammary gland, and lowest in the muscle. Significant changes in PEDF expression with time of sampling were limited to AT in primiparous and pluriparous cows. Our data support a regulatory role for PEDF. The similarities between the time course of the serum concentrations of PEDF and its mRNA abundance in scAT may point to a regulatory role for AT rather than the liver for PEDF in dairy cows.
Collapse
Affiliation(s)
- H Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran; Institute of Animal Science, Physiology & Hygiene Unit, University of Bonn, Bonn 53111, Germany
| | - B Saremi
- Institute of Animal Science, Physiology & Hygiene Unit, University of Bonn, Bonn 53111, Germany
| | - S Dänicke
- Institute of Animal Nutrition, Friedrich-Loeffler-Institute (FLI), Federal Research Institute for Animal Health, Braunschweig 38116, Germany
| | - J Rehage
- Clinic for Cattle, University of Veterinary Medicine, Hannover 30173, Germany
| | - M Mielenz
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, Dummerstorf 18196, Germany
| | - A Hosseini
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
| | - H Sauerwein
- Institute of Animal Science, Physiology & Hygiene Unit, University of Bonn, Bonn 53111, Germany.
| |
Collapse
|
40
|
Principe DR, DeCant B, Diaz AM, Mangan RJ, Hwang R, Lowy A, Shetuni BB, Sreekumar BK, Chung C, Bentrem DJ, Munshi HG, Jung B, Grippo PJ, Bishehsari F. PEDF inhibits pancreatic tumorigenesis by attenuating the fibro-inflammatory reaction. Oncotarget 2017; 7:28218-34. [PMID: 27058416 PMCID: PMC5053722 DOI: 10.18632/oncotarget.8587] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/17/2016] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is characterized by a pronounced fibro-inflammatory reaction that has been shown to contribute to cancer progression. Previous reports have demonstrated that pigment epithelium-derived factor (PEDF) has potent tumor suppressive effects in pancreatic cancer, though little is known about the mechanisms by which PEDF limits pancreatic tumorigenesis. We therefore employed human specimens, as well as mouse and in vitro models, to explore the effects of PEDF upon the pancreatic microenvironment. We found that PEDF expression is decreased in human pancreatic cancer samples compared to non-malignant tissue. Furthermore, PEDF-deficient patients displayed increased intratumoral inflammation/fibrosis. In mice, genetic ablation of PEDF increased cerulein-induced inflammation and fibrosis, and similarly enhanced these events in the background of oncogenic KRAS. In vitro, recombinant PEDF neutralized macrophage migration as well as inhibited macrophage-induced proliferation of tumor cells. Additionally, recombinant PEDF suppressed the synthesis of pro-inflammatory/pro-fibrotic cytokines both in vivo and in vitro, and reduced collagen I deposition and TGFβ synthesis by pancreatic stellate cells, consistent with reduced fibrosis. Combined, our results demonstrate that PEDF limits pancreatic cancer progression by attenuating the fibro-inflammatory reaction, and makes restoration of PEDF signaling a potential therapeutic approach to study in pancreatic cancer.
Collapse
Affiliation(s)
| | - Brian DeCant
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrew M Diaz
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Riley J Mangan
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Rosa Hwang
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew Lowy
- Department of Surgery, University of California San Diego, San Diego, CA, USA
| | | | - Bharath K Sreekumar
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Chuhan Chung
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - David J Bentrem
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hidayatullah G Munshi
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Barbara Jung
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Faraz Bishehsari
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
41
|
Rivera-Pérez J, Monter-Vera MDR, Barrientos-Alvarado C, Toscano-Garibay JD, Cuesta-Mejías T, Flores-Estrada J. Evaluation of VEGF and PEDF in prostate cancer: A preliminary study in serum and biopsies. Oncol Lett 2017; 15:1072-1078. [PMID: 29391897 PMCID: PMC5769414 DOI: 10.3892/ol.2017.7374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 10/25/2017] [Indexed: 01/18/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) and the pigment epithelium-derived factor (PEDF) serve an important role in prostate cancer (PCa). The aim of the present study was to evaluate whether the levels of VEGF and PEDF in serum are associated with the severity of PCa, and whether they can differentiate from patients with benign prostatic hyperplasia (BPH). Two groups of patients were recruited, patients with PCa or BPH that were newly diagnosed without other comorbidities, and were compared with healthy individuals. The levels of VEGF and PEDF were measured by ELISA in serum, and by immunohistochemistry in biopsies. A correlation analysis was performed for the values in biopsies and serum, comparing the VEGF/PEDF ratio, total-prostate-specific antigen (t-PSA) levels and the status of each sample as acinar Ad (Gleason score) or as benign hyperplasia. The results demonstrated that serum levels of VEGF, PEDF, and t-PSA between PCa and BPH were similar to each other, but different to healthy individuals (P<0.05). The VEGF/PEDF ratio in serum had a significant difference between acinar Ad with Gleason score 8–10 and BPH groups (P<0.05). The VEGF and PEDF immunostaining intensities were correlated with its circulating levels in all cases of PCa, but not in BPH. These preliminary results suggest that VEGF and PEDF levels by themselves or in combination with t-PSA did not differentiate between malignant, and benign prostate diseases. However, there was a significant difference observed in the VEGF/PEDF ratio in serum between the groups, suggesting that it may be used as an index for diagnosis and prognosis in a personalized manner, although more studies are necessary.
Collapse
Affiliation(s)
- Josué Rivera-Pérez
- Direction of Research, Hospital Juárez de México, 07760 Mexico City, Mexico
| | | | | | | | - Teresa Cuesta-Mejías
- Pathological Anatomy Service, Hospital Juárez de México, 07760 Mexico City, CDMX, Mexico
| | | |
Collapse
|
42
|
Duggan C, Tapsoba JDD, Wang CY, Schubert KEF, McTiernan A. Long-Term Effects of Weight Loss and Exercise on Biomarkers Associated with Angiogenesis. Cancer Epidemiol Biomarkers Prev 2017; 26:1788-1794. [PMID: 29042415 DOI: 10.1158/1055-9965.epi-17-0356] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/25/2017] [Accepted: 09/27/2017] [Indexed: 12/13/2022] Open
Abstract
Background: We tested the effect of weight loss on circulating levels of the angiogenic factors VEGF and pigment epithelium-derived factor (PEDF) in postmenopausal overweight/obese women, 18 months after completing a year-long 4-arm randomized controlled trial of behavioral weight loss and/or exercise versus control (i.e., 30 months postrandomization).Methods: The 439 overweight/obese, postmenopausal women, ages 50 to 75 years, were randomized to: diet (goal: 10% weight loss, N = 118), exercise (225 min/wk moderate-to-vigorous activity, N = 117), diet + exercise (N = 117), or control (N = 87). At 12 months, 399 women gave a blood sample; 156 returned at 30 months. Biomarkers were measured by immunoassay. Changes were compared using generalized estimating equations, adjusting for baseline BMI, age, and race/ethnicity.Results: Participants randomized to diet, exercise, and diet + exercise arms had greater reductions in VEGF at 30 months (-14.1% P = 0.02; -19.7% P = 0.003; -14.5% P = 0.002, respectively) versus controls (-4.5%). There were no statistically significant changes in PEDF in any intervention arm. Participants maintaining ≥10% of baseline weight loss at 30 months had greater reductions in VEGF versus those who gained weight/had no weight change (-22.3% vs. -10.2% respectively, P = 0.002). Participants maintaining any weight loss had significantly lower levels of PEDF at 30 months versus those who gained weight/no weight change.Conclusions: Sustained weight loss via diet and/or exercise results in reductions in angiogenic factors, and can be maintained up to 30-month follow-up. Limitations include relatively small numbers, and possible bias toward more successful weight loss among women who returned at 30 months.Impact: Maintaining weight loss can achieve long-term reductions in biomarkers of angiogenesis that can persist up to 18 months after completion of a weight loss intervention. Cancer Epidemiol Biomarkers Prev; 26(12); 1788-94. ©2017 AACR.
Collapse
Affiliation(s)
- Catherine Duggan
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Jean de Dieu Tapsoba
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ching-Yun Wang
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Anne McTiernan
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
43
|
Daubriac J, Pandya UM, Huang KT, Pavlides SC, Gama P, Blank SV, Shukla P, Crawford SE, Gold LI. Hormonal and Growth Regulation of Epithelial and Stromal Cells From the Normal and Malignant Endometrium by Pigment Epithelium-Derived Factor. Endocrinology 2017; 158:2754-2773. [PMID: 28911166 DOI: 10.1210/en.2017-00028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 06/16/2017] [Indexed: 12/19/2022]
Abstract
We discovered that pigment epithelium-derived factor (PEDF)-null mice have endometrial hyperplasia, the precursor to human type I endometrial cancer (ECA), which is etiologically linked to unopposed estrogen (E2), suggesting that this potent antiangiogenic factor might contribute to dysregulated growth and the development of type I ECA. Treatment of both ECA cell lines and primary ECA cells with recombinant PEDF dose dependently decreased cellular proliferation via an autocrine mechanism by blocking cells in G1 and G2 phases of the cell cycle. Consistent with the known opposing effects of E2 and progesterone (Pg) on endometrial proliferation, Pg increases PEDF protein synthesis and release, whereas E2 has the converse effect. Using PEDF luciferase promoter constructs containing two Pg and one E2 response elements, E2 reduced and Pg increased promoter activity due to distal response elements. Furthermore, E2 decreases and Pg increases PEDF secretion into conditioned media (CM) by both normal endometrial stromal fibroblasts (ESFs) and cancer-associated fibroblasts (CAFs), but only CM from ESFs mediated growth-inhibitory activity of primary endometrial epithelial cells (EECs). In addition, in cocultures with primary EECs, Pg-induced growth inhibition is mediated by ESFs, but not CAFs. This is consistent with reduced levels of Pg receptors on CAFs surrounding human malignant glands in vivo. Taken together, the data suggest that PEDF is a hormone-regulated negative autocrine mediator of endometrial proliferation, and that paracrine growth inhibition by soluble factors, possibly PEDF, released by ESFs in response to Pg, but not CAFs, exemplifies a tumor microenvironment that contributes to the pathogenesis of ECA.
Collapse
Affiliation(s)
- Julien Daubriac
- Department of Medicine, Division of Translational Medicine, New York University School of Medicine Langone Medical Center, New York, New York 10016
| | - Unnati M Pandya
- Department of Medicine, Division of Translational Medicine, New York University School of Medicine Langone Medical Center, New York, New York 10016
| | - Kuang-Tzu Huang
- Department of Medicine, Division of Translational Medicine, New York University School of Medicine Langone Medical Center, New York, New York 10016
| | - Savvas C Pavlides
- Department of Medicine, Division of Translational Medicine, New York University School of Medicine Langone Medical Center, New York, New York 10016
| | - Patricia Gama
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paolo, Sao Paolo 05508 000, Brazil
| | - Stephanie V Blank
- Department of Pathology, New York University School of Medicine Langone Medical Center, New York, New York 10016
- Department of Gynecological Oncology, New York University School of Medicine Langone Medical Center, New York, New York 10016
- Perlmutter Cancer Center, New York University School of Medicine Langone Medical Center, New York, New York 10016
| | - Pratibha Shukla
- Department of Pathology, New York University School of Medicine Langone Medical Center, New York, New York 10016
| | - Susan E Crawford
- NorthShore University Research Institute, Affiliate of Chicago Pritizker School of Medicine, Evanston, Illinois 60201
| | - Leslie I Gold
- Department of Medicine, Division of Translational Medicine, New York University School of Medicine Langone Medical Center, New York, New York 10016
- Department of Pathology, New York University School of Medicine Langone Medical Center, New York, New York 10016
- Perlmutter Cancer Center, New York University School of Medicine Langone Medical Center, New York, New York 10016
| |
Collapse
|
44
|
Martinez-Marin D, Jarvis C, Nelius T, de Riese W, Volpert OV, Filleur S. PEDF increases the tumoricidal activity of macrophages towards prostate cancer cells in vitro. PLoS One 2017; 12:e0174968. [PMID: 28403150 PMCID: PMC5389654 DOI: 10.1371/journal.pone.0174968] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 03/19/2017] [Indexed: 12/23/2022] Open
Abstract
Background Although inflammation and prostate cancer (PCa) have been linked, the molecular interactions between macrophages and PCa cells are poorly explored. Pigment Epithelium-Derived Factor (PEDF) is an anti-angiogenic and anti-tumor factor. We previously showed that PEDF induces macrophages recruitment in vitro, correlates with macrophages density in human prostate, and stimulates macrophages polarization towards the classically activated pathway. Here, we demonstrate that PEDF modulates the interaction between macrophages and PCa cells through a bidirectional signalling leading to tumor cell apoptosis and phagocytosis. Methods RAW 264.7 and THP-1 cells, and BMDMs were grown in vitro as mono- or co-cultures with PC3 or CL1 tumor cells. The effects of PEDF and its derived P18 peptide were measured on macrophages differentiation, migration, and superoxide production, and tumor cell apoptosis and phagocytosis. PEDF receptors (ATP5B, PNPLA2, and LRP6) and CD47 mRNA and protein expression were quantified in macrophages and tumor cells by quantitative RT-PCR, western blot, immunofluorescence and flow cytometry. Results We found that PEDF induced the migration of macrophages towards tumor 3D spheroids and 2D cultures. In co-culture, PEDF increased PCa cells phagocytosis through an indirect apoptosis-dependent mechanism. Moreover, PEDF stimulated the production of superoxide by macrophages. Conditioned media from macrophages exposed to PEDF induced tumor cells apoptosis in contrast to control conditioned media suggesting that ROS may be involved in tumor cells apoptosis. ATP5B and PNPLA2 PEDF receptors on macrophages and CD47 on tumor cells were respectively up- and down-regulated by PEDF. As PEDF, blocking CD47 induced phagocytosis. Inhibiting ATP5B reduced phagocytosis. Inversely, PNPLA2 inhibition blocks differentiation but maintains phagocytosis. CD47-induced phagocytosis was partially reverted by ATP5B inhibition suggesting a complementary action. Similar effects were observed with P18 PEDF-derived peptide. Conclusions These data established that modulating the molecular interactions between macrophages and PCa cells using PEDF may be a promising strategy for PCa treatment.
Collapse
Affiliation(s)
- Dalia Martinez-Marin
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Courtney Jarvis
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
- Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Thomas Nelius
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Werner de Riese
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Olga V. Volpert
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Stéphanie Filleur
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
- Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
- * E-mail:
| |
Collapse
|
45
|
Metzler VM, de Brot S, Robinson RS, Jeyapalan JN, Rakha E, Walton T, Gardner DS, Lund EF, Whitchurch J, Haigh D, Lochray JM, Robinson BD, Allegrucci C, Fray RG, Persson JL, Ødum N, Miftakhova RR, Rizvanov AA, Hughes IA, Tadokoro-Cuccaro R, Heery DM, Rutland CS, Mongan NP. Androgen dependent mechanisms of pro-angiogenic networks in placental and tumor development. Placenta 2017; 56:79-85. [PMID: 28238455 DOI: 10.1016/j.placenta.2017.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 11/25/2022]
Abstract
The placenta and tumors share important characteristics, including a requirement to establish effective angiogenesis. In the case of the placenta, optimal angiogenesis is required to sustain the blood flow required to maintain a successful pregnancy, whereas in tumors establishing new blood supplies is considered a key step in supporting metastases. Therefore the development of novel angiogenesis inhibitors has been an area of active research in oncology. A subset of the molecular processes regulating angiogenesis are well understood in the context of both early placentation and tumorigenesis. In this review we focus on the well-established role of androgen regulation of angiogenesis in cancer and relate these mechanisms to placental angiogenesis. The physiological actions of androgens are mediated by the androgen receptor (AR), a ligand dependent transcription factor. Androgens and the AR are essential for normal male embryonic development, puberty and lifelong health. Defects in androgen signalling are associated with a diverse range of clinical disorders in men and women including disorders of sex development (DSD), polycystic ovary syndrome in women and many cancers. We summarize the diverse molecular mechanisms of androgen regulation of angiogenesis and infer the potential significance of these pathways to normal and pathogenic placental function. Finally, we offer potential research applications of androgen-targeting molecules developed to treat cancer as investigative tools to help further delineate the role of androgen signalling in placental function and maternal and offspring health in animal models.
Collapse
Affiliation(s)
- Veronika M Metzler
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Simone de Brot
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Robert S Robinson
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Jennie N Jeyapalan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Emad Rakha
- School of Medicine and Sciences, University of Nottingham, Nottingham City Hospital, NG5 1PB, UK
| | - Thomas Walton
- Department of Urology, Nottingham University Hospitals NHS Trust, NG5 1PB, UK
| | - David S Gardner
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Emma F Lund
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | | | - Daisy Haigh
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Jack M Lochray
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Brian D Robinson
- Department of Pathology, Weill Cornell Medicine, New York 10065, USA
| | - Cinzia Allegrucci
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Rupert G Fray
- School of Biosciences, University of Nottingham, LE12 5RD, UK
| | - Jenny L Persson
- Department of Translational Medicine, Lund University, Malmö, Sweden; Department of Molecular Biology, Umeå University, Sweden
| | - Niels Ødum
- Department of Immunology and Microbiology, University of Copenhagen, Denmark
| | - Regina R Miftakhova
- Department of Molecular Biology, Umeå University, Sweden; Kazan Federal University, Kazan, Republic of Tatarstan 420008, Russian Federation
| | - Albert A Rizvanov
- Kazan Federal University, Kazan, Republic of Tatarstan 420008, Russian Federation
| | - Ieuan A Hughes
- Department of Paediatrics, University of Cambridge, Hills Rd, Cambridge CB2 0QQ, UK
| | | | - David M Heery
- School of Pharmacy, University of Nottingham, NG7 2TQ, UK
| | - Catrin S Rutland
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK.
| | - Nigel P Mongan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK; Department of Pharmacology, Weill Cornell Medicine, New York 10065, USA.
| |
Collapse
|
46
|
Enderli TA, Burtch SR, Templet JN, Carriero A. Animal models of osteogenesis imperfecta: applications in clinical research. Orthop Res Rev 2016; 8:41-55. [PMID: 30774469 PMCID: PMC6209373 DOI: 10.2147/orr.s85198] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Osteogenesis imperfecta (OI), commonly known as brittle bone disease, is a genetic disease characterized by extreme bone fragility and consequent skeletal deformities. This connective tissue disorder is caused by mutations in the quality and quantity of the collagen that in turn affect the overall mechanical integrity of the bone, increasing its vulnerability to fracture. Animal models of the disease have played a critical role in the understanding of the pathology and causes of OI and in the investigation of a broad range of clinical therapies for the disease. Currently, at least 20 animal models have been officially recognized to represent the phenotype and biochemistry of the 17 different types of OI in humans. These include mice, dogs, and fish. Here, we describe each of the animal models and the type of OI they represent, and present their application in clinical research for treatments of OI, such as drug therapies (ie, bisphosphonates and sclerostin) and mechanical (ie, vibrational) loading. In the future, different dosages and lengths of treatment need to be further investigated on different animal models of OI using potentially promising treatments, such as cellular and chaperone therapies. A combination of therapies may also offer a viable treatment regime to improve bone quality and reduce fragility in animals before being introduced into clinical trials for OI patients.
Collapse
Affiliation(s)
- Tanya A Enderli
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, FL, USA,
| | - Stephanie R Burtch
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, FL, USA,
| | - Jara N Templet
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, FL, USA,
| | - Alessandra Carriero
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, FL, USA,
| |
Collapse
|
47
|
Belinsky GS, Sreekumar B, Andrejecsk JW, Saltzman WM, Gong J, Herzog RI, Lin S, Horsley V, Carpenter TO, Chung C. Pigment epithelium-derived factor restoration increases bone mass and improves bone plasticity in a model of osteogenesis imperfecta type VI via Wnt3a blockade. FASEB J 2016; 30:2837-48. [PMID: 27127101 DOI: 10.1096/fj.201500027r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 04/18/2016] [Indexed: 12/23/2022]
Abstract
Null mutations in for pigment epithelium-derived factor (PEDF), the protein product of the SERPINF1 gene, are the cause of osteogenesis imperfecta (OI) type VI. The PEDF-knockout (KO) mouse captures crucial elements of the human disease, including diminished bone mineralization and propensity to fracture. Our group and others have demonstrated that PEDF directs human mesenchymal stem cell (hMSC) commitment to the osteoblast lineage and modulates Wnt/β-catenin signaling, a major regulator of bone development; however, the ability of PEDF to restore bone mass in a mouse model of OI type VI has not been determined. In this study, PEDF delivery increased trabecular bone volume/total volume by 52% in 6-mo-old PEDF-KO mice but not in wild-type mice. In young (19-d-old) PEDF-KO mice, PEDF restoration increased bone volume fraction by 35% and enhanced biomechanical parameters of bone plasticity. A Wnt-green fluorescent protein reporter demonstrated dynamic changes in Wnt/β-catenin signaling characterized by early activation and marked suppression during terminal differentiation of hMSCs. Continuous Wnt3a exposure impeded mineralization of hMSCs, whereas the combination of Wnt3a and PEDF potentiated mineralization. Interrogation of the PEDF sequence identified a conserved motif found in other Wnt modulators, such as the dickkopf proteins. Mutation of a single amino acid on a 34-mer PEDF peptide increased mineralization of hMSC cultures compared with the native peptide sequence. These results indicate that PEDF counters Wnt signaling to allow for osteoblast differentiation and provides a mechanistic insight into how the PEDF null state results in OI type VI.-Belinsky, G. S., Sreekumar, B., Andrejecsk, J. W., Saltzman, W. M., Gong, J., Herzog, R. I., Lin, S., Horsley, V., Carpenter, T. O., Chung, C. Pigment epithelium-derived factor restoration increases bone mass and improves bone plasticity in a model of osteogenesis imperfecta type VI via Wnt3a blockade.
Collapse
Affiliation(s)
- Glenn S Belinsky
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bharath Sreekumar
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jillian W Andrejecsk
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Jingjing Gong
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Raimund I Herzog
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Samantha Lin
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Thomas O Carpenter
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chuhan Chung
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
48
|
Chen X, Li C, He T, Mao J, Li C, Lyu J, Meng QH. Metformin inhibits prostate cancer cell proliferation, migration, and tumor growth through upregulation of PEDF expression. Cancer Biol Ther 2016; 17:507-14. [PMID: 26987032 DOI: 10.1080/15384047.2016.1156273] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Metformin has been reported to inhibit the growth of various types of cancers, including prostate cancer. Yet the mode of anti-cancer action of metformin and the underlying mechanisms remain not fully elucidated. We hypothesized that the antitumorigenic effects of metformin are mediated through upregulation of pigment epithelium-derived factor (PEDF) expression in prostate cancer cells. In this report, metformin treatment significantly inhibited the proliferation and colony formation of prostate cancer cells, in a dose- and time-dependent manner. Meanwhile, Metformin markedly suppressed migration and invasion and induced apoptosis of both LNCaP and PC3 cancer cells. Metformin also reduced PC3 tumor growth in BALB/c nude mice in vivo. Furthermore, metformin treatment was associated with higher PEDF expression in both prostate cancer cells and tumor tissue. Taken together, metformin inhibits prostate cancer cell proliferation, migration, invasion and tumor growth, and these activities are mediated by upregulation of PEDF expression. These findings provide a novel insight into the molecular functions of metformin as an anticancer agent.
Collapse
Affiliation(s)
- Xiaowan Chen
- a Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University , Wenzhou , Zhejiang , China
| | - Chenli Li
- a Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University , Wenzhou , Zhejiang , China
| | - Tiantian He
- a Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University , Wenzhou , Zhejiang , China
| | - Jiating Mao
- a Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University , Wenzhou , Zhejiang , China
| | - Chunmei Li
- a Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University , Wenzhou , Zhejiang , China
| | - Jianxin Lyu
- a Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University , Wenzhou , Zhejiang , China
| | - Qing H Meng
- b Department of Laboratory Medicine , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
49
|
Rajagopal A, Homan EP, Joeng KS, Suzuki M, Bertin T, Cela R, Munivez E, Dawson B, Jiang MM, Gannon F, Crawford S, Lee BH. Restoration of the serum level of SERPINF1 does not correct the bone phenotype in Serpinf1 null mice. Mol Genet Metab 2016; 117:378-82. [PMID: 26693895 PMCID: PMC4788589 DOI: 10.1016/j.ymgme.2015.11.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/30/2015] [Accepted: 11/30/2015] [Indexed: 11/16/2022]
Abstract
Osteogenesis imperfecta (OI) is a group of genetic disorders characterized by bone fragility and deformity. OI type VI is unique owing to the mineralization defects observed in patient biopsies. Furthermore, it has been reported to respond less well to standard therapy with bisphosphonates [1]. Others and we have previously identified SERPINF1 mutations in patients with OI type VI. SERPINF1 encodes pigment epithelium derived factor (PEDF), a secreted collagen-binding glycoprotein that is absent in the sera of patients with OI type VI. Serpinf1 null mice show increased osteoid and decreased bone mass, and thus recapitulate the OI type VI phenotype. We tested whether restoration of circulating PEDF in the blood could correct the phenotype of OI type VI in the context of protein replacement. To do so, we utilized a helper-dependent adenoviral vector (HDAd) to express human SERPINF1 in the mouse liver and assessed whether PEDF secreted from the liver was able to rescue the bone phenotype observed in Serpinf1(-/-) mice. We confirmed that expression of SERPINF1 in the liver restored the serum level of PEDF. We also demonstrated that PEDF secreted from the liver was biologically active by showing the expected metabolic effects of increased adiposity and impaired glucose tolerance in Serpinf1(-/-) mice. Interestingly, overexpression of PEDF in vitro increased mineralization with a concomitant increase in the expression of bone gamma-carboxyglutamate protein, alkaline phosphatase and collagen, type I, alpha I, but the increased serum PEDF level did not improve the bone phenotype of Serpinf1(-/-) mice. These results suggest that PEDF may function in a context-dependent and paracrine fashion in bone homeostasis.
Collapse
Affiliation(s)
- Abbhirami Rajagopal
- Molecular and Human Genetics Department, Baylor College of Medicine, United States
| | - Erica P Homan
- Molecular and Human Genetics Department, Baylor College of Medicine, United States
| | - Kyu Sang Joeng
- Molecular and Human Genetics Department, Baylor College of Medicine, United States
| | - Masataka Suzuki
- Department of Medicine, Center for Cell and Gene Therapy, Baylor College of Medicine, United States
| | - Terry Bertin
- Molecular and Human Genetics Department, Baylor College of Medicine, United States
| | - Racel Cela
- Molecular and Human Genetics Department, Baylor College of Medicine, United States
| | - Elda Munivez
- Molecular and Human Genetics Department, Baylor College of Medicine, United States
| | - Brian Dawson
- Molecular and Human Genetics Department, Baylor College of Medicine, United States
| | - Ming-Ming Jiang
- Molecular and Human Genetics Department, Baylor College of Medicine, United States
| | - Frank Gannon
- Department of Pathology & Immunology, Baylor College of Medicine, United States
| | - Susan Crawford
- Department of Pathology, Saint Louis University, School of Medicine, United States
| | - Brendan H Lee
- Molecular and Human Genetics Department, Baylor College of Medicine, United States.
| |
Collapse
|
50
|
Yang L, Zhang Y, Cheng L, Yue D, Ma J, Zhao D, Hou X, Xiang R, Cheng P. Mesenchymal Stem Cells Engineered to Secrete Pigment Epithelium-Derived Factor Inhibit Tumor Metastasis and the Formation of Malignant Ascites in a Murine Colorectal Peritoneal Carcinomatosis Model. Hum Gene Ther 2016; 27:267-77. [PMID: 26756933 DOI: 10.1089/hum.2015.135] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The therapeutic effects of conventional treatments for advanced colorectal cancer with colorectal peritoneal carcinomatosis (CRPC) and malignant ascites are not very encouraging. Vascular endothelial growth factor-A/vascular permeability factors (VEGF-A/VPF) play key roles in the formation of malignant ascites. In previous work, we demonstrated that pigment epithelium-derived factor (PEDF) antagonized VEGF-A and could repress tumor growth and suppress metastasis in several cancer types. Thus, PEDF may be a therapeutic candidate for treating malignant ascites. Mesenchymal stem cells (MSCs) are promising tools for delivering therapeutic agents in cancer treatment. In the study, MSCs derived from bone marrow were efficiently engineered to secrete human PEDF by adenoviral transduction. Then, intraperitoneal Ad-PEDF-transduced MSCs were analyzed with respect to CRPC and malignant ascites in a CT26 CRPC model. MSCs engineered to secrete PEDF through adenoviral transduction significantly inhibited tumor metastasis and malignant ascites formation in CT26 CRPC mice. Antitumor mechanisms of MSCs-PEDF (MSCs transduced with Ad-PEDF: MOI 500) were associated with inhibiting tumor angiogenesis, inducing apoptosis, and restoring the VEGF-A/sFLT-1 ratio in ascites. Moreover, MSC-mediated Ad-PEDF delivery reduced production of adenovirus-neutralizing antibodies, prolonged PEDF expression, and induced MSCs-PEDF migration toward tumor cells. As a conclusion, MSCs engineered to secrete PEDF by adenoviral transduction may be a therapeutic approach for suppressing tumor metastasis and inhibiting malignant ascites production in CRPC.
Collapse
Affiliation(s)
- Liping Yang
- 1 Tumor Biotherapy Center/Key Laboratory of Biological Therapy and Regenerative Medicine Transformation, Gansu Province, Donggang Branch of The First Hospital of Lanzhou University, Lanzhou, People's Republic of China
| | - Yuwei Zhang
- 2 Division of Endocrinology and Metabolism, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Liuliu Cheng
- 3 State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy/Cancer Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Dan Yue
- 3 State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy/Cancer Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Jinhu Ma
- 3 State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy/Cancer Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Da Zhao
- 4 Oncology Medicine Department, Donggang Branch of The First Hospital of Lanzhou University, Lanzhou, People's Republic of China
| | - Xiaoming Hou
- 4 Oncology Medicine Department, Donggang Branch of The First Hospital of Lanzhou University, Lanzhou, People's Republic of China
| | - Rong Xiang
- 5 School of Medicine/Collaborative Innovation Center for Biotherapy, Nankai University , Tianjin, People's Republic of China
| | - Ping Cheng
- 3 State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy/Cancer Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|