1
|
Gauthier C, El Cheikh K, Basile I, Daurat M, Morère E, Garcia M, Maynadier M, Morère A, Gary-Bobo M. Cation-independent mannose 6-phosphate receptor: From roles and functions to targeted therapies. J Control Release 2024; 365:759-772. [PMID: 38086445 DOI: 10.1016/j.jconrel.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
The cation-independent mannose 6-phosphate receptor (CI-M6PR) is a ubiquitous transmembrane receptor whose main intracellular role is to direct enzymes carrying mannose 6-phosphate moieties to lysosomal compartments. Recently, the small membrane-bound portion of this receptor has appeared to be implicated in numerous pathophysiological processes. This review presents an overview of the main ligand partners and the roles of CI-M6PR in lysosomal storage diseases, neurology, immunology and cancer fields. Moreover, this membrane receptor has already been noted for its strong potential in therapeutic applications thanks to its cellular internalization activity and its ability to address pathogenic factors to lysosomes for degradation. A number of therapeutic delivery approaches using CI-M6PR, in particular with enzymes, antibodies or nanoparticles, are currently being proposed.
Collapse
Affiliation(s)
- Corentin Gauthier
- NanoMedSyn, Montpellier, France; IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | | | | - Elodie Morère
- NanoMedSyn, Montpellier, France; IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | | - Alain Morère
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | | |
Collapse
|
2
|
Jaiswal P, Tripathi V, Nayak A, Kataria S, Lukashevich V, Das A, Parmar HS. A molecular link between diabetes and breast cancer: Therapeutic potential of repurposing incretin-based therapies for breast cancer. Curr Cancer Drug Targets 2021; 21:829-848. [PMID: 34468298 DOI: 10.2174/1568009621666210901101851] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
Female breast cancer recently surpassed lung cancer and became the most commonly diagnosed cancer worldwide. As per the recent data from WHO, breast cancer accounts for one out of every 8 cancer cases diagnosed among an estimated 2.3 million new cancer cases. Breast cancer is the most prevailing cancer type among women causing the highest number of cancer-related mortality. It has been estimated that in 2020, 68,5000 women died due to this disease. Breast cancers have varying degrees of molecular heterogeneity; therefore, they are divided into various molecular clinical sub types. Recent reports suggest that type 2 diabetes (one of the common chronic diseases worldwide) is linked to the higher incidence, accelerated progression, and aggressiveness of different cancers; especially breast cancer. Breast cancer is hormone-dependent in nature and has a cross-talk with metabolism. A number of antidiabetic therapies are known to exert beneficial effects on various types of cancers, including breast cancer. However, only a few reports are available on the role of incretin-based antidiabetic therapies in cancer as a whole and in breast cancer in particular. The present review sheds light on the potential of incretin based therapies on breast cancer and explores the plausible underlying mechanisms. Additionally, we have also discussed the sub types of breast cancer as well as the intricate relationship between diabetes and breast cancer.
Collapse
Affiliation(s)
- Pooja Jaiswal
- School of Biotechnology, Devi Ahilya University, Indore-452001. M.P., India
| | - Versha Tripathi
- School of Biotechnology, Devi Ahilya University, Indore-452001. M.P., India
| | - Aakruti Nayak
- School of Biotechnology, Devi Ahilya University, Indore-452001. M.P., India
| | - Shreya Kataria
- School of Biotechnology, Devi Ahilya University, Indore-452001. M.P., India
| | - Vladimir Lukashevich
- Institute of Physiology of the National Academy of Sciences of Belarus, Minsk-220072. Belarus
| | - Apurba Das
- Department of Chemical Sciences, IIT, Indore, Simrol, Indore, M.P., India
| | | |
Collapse
|
3
|
Miller JJ, Bohnsack RN, Olson LJ, Ishihara M, Aoki K, Tiemeyer M, Dahms NM. Tissue plasminogen activator is a ligand of cation-independent mannose 6-phosphate receptor and consists of glycoforms that contain mannose 6-phosphate. Sci Rep 2021; 11:8213. [PMID: 33859256 PMCID: PMC8050316 DOI: 10.1038/s41598-021-87579-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/30/2021] [Indexed: 01/18/2023] Open
Abstract
Plasmin is the key enzyme in fibrinolysis. Upon interaction with plasminogen activators, the zymogen plasminogen is converted to active plasmin. Some studies indicate plasminogen activation is regulated by cation-independent mannose 6-phosphate receptor (CI-MPR), a protein that facilitates lysosomal enzyme trafficking and insulin-like growth factor 2 downregulation. Plasminogen regulation may be accomplished by CI-MPR binding to plasminogen or urokinase plasminogen activator receptor. We asked whether other members of the plasminogen activation system, such as tissue plasminogen activator (tPA), also interact with CI-MPR. Because tPA is a glycoprotein with three N-linked glycosylation sites, we hypothesized that tPA contains mannose 6-phosphate (M6P) and binds CI-MPR in a M6P-dependent manner. Using surface plasmon resonance, we found that two sources of tPA bound the extracellular region of human and bovine CI-MPR with low-mid nanomolar affinities. Binding was partially inhibited with phosphatase treatment or M6P. Subsequent studies revealed that the five N-terminal domains of CI-MPR were sufficient for tPA binding, and this interaction was also partially mediated by M6P. The three glycosylation sites of tPA were analyzed by mass spectrometry, and glycoforms containing M6P and M6P-N-acetylglucosamine were identified at position N448 of tPA. In summary, we found that tPA contains M6P and is a CI-MPR ligand.
Collapse
Affiliation(s)
- James J Miller
- Department of Biochemistry, Medical College of Wisconsin, 8701 W. Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Richard N Bohnsack
- Department of Biochemistry, Medical College of Wisconsin, 8701 W. Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Linda J Olson
- Department of Biochemistry, Medical College of Wisconsin, 8701 W. Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Mayumi Ishihara
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA.
| | - Nancy M Dahms
- Department of Biochemistry, Medical College of Wisconsin, 8701 W. Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| |
Collapse
|
4
|
Buchroithner B, Spurný P, Mayr S, Heitz J, Sivun D, Jacak J, Ludwig J. An Improved Transwell Design for Microelectrode Ion-Flux Measurements. MICROMACHINES 2021; 12:273. [PMID: 33800830 PMCID: PMC7998906 DOI: 10.3390/mi12030273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 11/16/2022]
Abstract
The microelectrode ion flux estimation (MIFE) is a powerful, non-invasive electrophysiological method for cellular membrane transport studies. Usually, the MIFE measurements are performed in a tissue culture dish or directly with tissues (roots, parts of the plants, and cell tissues). Here, we present a transwell system that allows for MIFE measurements on a cell monolayer. We introduce a measurement window in the transwell insert membrane, which provides direct access for the cells to the media in the upper and lower compartment of the transwell system and allows direct cell-to-cell contact coculture. Three-dimensional multiphoton lithography (MPL) was used to construct a 3D grid structure for cell support in the measurement window. The optimal polymer grid constant was found for implementation in transwell MIFE measurements. We showed that human umbilical vein endothelial cells (HUVECs) efficiently grow and maintain their physiological response on top of the polymer structures.
Collapse
Affiliation(s)
- Boris Buchroithner
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstr. 21, 4020 Linz, Austria; (B.B.); (S.M.); (J.J.)
| | - Pavel Spurný
- Institute of Microbiology of the Czech Academy of Sciences, Zamek 136, 37333 Nove Hrady, Czech Republic; (P.S.); (J.L.)
| | - Sandra Mayr
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstr. 21, 4020 Linz, Austria; (B.B.); (S.M.); (J.J.)
| | - Johannes Heitz
- Institute of Applied Physics, Johannes Kepler University Linz, Altenberger Str. 69, 4040 Linz, Austria;
| | - Dmitry Sivun
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstr. 21, 4020 Linz, Austria; (B.B.); (S.M.); (J.J.)
| | - Jaroslaw Jacak
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstr. 21, 4020 Linz, Austria; (B.B.); (S.M.); (J.J.)
| | - Jost Ludwig
- Institute of Microbiology of the Czech Academy of Sciences, Zamek 136, 37333 Nove Hrady, Czech Republic; (P.S.); (J.L.)
| |
Collapse
|
5
|
Al-Share B, Hammad N, Diab M. Pancreatic adenocarcinoma: molecular drivers and the role of targeted therapy. Cancer Metastasis Rev 2021; 40:355-371. [PMID: 33398620 DOI: 10.1007/s10555-020-09948-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/15/2020] [Indexed: 01/05/2023]
Abstract
Prognosis from pancreatic ductal adenocarcinoma (PDAC) continues to be poor despite the many efforts channeled to improve its management. Although the mainstay treatment is still traditional chemotherapy, recent advances highlighted a promising potential for targeted therapy in the management of this disease. Those advances emphasize the significance of timely genomic profiling of tumor tissue as well as germline testing of patients to identify potential markers of targeted therapy. While targeted therapy is reserved for a relatively small subset of patients with PDAC, ongoing research is uncovering additional markers, and targeted agents, that will hopefully translate to better outcomes for patients.
Collapse
Affiliation(s)
- Bayan Al-Share
- Department of Oncology, Wayne State University, Karmanos Cancer Institute, Detroit, MI, USA
| | - Nour Hammad
- Department of Oncology, Ascension Providence Hospital and Medical Center/Michigan State University/Collage of Human Medicine, Southfield, MI, USA
| | - Maria Diab
- Department of Oncology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
6
|
Lian S, Li S, Sah DK, Kim NH, Lakshmanan VK, Jung YD. Suppression of Urokinase-Type Plasminogen Activator Receptor by Docosahexaenoic Acid Mediated by Heme Oxygenase-1 in 12- O-Tetradecanoylphorbol-13-Acetate-Induced Human Endothelial Cells. Front Pharmacol 2021; 11:577302. [PMID: 33381031 PMCID: PMC7768974 DOI: 10.3389/fphar.2020.577302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/15/2020] [Indexed: 11/28/2022] Open
Abstract
Urokinase-type plasminogen activator receptor (uPAR) plays a crucial role in inflammation and tumor metastasis. Docosahexaenoic acid (DHA), a representative omega-3 polyunsaturated fatty acid, has been shown to exhibit anti-inflammatory and anti-tumor properties. However, the mechanism by which DHA negatively regulates uPAR expression is not yet understood. The aim of this study was to investigate the effect of DHA on 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced uPAR expression and potential role of heme oxygenase-1 (HO-1) in DHA-induced inhibition of uPAR in human endothelial ECV304 cells. Results showed that TPA induced uPAR expression in a time dependent manner, while DHA inhibited uPAR expression in a concentration-dependent manner. Moreover, treatment with DHA induced HO-1 expression in a time- and concentration-dependent manner. In addition, DHA-induced inhibition of uPAR expression and cell invasion in TPA-stimulated cells was reversed by si-HO-1 RNA. Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). Additionally, carbon monoxide, an HO-1 product, attenuated TPA-induced uPAR expression and cell invasion. Collectively, these data suggest a novel role of DHA-induced HO-1 in reducing uPAR expression and cell invasion in human endothelial ECV304 cells.
Collapse
Affiliation(s)
- Sen Lian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangdong, China
| | - Shinan Li
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Korea
| | - Dhiraj Kumar Sah
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Korea
| | - Nam Ho Kim
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Korea
| | - Vinoth-Kumar Lakshmanan
- Centre for Preclinical and Translational Medical Research (CPTMR), Central Research Facility (CRF), Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, India.,Thumbay Research Institute for Precision Medicine and Department of Biomedical Sciences, Gulf Medical University, Ajman, United Arab Emirates
| | - Young Do Jung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
7
|
Almeria C, Weiss R, Roy M, Tripisciano C, Kasper C, Weber V, Egger D. Hypoxia Conditioned Mesenchymal Stem Cell-Derived Extracellular Vesicles Induce Increased Vascular Tube Formation in vitro. Front Bioeng Biotechnol 2019; 7:292. [PMID: 31709251 PMCID: PMC6819375 DOI: 10.3389/fbioe.2019.00292] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) display a variety of therapeutically relevant effects, such as the induction of angiogenesis, particularly under hypoxic conditions. It is generally recognized that MSCs exert their effects by secretion of paracrine factors and by stimulation of host cells. Furthermore, there is increasing evidence that some therapeutically relevant effects of MSCs are mediated by MSC-derived extracellular vesicles (EVs). Since our current knowledge on MSC-derived EVs released under hypoxic conditions is very limited, we aimed to characterize MSC-derived EVs from normoxic vs. hypoxic conditions (5% O2). Adipose-derived MSCs were grown under normoxic and hypoxic conditions, and EVs were analyzed by flow cytometry using lactadherin as a marker for EVs exposing phosphatidylserine, CD63 and CD81 as EV markers, as well as CD73 and CD90 as MSC surface markers. Particle concentration and size distribution were measured by nanoparticle tracking analysis (NTA), and the EV surface antigen signature was characterized using bead-based multiplex flow cytometry. Furthermore, we evaluated the potential of MSC-derived EVs obtained under hypoxic conditions to support angiogenesis using an in vitro assay with an hTERT-immortalized human umbilical vein endothelial cell (HUVEC) line. Proliferation and viability of MSCs were increased under hypoxic conditions. EV concentration, size, and surface signature did not differ significantly between normoxic and hypoxic conditions, with the exception of CD44, which was significantly upregulated on normoxic EVs. EVs from hypoxic conditions exhibited increased tube formation as compared to normoxic EVs or to the corresponding supernatants from both groups, indicating that tube formation is facilitated by EVs rather than by soluble factors. In conclusion, hypoxia conditioned MSC-derived EVs appear to be functionally more potent than normoxic MSC-derived EVs regarding the induction of angiogenesis.
Collapse
Affiliation(s)
- Ciarra Almeria
- Department of Biotechnology, University of Natural Resources and Life Science, Vienna, Austria
| | - René Weiss
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Department for Biomedical Research, Danube University Krems, Krems, Austria
| | - Michelle Roy
- Department of Biotechnology, University of Natural Resources and Life Science, Vienna, Austria
| | - Carla Tripisciano
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Department for Biomedical Research, Danube University Krems, Krems, Austria
| | - Cornelia Kasper
- Department of Biotechnology, University of Natural Resources and Life Science, Vienna, Austria
| | - Viktoria Weber
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Department for Biomedical Research, Danube University Krems, Krems, Austria
| | - Dominik Egger
- Department of Biotechnology, University of Natural Resources and Life Science, Vienna, Austria
| |
Collapse
|
8
|
Ohradanova-Repic A, Machacek C, Donner C, Mühlgrabner V, Petrovčíková E, Zahradníková A, Vičíková K, Hořejší V, Stockinger H, Leksa V. The mannose 6-phosphate/insulin-like growth factor 2 receptor mediates plasminogen-induced efferocytosis. J Leukoc Biol 2019; 105:519-530. [PMID: 30657605 PMCID: PMC6392118 DOI: 10.1002/jlb.1ab0417-160rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 12/17/2018] [Accepted: 12/24/2018] [Indexed: 12/14/2022] Open
Abstract
The plasminogen system is harnessed in a wide variety of physiological processes, such as fibrinolysis, cell migration, or efferocytosis; and accordingly, it is essential upon inflammation, tissue remodeling, wound healing, and for homeostatic maintenance in general. Previously, we identified a plasminogen receptor in the mannose 6-phosphate/insulin-like growth factor 2 receptor (M6P/IGF2R, CD222). Here, we demonstrate by means of genetic knockdown, knockout, and rescue approaches combined with functional studies that M6P/IGF2R is up-regulated on the surface of macrophages, recognizes plasminogen exposed on the surface of apoptotic cells, and mediates plasminogen-induced efferocytosis. The level of uptake of plasminogen-coated apoptotic cells inversely correlates with the TNF-α production by phagocytes indicating tissue clearance without inflammation by this mechanism. Our results reveal an up-to-now undetermined function of M6P/IGF2R in clearance of apoptotic cells, which is crucial for tissue homeostasis.
Collapse
Affiliation(s)
- Anna Ohradanova-Repic
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Christian Machacek
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Clemens Donner
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Vanessa Mühlgrabner
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Eva Petrovčíková
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovak Republic.,Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Alexandra Zahradníková
- Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovak Republic.,Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Kristína Vičíková
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Václav Hořejší
- Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Hannes Stockinger
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Vladimir Leksa
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria.,Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| |
Collapse
|
9
|
Zhang H, Peng C, Huang H, Lai Y, Hu C, Li F, Wang D. Effects of amiloride on physiological activity of stem cells of human lung cancer and possible mechanism. Biochem Biophys Res Commun 2018; 504:1-5. [PMID: 29958884 DOI: 10.1016/j.bbrc.2018.06.138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 06/25/2018] [Indexed: 02/07/2023]
Abstract
Lung cancer is a common malignant tumor, the cancer stem cells (CSCs) were regarded responsible for the development of cancer tissue. The effects of amiloride on lung cancer stem cells and the possible mechanism were not much investigated. In this study, human NCI-H1975 lung CSCs were selected by flow cytometry, and the effects of amiloride at different concentrations (0, 12.5, 25, 50, and 100 μmol/L) were evaluated on proliferation, migration, invasion and apoptosis of CSCs using cell counting kit-8 and Transwell migration assays as well as flow cytometry. Wstern blot analysis was performed to investigate the effect of amiloride on the level of proteins in uPA system, NF-kB pathway, and PI3K-AKT-mTOR pathway in CSCs. As a result, we found that amiloride inhibited proliferation, migration and invasion of lung CSCs, and promoted apoptosis. Further, we found that amiloride decreased levels of target proteins in the uPA system, as well as the NF-kB and PI3K-AKT-mTOR pathways. These results indicated that amiloride could inhibit proliferation, migration and invasion of lung CSCs, and promotes apoptosis, these effects may be related to decreased levels of proteins in the uPA system, the NF-kB pathway, and the PI3K-AKT-mTOR pathway.
Collapse
Affiliation(s)
- Hengshuo Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Caixia Peng
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - He Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Yongxin Lai
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Chenchen Hu
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Fei Li
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Degui Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
10
|
HGF promotes HTR-8/SVneo cell migration through activation of MAPK/PKA signaling leading to up-regulation of WNT ligands and integrins that target β-catenin. Mol Cell Biochem 2018; 453:11-32. [DOI: 10.1007/s11010-018-3428-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/16/2018] [Indexed: 02/01/2023]
|
11
|
Xia Y, Yuan M, Li S, Thuan UT, Nguyen TT, Kang TW, Liao W, Lian S, Jung YD. Apigenin Suppresses the IL-1β-Induced Expression of the Urokinase-Type Plasminogen Activator Receptor by Inhibiting MAPK-Mediated AP-1 and NF-κB Signaling in Human Bladder Cancer T24 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:7663-7673. [PMID: 29945448 DOI: 10.1021/acs.jafc.8b02351] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The urokinase-type plasminogen activator receptor (uPAR), a glycoprotein localized on the cell surface with a glycosylphosphatidylinositol anchor, plays a crucial role in cell invasion, and the metastasis of several cancers, including bladder cancer, and its expression are significantly negatively correlated with patient survival rates. Apigenin, a naturally produced phytochemical compound found in fruits, vegetables, and plant leaves, has been shown to mediate a variety of cancer-metastasis-related molecules in various cancers. The effect of apigenin on uPAR expression is still unknown. In this study, we examined the effects of apigenin on IL-1β-induced uPAR expression and investigated its potential mechanisms. We discovered in this study that IL-1β could remarkably induce uPAR expression in bladder cancer T24 cells and that apigenin-inhibited IL-1β could induce uPAR expression concentration-dependently. Interestingly, NF-κB and AP-1 transcription factors were critically required for IL-1β-induced high uPAR expression. Apigenin suppressed the transcriptional activity of both AP-1 and NF-κB by inhibiting ERK1/2 and JNK signaling pathways. These results suggest that apigenin can exert anti-invasion effects by inhibiting uPAR expression via mediating (ERK1/2, JNK)/AP-1 and (ERK1/2, JNK)/NF-κB signaling pathways in human T24 cells. Our present study generated novel and valuable biological insight into anti-invasion through treatment with a small native compound.
Collapse
Affiliation(s)
- Yong Xia
- Research Institute of Medical Sciences , Chonnam National University Medical School , Gwangju 501-190 , Republic of Korea
- Department of Urology , New York University School of Medicine , New York , New York 10016 , United States
| | | | - Shinan Li
- Research Institute of Medical Sciences , Chonnam National University Medical School , Gwangju 501-190 , Republic of Korea
| | - Ung Trong Thuan
- Research Institute of Medical Sciences , Chonnam National University Medical School , Gwangju 501-190 , Republic of Korea
| | - Thi Thinh Nguyen
- Research Institute of Medical Sciences , Chonnam National University Medical School , Gwangju 501-190 , Republic of Korea
| | - Taek Won Kang
- Research Institute of Medical Sciences , Chonnam National University Medical School , Gwangju 501-190 , Republic of Korea
| | | | - Sen Lian
- Guangdong Provincial Key Laboratory of Biochip , Guangzhou , 510515 , Guangdong , China
| | - Young Do Jung
- Research Institute of Medical Sciences , Chonnam National University Medical School , Gwangju 501-190 , Republic of Korea
| |
Collapse
|
12
|
Zwirzitz A, Reiter M, Skrabana R, Ohradanova-Repic A, Majdic O, Gutekova M, Cehlar O, Petrovčíková E, Kutejova E, Stanek G, Stockinger H, Leksa V. Lactoferrin is a natural inhibitor of plasminogen activation. J Biol Chem 2018; 293:8600-8613. [PMID: 29669808 DOI: 10.1074/jbc.ra118.003145] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/07/2018] [Indexed: 12/31/2022] Open
Abstract
The plasminogen system is essential for dissolution of fibrin clots, and in addition, it is involved in a wide variety of other physiological processes, including proteolytic activation of growth factors, cell migration, and removal of protein aggregates. On the other hand, uncontrolled plasminogen activation contributes to many pathological processes (e.g. tumor cells' invasion in cancer progression). Moreover, some virulent bacterial species (e.g. Streptococci or Borrelia) bind human plasminogen and hijack the host's plasminogen system to penetrate tissue barriers. Thus, the conversion of plasminogen to the active serine protease plasmin must be tightly regulated. Here, we show that human lactoferrin, an iron-binding milk glycoprotein, blocks plasminogen activation on the cell surface by direct binding to human plasminogen. We mapped the mutual binding sites to the N-terminal region of lactoferrin, encompassed also in the bioactive peptide lactoferricin, and kringle 5 of plasminogen. Finally, lactoferrin blocked tumor cell invasion in vitro and also plasminogen activation driven by Borrelia Our results explain many diverse biological properties of lactoferrin and also suggest that lactoferrin may be useful as a potential tool for therapeutic interventions to prevent both invasive malignant cells and virulent bacteria from penetrating host tissues.
Collapse
Affiliation(s)
| | - Michael Reiter
- From the Institute for Hygiene and Applied Immunology and
| | - Rostislav Skrabana
- the Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, and
| | | | - Otto Majdic
- Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, A-1090 Vienna, Austria and
| | - Marianna Gutekova
- the Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava 814 38, Slovak Republic
| | - Ondrej Cehlar
- the Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, and
| | - Eva Petrovčíková
- the Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava 814 38, Slovak Republic
| | - Eva Kutejova
- the Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava 814 38, Slovak Republic
| | - Gerold Stanek
- From the Institute for Hygiene and Applied Immunology and
| | | | - Vladimir Leksa
- From the Institute for Hygiene and Applied Immunology and .,the Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava 814 38, Slovak Republic
| |
Collapse
|
13
|
Liu R, Heiss EH, Waltenberger B, Blažević T, Schachner D, Jiang B, Krystof V, Liu W, Schwaiger S, Peña-Rodríguez LM, Breuss JM, Stuppner H, Dirsch VM, Atanasov AG. Constituents of Mediterranean Spices Counteracting Vascular Smooth Muscle Cell Proliferation: Identification and Characterization of Rosmarinic Acid Methyl Ester as a Novel Inhibitor. Mol Nutr Food Res 2018; 62:e1700860. [PMID: 29405576 DOI: 10.1002/mnfr.201700860] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/23/2018] [Indexed: 12/15/2022]
Abstract
SCOPE Aberrant vascular smooth muscle cell (VSMC) proliferation is involved in atherosclerotic plaque formation and restenosis. Mediterranean spices have been reported to confer cardioprotection, but their direct influence on VSMCs has largely not been investigated. This study aims at examining rosmarinic acid (RA) and 11 related constituents for inhibition of VSMC proliferation in vitro, and at characterizing the most promising compound for their mode of action and influence on neointima formation in vivo. METHODS AND RESULTS RA, rosmarinic acid methyl ester (RAME), and caffeic acid methyl ester inhibit VSMC proliferation in a resazurin conversion assay with IC50 s of 5.79, 3.12, and 6.78 µm, respectively. RAME significantly reduced neointima formation in vivo in a mouse femoral artery cuff model. Accordingly, RAME leads to an accumulation of VSMCs in the G0 /G1 cell-cycle phase, as indicated by blunted retinoblastoma protein phosphorylation upon mitogen stimulation and inhibition of cyclin-dependent kinase 2 in vitro. CONCLUSION RAME represses PDGF-induced VSMC proliferation in vitro and reduces neointima formation in vivo. These results recommend RAME as an interesting compound with VSMC-inhibiting potential. Future metabolism and pharmacokinetics studies might help to further evaluate the potential relevance of RAME and other spice-derived polyphenolics for vasoprotection.
Collapse
MESH Headings
- Animals
- Cardiovascular Agents/adverse effects
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/therapeutic use
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Cinnamates/administration & dosage
- Cinnamates/adverse effects
- Cinnamates/pharmacology
- Cinnamates/therapeutic use
- Depsides/administration & dosage
- Depsides/adverse effects
- Depsides/pharmacology
- Depsides/therapeutic use
- Diet, Mediterranean
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Human Umbilical Vein Endothelial Cells/cytology
- Humans
- Male
- Mediterranean Region
- Methylation
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Random Allocation
- Rats
- Retinoblastoma Protein/metabolism
- Rosmarinus/chemistry
- Rosmarinus/growth & development
- Spices/analysis
- Rosmarinic Acid
Collapse
Affiliation(s)
- Rongxia Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, University of Yantai, Yantai, China
| | - Elke H Heiss
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Tina Blažević
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Daniel Schachner
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Baohong Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Vladimir Krystof
- Laboratory of Growth Regulators, Palacky University & Institute of Experimental Botany AS CR, Olomouc, Czech Republic
| | - Wanhui Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, University of Yantai, Yantai, China
| | - Stefan Schwaiger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Luis M Peña-Rodríguez
- Unidad de Biotecnología, Centro de investigación Científica de Yucatán, Mérida, México
| | - Johannes M Breuss
- Center for Physiology and Pharmacology, Institute for Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Hermann Stuppner
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Verena M Dirsch
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Atanas G Atanasov
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
| |
Collapse
|
14
|
Biotin-Chasing Assay to Evaluate uPAR Stability and Cleavage on the Surface of Cells. Methods Mol Biol 2018. [PMID: 29318541 DOI: 10.1007/978-1-4939-7595-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The plasminogen activation system, i.e., the fibrinolytic system, is one of the major plasma proteolytic pathways. The proteolytic conversion of the zymogen plasminogen to the active serine protease plasmin is on the cell surface catalyzed by the serine protease urokinase-type plasminogen activator (urokinase, uPA). Upon binding to the urokinase receptor (uPAR, CD87), single-chain pro-uPA is processed to double-chain uPA which in turn specifically converts cell-bound plasminogen to plasmin. Plasmin is harnessed in many physiological processes, e.g., blood clots' resolution, or proteolytic activation of growth factors. Plasmin is essential also for migratory cells, for instance, activated immune cells; however, malignant cells hijack plasmin for invasion as well. The activation of plasminogen to plasmin is thus at the physiological level tightly controlled. One of the negative regulators of plasminogen activation has been identified in the cation-independent mannose 6-phosphate/insulin-like growth factor 2 receptor (M6P/IGF2R, CIMPR, CD222). M6P/IGF2R is a multifunctional receptor involved in protein sorting, internalization, and degradation, being considered a tumor suppressor. M6P/IGF2R binds both plasminogen and uPAR and facilitates in this way the proteolytic cleavage of uPAR resulting in the loss of the uPA binding on the cell surface. Hence, this molecular device contributes to the negative feedback loop in regulation of pericellular plasminogen activation and cell invasion.In this chapter, we describe the experimental approach, i.e., biotin-chasing assay, to evaluate uPAR stability and cleavage on the surface of cells.
Collapse
|
15
|
Malainer C, Schachner D, Sangiovanni E, Atanasov AG, Schwaiger S, Stuppner H, Heiss EH, Dirsch VM. Eurycomalactone Inhibits Expression of Endothelial Adhesion Molecules at a Post-Transcriptional Level. JOURNAL OF NATURAL PRODUCTS 2017; 80:3186-3193. [PMID: 29148754 PMCID: PMC5744186 DOI: 10.1021/acs.jnatprod.7b00503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The C-19 quassinoid eurycomalactone (1) has recently been shown to be a potent (IC50 = 0.5 μM) NF-κB inhibitor in a luciferase reporter model. In this study, we show that 1 with similar potency inhibited the expression of the NF-κB-dependent target genes ICAM-1, VCAM-1, and E-selectin in TNFα-activated human endothelial cells (HUVECtert) by flow cytometry experiments. Surprisingly, 1 (2 μM) did not inhibit TNFα-induced IKKα/β or IκBα phosphorylation significantly. Also, the TNFα-induced degradation of IκBα remained unchanged in response to 1 (2 μM). In addition, pretreatment of HUVECtert with 1 (2 μM) had no statistically significant effect on TNFα-mediated nuclear translocation of the NF-κB subunit p65 (RelA). Quantitative RT-PCR revealed that 1 (0.5-5 μM) exhibited diverse effects on the TNFα-induced transcription of ICAM-1, VCAM-1, and SELE genes since the mRNA level either remained unchanged (ICAM-1, E-selectin, and VCAM-1 at 0.5 μM 1), was reduced (VCAM-1 at 5 μM 1), or even increased (E-selectin at 5 μM 1). Finally, the time-dependent depletion of a short-lived protein (cyclin D1) as well as the measurement of de novo protein synthesis in the presence of 1 (2-5 μM) suggested that 1 might act as a protein synthesis inhibitor rather than an inhibitor of early NF-κB signaling.
Collapse
Affiliation(s)
- Clemens Malainer
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Daniel Schachner
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Enrico Sangiovanni
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
- Department
of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy
| | - Atanas G. Atanasov
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
- Institute
of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Stefan Schwaiger
- Institute
of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck, University Innsbruck, Innrain 80/82, Innsbruck 6020, Austria
| | - Hermann Stuppner
- Institute
of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck, University Innsbruck, Innrain 80/82, Innsbruck 6020, Austria
| | - Elke H. Heiss
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Verena M. Dirsch
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
- Tel: +43-1-4277-55270. Fax: +43-1-4277-55969. E-mail:
| |
Collapse
|
16
|
Nikzad-Langerodi R, Ortmann S, Pferschy-Wenzig E, Bochkov V, Zhao Y, Miao J, Saukel J, Ladurner A, Heiss E, Dirsch V, Bauer R, Atanasov A. Assessment of anti-inflammatory properties of extracts from Honeysuckle (Lonicera sp. L., Caprifoliaceae) by ATR-FTIR spectroscopy. Talanta 2017; 175:264-272. [DOI: 10.1016/j.talanta.2017.07.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 12/16/2022]
|
17
|
Leksa V, Ilková A, Vičíková K, Stockinger H. Unravelling novel functions of the endosomal transporter mannose 6-phosphate/insulin-like growth factor receptor (CD222) in health and disease: An emerging regulator of the immune system. Immunol Lett 2017; 190:194-200. [PMID: 28823520 DOI: 10.1016/j.imlet.2017.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/04/2017] [Accepted: 08/10/2017] [Indexed: 02/02/2023]
Abstract
Properly balanced cellular responses require both the mutual interactions of soluble factors with cell surface receptors and the crosstalk of intracellular molecules. In particular, immune cells exposed unceasingly to an array of positive and negative stimuli must distinguish between what has to be tolerated and attacked. Protein trafficking is one of crucial pathways involved in this labour. The approximately >270-kDa protein transporter called mannose 6- phosphate/insulin-like growth factor 2 receptor (M6P/IGF2R, CD222) is a type I transmembrane glycoprotein present largely intracellularly in the Golgi apparatus and endosomal compartments, but also at the cell surface. It is expressed ubiquitously in a vast majority of higher eukaryotic cell types. Through binding and trafficking multiple unrelated extracellular and intracellular ligands, CD222 is involved in the regulation of a plethora of functions, and thus implicated in many physiological but also pathophysiological conditions. This review describes, first, general features of CD222, such as its evolution, genomic structure and regulation, protein structure and ligands; and second, its specific functions with a special focus on the immune system.
Collapse
Affiliation(s)
- Vladimir Leksa
- Centre for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria; Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | - Antónia Ilková
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Kristína Vičíková
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Hannes Stockinger
- Centre for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| |
Collapse
|
18
|
Lončar-Brzak B, Klobučar M, Veliki-Dalić I, Sabol I, Kraljević Pavelić S, Krušlin B, Mravak-Stipetić M. Expression of small leucine-rich extracellular matrix proteoglycans biglycan and lumican reveals oral lichen planus malignant potential. Clin Oral Investig 2017; 22:1071-1082. [PMID: 28779221 DOI: 10.1007/s00784-017-2190-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 07/21/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The aim of this study was to examine molecular alterations on the protein level in lesions of oral lichen planus (OLP), oral squamous cell carcinoma (OSCC) and healthy mucosa. MATERIALS AND METHODS Global protein profiling methods based on liquid chromatography coupled to mass spectrometry (LC-MS) were used, with a special emphasis on evaluation of deregulated extracellular matrix molecules expression, as well as on analyses of IG2F and IGFR2 expression in healthy mucosa, OLP and OSCC tissues by comparative semi-quantitative immunohistochemistry. RESULTS Mass spectrometry-based proteomics profiling of healthy mucosa, OLP and OSCC tissues (and accompanied histologically unaltered tissues, respectively) identified 55 extracellular matrix proteins. Twenty among identified proteins were common to all groups of samples. Expression of small leucine-rich extracellular matrix proteoglycans lumican and biglycan was found both in OSCC and OLP and they were validated by Western blot analysis as putative biomarkers. A significant increase (p < 0.05) of biglycan expression in OLP-AT group was determined in comparison with OLP-T group, while lumican showed significant up-regulation (p < 0.05) in OLP-T and OSCC-T groups vs. adjacent and control tissue groups. Biglycan expression was only determined in OSCC-AT group. Immunohistochemical analysis of IGF2 and IG2FR expression revealed no significant difference among groups of samples. CONCLUSION/CLINICAL RELEVANCE Biglycan and lumican were identified as important pathogenesis biomarkers of OLP that point to its malignant potential.
Collapse
Affiliation(s)
- Božana Lončar-Brzak
- School of Dental Medicine, Department of Oral Medicine, University of Zagreb, Zagreb, Croatia
| | - Marko Klobučar
- Department of Biotechnology and Centre for High-throughput technologies, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia
| | - Irena Veliki-Dalić
- Department of Pathology, Clinical Hospital for Tumours, Clinical Hospital Centre Sisters of Mercy, Zagreb, Croatia
| | - Ivan Sabol
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Sandra Kraljević Pavelić
- Department of Biotechnology and Centre for High-throughput technologies, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia.
| | - Božo Krušlin
- School of Medicine, Department of Pathology, Clinical Hospital Centre Sisters of Mercy, University of Zagreb, Zagreb, Croatia
| | - Marinka Mravak-Stipetić
- School of Dental Medicine, Department of Oral Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
19
|
Liu R, Heiss EH, Sider N, Schinkovitz A, Gröblacher B, Guo D, Bucar F, Bauer R, Dirsch VM, Atanasov AG. Identification and characterization of [6]-shogaol from ginger as inhibitor of vascular smooth muscle cell proliferation. Mol Nutr Food Res 2015; 59:843-52. [PMID: 25631547 PMCID: PMC4573514 DOI: 10.1002/mnfr.201400791] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 01/28/2023]
Abstract
SCOPE Vascular smooth muscle cell (VSMC) proliferation is involved in the pathogenesis of cardiovascular disease, making the identification of new counteracting agents and their mechanisms of action relevant. Ginger and its constituents have been reported to improve cardiovascular health, but no studies exist addressing a potential interference with VSMC proliferation. METHODS AND RESULTS The dichloromethane extract of ginger inhibited VSMC proliferation when monitored by resazurin metabolic conversion (IC50 = 2.5 μg/mL). The examination of major constituents from ginger yielded [6]-shogaol as the most active compound (IC50 = 2.7 μM). In the tested concentration range [6]-shogaol did not exhibit cytotoxicity toward VSMC and did not interfere with endothelial cell proliferation. [6]-shogaol inhibited DNA synthesis and induced accumulation of the VSMC in the G0 /G1 cell-cycle phase accompanied with activation of the nuclear factor-erythroid 2-related factor 2 (Nrf2)/HO-1 pathway. Since [6]-shogaol lost its antiproliferative activity in the presence of the heme oxygenase-1 (HO-1) inhibitor tin protoporphyrin IX, HO-1 induction appears to contribute to the antiproliferative effect. CONCLUSION This study demonstrates for the first time inhibitory potential of ginger constituents on VSMC proliferation. The presented data suggest that [6]-shogaol exerts its antiproliferative effect through accumulation of cells in the G0 /G1 cell-cycle phase associated with activation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Rongxia Liu
- Department of Pharmacognosy, University of Vienna, Vienna, Austria; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Fakhrudin N, Waltenberger B, Cabaravdic M, Atanasov AG, Malainer C, Schachner D, Heiss EH, Liu R, Noha SM, Grzywacz AM, Mihaly-Bison J, Awad EM, Schuster D, Breuss JM, Rollinger JM, Bochkov V, Stuppner H, Dirsch VM. Identification of plumericin as a potent new inhibitor of the NF-κB pathway with anti-inflammatory activity in vitro and in vivo. Br J Pharmacol 2014; 171:1676-86. [PMID: 24329519 PMCID: PMC3966748 DOI: 10.1111/bph.12558] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 11/28/2013] [Accepted: 12/09/2013] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE The transcription factor NF-κB orchestrates many pro-inflammatory signals and its inhibition is considered a promising strategy to combat inflammation. Here we report the characterization of the natural product plumericin as a highly potent inhibitor of the NF-κB pathway with a novel chemical scaffold, which was isolated via a bioactivity-guided approach, from extracts of Himatanthus sucuuba, an Amazonian plant traditionally used to treat inflammation-related disorders. EXPERIMENTAL APPROACH A NF-κB luciferase reporter gene assay was used to identify NF-κB pathway inhibitors from H. sucuuba extracts. Monitoring of TNF-α-induced expression of the adhesion molecules VCAM-1, ICAM-1 and E-selectin by flow cytometry was used to confirm NF-κB inhibition in endothelial cells, and thioglycollate-induced peritonitis in mice to confirm effects in vivo. Western blotting and transfection experiments were used to investigate the mechanism of action of plumericin. KEY RESULTS Plumericin inhibited NF-κB-mediated transactivation of a luciferase reporter gene (IC50 1 μM), abolished TNF-α-induced expression of the adhesion molecules VCAM-1, ICAM-1 and E-selectin in endothelial cells and suppressed thioglycollate-induced peritonitis in mice. Plumericin exerted its NF-κB pathway inhibitory effect by blocking IκB phosphorylation and degradation. Plumericin also inhibited NF-κB activation induced by transfection with the constitutively active catalytic subunit of the IκB kinase (IKK-β), suggesting IKK involvement in the inhibitory action of this natural product. CONCLUSION AND IMPLICATIONS Plumericin is a potent inhibitor of NF-κB pathways with a new chemical scaffold. It could be further explored as a novel anti-inflammatory lead compound.
Collapse
Affiliation(s)
- N Fakhrudin
- Department of Pharmacognosy, University of Vienna, Vienna, Austria; Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bohnsack RN, Warejcka DJ, Wang L, Gillespie SR, Bernstein AM, Twining SS, Dahms NM. Expression of insulin-like growth factor 2 receptor in corneal keratocytes during differentiation and in response to wound healing. Invest Ophthalmol Vis Sci 2014; 55:7697-708. [PMID: 25358730 DOI: 10.1167/iovs.14-15179] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Insulin-like growth factor 2 receptor (IGF2R) associates with ligands that influence wound healing outcomes. However, the expression pattern of IGF2R and its role in the cornea is unknown. METHODS Human keratocytes were isolated from donor corneas. Fibroblasts (fibroblast growth factor 2 [FGF2]-treated) or myofibroblasts (TGF-β1-treated) were analyzed for IGF2R and α-smooth muscle actin (α-SMA) expression by Western blotting and immunolocalization. Mouse corneas were wounded in vivo and porcine corneas ex vivo. The IGF2R and α-SMA protein expression were visualized and quantified by immunohistochemistry. The IGF2R gene expression in human corneal fibroblasts was knocked-down with targeted lentiviral shRNA. RESULTS The IGF2R is expressed in epithelial and stromal cells of normal human, mouse, and porcine corneas. The IGF2R increases (11.2 ± 0.4-fold) in the epithelial and (11.7 ± 0.9-fold) stromal layers of in vivo wounded mouse corneas. Double-staining with α-SMA- and IGF2R-specific antibodies reveals that IGF2R protein expression is increased in stromal myofibroblasts in the wounded cornea relative to keratocytes in the normal cornea (11.2 ± 0.8-fold). Human primary stromal keratocytes incubated with FGF2 or TGF-β1 in vitro demonstrate increased expression (2.0 ± 0.4-fold) of IGF2R in myofibroblasts relative to fibroblasts. Conversion of IGF2R shRNA-lentiviral particle transduced corneal fibroblasts to myofibroblasts reveals a dependence on IGF2R expression, as only 40% ± 10% of cells transduced converted to myofibroblasts compared to 86% ± 3% in control cells. CONCLUSIONS The IGF2R protein expression is increased during corneal wound healing and IGF2R regulates human corneal fibroblast to myofibroblast differentiation.
Collapse
Affiliation(s)
- Richard N Bohnsack
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Debra J Warejcka
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Lingyan Wang
- Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | | | - Audrey M Bernstein
- Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Sally S Twining
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States Department of Ophthalmology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Nancy M Dahms
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
22
|
Pfisterer K, Forster F, Paster W, Supper V, Ohradanova-Repic A, Eckerstorfer P, Zwirzitz A, Donner C, Boulegue C, Schiller HB, Ondrovičová G, Acuto O, Stockinger H, Leksa V. The late endosomal transporter CD222 directs the spatial distribution and activity of Lck. THE JOURNAL OF IMMUNOLOGY 2014; 193:2718-32. [PMID: 25127865 DOI: 10.4049/jimmunol.1303349] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The spatial and temporal organization of T cell signaling molecules is increasingly accepted as a crucial step in controlling T cell activation. CD222, also known as the cation-independent mannose 6-phosphate/insulin-like growth factor 2 receptor, is the central component of endosomal transport pathways. In this study, we show that CD222 is a key regulator of the early T cell signaling cascade. Knockdown of CD222 hampers the effective progression of TCR-induced signaling and subsequent effector functions, which can be rescued via reconstitution of CD222 expression. We decipher that Lck is retained in the cytosol of CD222-deficient cells, which obstructs the recruitment of Lck to CD45 at the cell surface, resulting in an abundant inhibitory phosphorylation signature on Lck at the steady state. Hence, CD222 specifically controls the balance between active and inactive Lck in resting T cells, which guarantees operative T cell effector functions.
Collapse
Affiliation(s)
- Karin Pfisterer
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria
| | - Florian Forster
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria
| | - Wolfgang Paster
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria; Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Verena Supper
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria
| | - Anna Ohradanova-Repic
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria
| | - Paul Eckerstorfer
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria
| | - Alexander Zwirzitz
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria
| | - Clemens Donner
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria
| | - Cyril Boulegue
- Department of Molecular Medicine, Max-Planck Institute of Biochemistry, Martinsried 82152, Germany; and
| | - Herbert B Schiller
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria; Department of Molecular Medicine, Max-Planck Institute of Biochemistry, Martinsried 82152, Germany; and
| | - Gabriela Ondrovičová
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 84551 Bratislava, Slovak Republic
| | - Oreste Acuto
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Hannes Stockinger
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria;
| | - Vladimir Leksa
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna A-1090, Austria; Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 84551 Bratislava, Slovak Republic
| |
Collapse
|
23
|
Ou JM, Lian WS, Qiu MK, Dai YX, Dong Q, Shen J, Dong P, Wang XF, Liu YB, Quan ZW, Fei ZW. Knockdown of IGF2R suppresses proliferation and induces apoptosis in hemangioma cells in vitro and in vivo. Int J Oncol 2014; 45:1241-9. [PMID: 24968760 DOI: 10.3892/ijo.2014.2512] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 05/26/2014] [Indexed: 01/18/2023] Open
Abstract
Insulin-like growth factor-II (IGF-II)/IGF2R signaling plays a pivotal role in cell growth, migration and differentiation in many malignancies. An individual with high IGF-II expression levels has a high risk of developing cancer, but IGF2R is often considered to be a tumor suppressor. To date, little has been reported about the role of IGF-II/IGF2R signaling in hemangiomas (HAs). Thus, uncovering the mechanisms of IGF-II/IGF2R signaling is very important to understanding the development of HAs. In the present study, the expression of IGF-II and IGF2R was investigated in 27 cases of HAs of different phases by immunohistochemistry. Through lentivirus-mediated IGF2R siRNA (Lv-siIGF2R) in HA-derived endothelial cells (HDECs), we observed the effects of IGF2R knockdown on the biological behavior of HA cells. We found that the expression of IGF-II and IGF2R was significantly increased in proliferating phase HAs, but decreased in involuting phase HAs. Furthermore, knockdown of IGF2R in vitro significantly diminished the proliferative activity and induced apoptosis and cycle arrest with decreased expression of PCNA, Ki-67, Bcl-2, Cyclin D1 and E and increased the expression of Bax in the proliferative phase HAs (HDEC and CRL-2586 EOMA cells). In addition, the tumor volumes in a subcutaneous HDEC nude mouse model treated with Lv-siIGF2R were significantly smaller than those of the control group. Taken together, our findings indicate that the expression of IGF-II and IGF2R is increased in proliferating phase HAs, and knockdown of IGF2R suppresses proliferation and induces apoptosis in HA cells in vitro and in vivo, suggesting that IGF2R may represent a novel therapeutic target for the treatment of human HAs.
Collapse
Affiliation(s)
- J-M Ou
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - W-S Lian
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - M-K Qiu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Y-X Dai
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Q Dong
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - J Shen
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - P Dong
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - X-F Wang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Y-B Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Z-W Quan
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Z-W Fei
- Department of General Surgery, Xinhua Hospital (Chong Ming) Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 202150, P.R. China
| |
Collapse
|
24
|
Conde J, Tian F, Hernández Y, Bao C, Cui D, Janssen KP, Ibarra MR, Baptista PV, Stoeger T, de la Fuente JM. In vivo tumor targeting via nanoparticle-mediated therapeutic siRNA coupled to inflammatory response in lung cancer mouse models. Biomaterials 2013; 34:7744-53. [DOI: 10.1016/j.biomaterials.2013.06.041] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 06/23/2013] [Indexed: 11/28/2022]
|
25
|
Noh H, Hong S, Huang S. Role of urokinase receptor in tumor progression and development. Am J Cancer Res 2013; 3:487-95. [PMID: 23843896 PMCID: PMC3706692 DOI: 10.7150/thno.4218] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 08/15/2012] [Indexed: 12/21/2022] Open
Abstract
Elevated level of urokinase receptor (uPAR) is detected in various aggressive cancer types and is closely associated with poor prognosis of cancers. Binding of uPA to uPAR triggers the conversion of plasminogen to plasmin and the subsequent activation of metalloproteinases. These events confer tumor cells with the capability to degrade the components of the surrounding extracellular matrix, thus contributing to tumor cell invasion and metastasis. uPA-uPAR interaction also elicits signals that stimulate cell proliferation/survival and the expression of tumor-promoting genes, thus assisting tumor development. In addition to its interaction with uPA, uPAR also interacts with vitronectin and this interaction promotes cancer metastasis by activating Rac and stimulating cell migration. Although underlying mechanisms are yet to be fully elucidated, uPAR has been shown to facilitate epithelial-mesenchymal transition (EMT) and induce cancer stem cell-like properties in breast cancer cells. The fact that uPAR lacks intracellular domain suggests that its signaling must be mediated through its co-receptors. Indeed, uPAR interacts with diverse transmembrane proteins including integrins, ENDO180, G protein-coupled receptors and growth factor receptors in cancer cells and these interactions are proven to be critical for the role of uPAR in tumorigenesis. Inhibitory peptide that prevents uPA-uPAR interaction has shown the promise to prolong patients' survival in the early stage of clinical trial. The importance of uPAR's co-receptor in uPAR's tumor-promoting effects implicate that anti-cancer therapeutic agents may also be developed by disrupting the interactions between uPAR and its functional partners.
Collapse
|
26
|
Caixeiro NJ, Martin JL, Scott CD. Silencing the mannose 6-phosphate/IGF-II receptor differentially affects tumorigenic properties of normal breast epithelial cells. Int J Cancer 2013; 133:2542-50. [PMID: 23686499 DOI: 10.1002/ijc.28276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 05/07/2013] [Indexed: 11/09/2022]
Abstract
Although loss of the mannose 6-phosphate/insulin-like growth factor-II receptor (M6P/IGF-IIR) in breast cancer is believed to play a role in tumorigenesis, it has not been demonstrated that M6P/IGF-IIR loss is sufficient to confer a malignant phenotype in an untransformed cell. We investigated the impact of M6P/IGF-IIR silencing using phenotypically normal (MCF-10A) and oncogenically transformed (MCF-10T, the c-Ha-ras transformed derivative of MCF-10A) human breast epithelial cell lines as model systems. In both cell lines, silencing of M6P/IGF-IIR increased cell proliferation and motility, with the effects being more pronounced in MCF-10A cells. Although anchorage-independent growth was increased by M6P/IGF-IIR silencing in MCF-10T cells, MCF-10A cells did not acquire the ability to grow in soft agar. Conversely, reduced M6P/IGF-IIR expression increased the invasive potential of MCF-10A cells, but did not enhance the already high rate of invasion of MCF-10T cells. M6P/IGF-IIR silencing had no effect on basal or IGF-II-stimulated IGF-I receptor (IGF-IR) or AKT phosphorylation in either cell line, but both were abrogated by IGF-IR kinase inhibition, which also reduced the stimulatory effect of M6P/IGF-IIR silencing on proliferation under basal and IGF-II-stimulated conditions in both cell lines. However, cell motility was neither stimulated by IGF-II nor reduced by IGF-IR inhibition, suggesting that potentiation of specific tumorigenic features in response to M6P/IGF-IIR silencing involves IGF-II- dependent and -independent mechanisms. Collectively, these data suggest that M6P/IGF-IIR silencing alone is insufficient to confer a tumorigenic phenotype, but can enhance tumorigenicity in an already transformed cell.
Collapse
Affiliation(s)
- Nicole J Caixeiro
- Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St Leonards, NSW, Australia
| | | | | |
Collapse
|
27
|
Schiller HB, Hermann MR, Polleux J, Vignaud T, Zanivan S, Friedel CC, Sun Z, Raducanu A, Gottschalk KE, Théry M, Mann M, Fässler R. β1- and αv-class integrins cooperate to regulate myosin II during rigidity sensing of fibronectin-based microenvironments. Nat Cell Biol 2013; 15:625-36. [PMID: 23708002 DOI: 10.1038/ncb2747] [Citation(s) in RCA: 339] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 04/04/2013] [Indexed: 12/14/2022]
Abstract
How different integrins that bind to the same type of extracellular matrix protein mediate specific functions is unclear. We report the functional analysis of β1- and αv-class integrins expressed in pan-integrin-null fibroblasts seeded on fibronectin. Reconstitution with β1-class integrins promotes myosin-II-independent formation of small peripheral adhesions and cell protrusions, whereas expression of αv-class integrins induces the formation of large focal adhesions. Co-expression of both integrin classes leads to full myosin activation and traction-force development on stiff fibronectin-coated substrates, with αv-class integrins accumulating in adhesion areas exposed to high traction forces. Quantitative proteomics linked αv-class integrins to a GEF-H1-RhoA pathway coupled to the formin mDia1 but not myosin II, and α5β1 integrins to a RhoA-Rock-myosin II pathway. Our study assigns specific functions to distinct fibronectin-binding integrins, demonstrating that α5β1integrins accomplish force generation, whereas αv-class integrins mediate the structural adaptations to forces, which cooperatively enable cells to sense the rigidity of fibronectin-based microenvironments.
Collapse
Affiliation(s)
- Herbert B Schiller
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
The mannose 6-phosphate-binding sites of M6P/IGF2R determine its capacity to suppress matrix invasion by squamous cell carcinoma cells. Biochem J 2013; 451:91-9. [PMID: 23347038 PMCID: PMC3632087 DOI: 10.1042/bj20121422] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The M6P (mannose 6-phosphate)/IGF2R (insulin-like growth factor II receptor) interacts with a
variety of factors that impinge on tumour invasion and metastasis. It has been shown that expression
of wild-type M6P/IGF2R reduces the tumorigenic and invasive properties of receptor-deficient SCC-VII
squamous cell carcinoma cells. We have now used mutant forms of M6P/IGF2R to assess the relevance of
the different ligand-binding sites of the receptor for its biological activities in this cellular
system. The results of the present study demonstrate that M6P/IGF2R does not require a functional
binding site for insulin-like growth factor II for inhibition of anchorage-independent growth and
matrix invasion by SCC-VII cells. In contrast, the simultaneous mutation of both M6P-binding sites
is sufficient to impair all cellular functions of the receptor tested. These findings highlight that
the interaction between M6P/IGF2R and M6P-modified ligands is not only important for intracellular
accumulation of lysosomal enzymes and formation of dense lysosomes, but is also crucial for the
ability of the receptor to suppress SCC-VII growth and invasion. The present study also shows that
some of the biological activities of M6P/IGF2R in SCC-VII cells strongly depend on a functional
M6P-binding site within domain 3, thus providing further evidence for the non-redundant cellular
functions of the individual carbohydrate-binding domains of the receptor.
Collapse
|
29
|
|
30
|
Djiogue S, Nwabo Kamdje AH, Vecchio L, Kipanyula MJ, Farahna M, Aldebasi Y, Seke Etet PF. Insulin resistance and cancer: the role of insulin and IGFs. Endocr Relat Cancer 2013; 20:R1-R17. [PMID: 23207292 DOI: 10.1530/erc-12-0324] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Insulin, IGF1, and IGF2 are the most studied insulin-like peptides (ILPs). These are evolutionary conserved factors well known as key regulators of energy metabolism and growth, with crucial roles in insulin resistance-related metabolic disorders such as obesity, diseases like type 2 diabetes mellitus, as well as associated immune deregulations. A growing body of evidence suggests that insulin and IGF1 receptors mediate their effects on regulating cell proliferation, differentiation, apoptosis, glucose transport, and energy metabolism by signaling downstream through insulin receptor substrate molecules and thus play a pivotal role in cell fate determination. Despite the emerging evidence from epidemiological studies on the possible relationship between insulin resistance and cancer, our understanding on the cellular and molecular mechanisms that might account for this relationship remains incompletely understood. The involvement of IGFs in carcinogenesis is attributed to their role in linking high energy intake, increased cell proliferation, and suppression of apoptosis to cancer risks, which has been proposed as the key mechanism bridging insulin resistance and cancer. The present review summarizes and discusses evidence highlighting recent advances in our understanding on the role of ILPs as the link between insulin resistance and cancer and between immune deregulation and cancer in obesity, as well as those areas where there remains a paucity of data. It is anticipated that issues discussed in this paper will also recover new therapeutic targets that can assist in diagnostic screening and novel approaches to controlling tumor development.
Collapse
Affiliation(s)
- Sefirin Djiogue
- Department of Animal Biology and Physiology, University of Yaoundé 1, PO Box 812, Yaoundé, Cameroon
| | | | | | | | | | | | | |
Collapse
|
31
|
Mani T, Wang F, Knabe WE, Sinn AL, Khanna M, Jo I, Sandusky GE, Sledge GW, Jones DR, Khanna R, Pollok KE, Meroueh SO. Small-molecule inhibition of the uPAR·uPA interaction: synthesis, biochemical, cellular, in vivo pharmacokinetics and efficacy studies in breast cancer metastasis. Bioorg Med Chem 2013; 21:2145-55. [PMID: 23411397 DOI: 10.1016/j.bmc.2012.12.047] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/13/2012] [Accepted: 12/20/2012] [Indexed: 10/27/2022]
Abstract
The uPAR·uPA protein-protein interaction (PPI) is involved in signaling and proteolytic events that promote tumor invasion and metastasis. A previous study had identified 4 (IPR-803) from computational screening of a commercial chemical library and shown that the compound inhibited uPAR·uPA PPI in competition biochemical assays and invasion cellular studies. Here, we synthesize 4 to evaluate in vivo pharmacokinetic (PK) and efficacy studies in a murine breast cancer metastasis model. First, we show, using fluorescence polarization and saturation transfer difference (STD) NMR, that 4 binds directly to uPAR with sub-micromolar affinity of 0.2 μM. We show that 4 blocks invasion of breast MDA-MB-231, and inhibits matrix metalloproteinase (MMP) breakdown of the extracellular matrix (ECM). Derivatives of 4 also inhibited MMP activity and blocked invasion in a concentration-dependent manner. Compound 4 also impaired MDA-MB-231 cell adhesion and migration. Extensive in vivo PK studies in NOD-SCID mice revealed a half-life of nearly 5h and peak concentration of 5 μM. Similar levels of the inhibitor were detected in tumor tissue up to 10h. Female NSG mice inoculated with highly malignant TMD-MDA-MB-231 in their mammary fat pads showed that 4 impaired metastasis to the lungs with only four of the treated mice showing severe or marked metastasis compared to ten for the untreated mice. Compound 4 is a promising template for the development of compounds with enhanced PK parameters and greater efficacy.
Collapse
Affiliation(s)
- Timmy Mani
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Van Nuys Medical Science Building, MS 4023, 635 Barnhill Drive, Indianapolis, IN 46202-5122, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Wang XY, Fillafer C, Pichl C, Deinhammer S, Hofer-Warbinek R, Wirth M, Gabor F. A multichannel acoustically driven microfluidic chip to study particle-cell interactions. BIOMICROFLUIDICS 2013; 7:44127. [PMID: 24404060 PMCID: PMC3772939 DOI: 10.1063/1.4819273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/09/2013] [Indexed: 05/13/2023]
Abstract
Microfluidic devices have emerged as important tools for experimental physiology. They allow to study the effects of hydrodynamic flow on physiological and pathophysiological processes, e.g., in the circulatory system of the body. Such dynamic in vitro test systems are essential in order to address fundamental problems in drug delivery and targeted imaging, such as the binding of particles to cells under flow. In the present work an acoustically driven microfluidic platform is presented in which four miniature flow channels can be operated in parallel at distinct flow velocities with only slight inter-experimental variations. The device can accommodate various channel architectures and is fully compatible with cell culture as well as microscopy. Moreover, the flow channels can be readily separated from the surface acoustic wave pumps and subsequently channel-associated luminescence, absorbance, and/or fluorescence can be determined with a standard microplate reader. In order to create artificial blood vessels, different coatings were evaluated for the cultivation of endothelial cells in the microchannels. It was found that 0.01% fibronectin is the most suitable coating for growth of endothelial monolayers. Finally, the microfluidic system was used to study the binding of 1 μm polystyrene microspheres to three different types of endothelial cell monolayers (HUVEC, HUVECtert, HMEC-1) at different average shear rates. It demonstrated that average shear rates between 0.5 s(-1) and 2.25 s(-1) exert no significant effect on cytoadhesion of particles to all three types of endothelial monolayers. In conclusion, the multichannel microfluidic platform is a promising device to study the impact of hydrodynamic forces on cell physiology and binding of drug carriers to endothelium.
Collapse
Affiliation(s)
- Xue-Yan Wang
- Faculty of Life Sciences, Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Vienna A-1090, Austria
| | - Christian Fillafer
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts 02215, USA
| | - Clara Pichl
- Faculty of Life Sciences, Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Vienna A-1090, Austria
| | - Stephanie Deinhammer
- Faculty of Life Sciences, Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Vienna A-1090, Austria
| | - Renate Hofer-Warbinek
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna A-1090, Austria
| | - Michael Wirth
- Faculty of Life Sciences, Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Vienna A-1090, Austria
| | - Franz Gabor
- Faculty of Life Sciences, Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Vienna A-1090, Austria
| |
Collapse
|
33
|
Stie J, Fox D. Induction of brain microvascular endothelial cell urokinase expression by Cryptococcus neoformans facilitates blood-brain barrier invasion. PLoS One 2012; 7:e49402. [PMID: 23145170 PMCID: PMC3493525 DOI: 10.1371/journal.pone.0049402] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 10/10/2012] [Indexed: 11/30/2022] Open
Abstract
The invasive ability of the blood-borne fungal pathogen Cryptococcus neoformans can be enhanced through interactions with host plasma components, such as plasminogen. Previously we showed by in vitro studies that plasminogen coats the surface of C. neoformans and is converted to the active serine protease, plasmin, by host plasminogen activators. Viable, but not formaldehyde- or sodium azide-killed, cryptococcal strains undergo brain microvascular endothelial cell-dependent plasminogen-to-plasmin activation, which results in enhanced, plasmin-dependent cryptococcal invasion of primary bovine brain microvascular endothelial cells and fungal ability to degrade plasmin substrates. In the present work, brain microvascular endothelial cells cultured with viable, but not killed, cryptococcal strains led to significant increases in both urokinase mRNA transcription and cell-associated urokinase protein expression. Soluble urokinase was also detected in conditioned medium from brain microvascular endothelial cells cultured with viable, but not killed, C. neoformans. Exposure of plasminogen pre-coated viable C. neoformans to conditioned medium from strain-matched brain microvascular endothelial cell-fungal co-cultures resulted in plasminogen-to-plasmin activation and plasmin-dependent cryptococcal invasion. siRNA-mediated silencing of urokinase gene expression or the use of specific inhibitors of urokinase activity abrogated both plasminogen-to-plasmin activation on C. neoformans and cryptococcal-brain microvascular endothelial cell invasion. Our results suggest that pathogen exploitation of the host urokinase-plasmin(ogen) system may contribute to C. neoformans virulence during invasive cryptococcosis.
Collapse
Affiliation(s)
- Jamal Stie
- Research Institute for Children, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Deborah Fox
- Research Institute for Children, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Department of Pediatrics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
34
|
Grismayer B, Sölch S, Seubert B, Kirchner T, Schäfer S, Baretton G, Schmitt M, Luther T, Krüger A, Kotzsch M, Magdolen V. Rab31 expression levels modulate tumor-relevant characteristics of breast cancer cells. Mol Cancer 2012; 11:62. [PMID: 22920728 PMCID: PMC3499445 DOI: 10.1186/1476-4598-11-62] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 08/16/2012] [Indexed: 11/12/2022] Open
Abstract
Background Rab proteins constitute a large family of monomeric GTP-binding proteins that regulate intracellular vesicle transport. Several Rab proteins, including rab31, have been shown to affect cancer progression and are related with prognosis in various types of cancer including breast cancer. Recently, the gene encoding rab31 was found to be overexpressed in estrogen receptor-positive breast cancer tissue. In a previous study we found a significant association of high rab31 mRNA expression with poor prognosis in node-negative breast cancer patients. In the present study, we aimed to investigate the impact of rab31 (over)-expression on important aspects of tumor progression in vitro and in vivo. Methods Breast cancer cells displaying low (MDA-MB-231) or no (CAMA-1) endogenous rab31 expression were stably transfected with a rab31 expression plasmid. Batch-transfected cells as well as selected cell clones, expressing different levels of rab31 protein, were analyzed with regard to proliferation, cell adhesion, the invasive capacity of tumor cells, and in vivo in a xenograft tumor model. Polyclonal antibodies directed to recombinantly expressed rab31 were generated and protein expression analyzed by immunohistochemistry, Western blot analysis, and a newly developed sensitive ELISA. Results Elevated rab31 protein levels were associated with enhanced proliferation of breast cancer cells. Interestingly, weak to moderate overexpression of rab31 in cell lines with no detectable endogenous rab31 expression was already sufficient to elicit distinct effects on cell proliferation. By contrast, increased expression of rab31 in breast cancer cells led to reduced adhesion towards several extracellular matrix proteins and decreased invasive capacity through MatrigelTM. Again, the rab31-mediated effects on cell adhesion and invasion were dose-dependent. Finally, in a xenograft mouse model, we observed a significantly impaired metastatic dissemination of rab31 overexpressing MDA-MB-231 breast cancer cells to the lung. Conclusions Overexpression of rab31 in breast cancer cells leads to a switch from an invasive to a proliferative phenotype as indicated by an increased cell proliferation, reduced adhesion and invasion in vitro, and a reduced capacity to form lung metastases in vivo.
Collapse
Affiliation(s)
- Bettina Grismayer
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str, 22, Munich 81675, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Puxbaum V, Nimmerfall E, Bäuerl C, Taub N, Blaas PM, Wieser J, Mikula M, Mikulits W, Ng KM, Yeoh GC, Mach L. M6P/IGF2R modulates the invasiveness of liver cells via its capacity to bind mannose 6-phosphate residues. J Hepatol 2012; 57:337-43. [PMID: 22521359 PMCID: PMC3401376 DOI: 10.1016/j.jhep.2012.03.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 03/10/2012] [Accepted: 03/23/2012] [Indexed: 01/18/2023]
Abstract
BACKGROUND & AIMS The mannose 6-phosphate/insulin-like growth factor II receptor (M6P/IGF2R), a multifunctional protein, plays a central role in intracellular targeting of lysosomal enzymes and control of insulin-like growth factor II (IGF-II) bioactivity. Importantly, the gene encoding this receptor is frequently inactivated in a wide range of malignant tumors including hepatocellular carcinomas. Thus, M6P/IGF2R is considered a putative liver tumor suppressor. The aim of this study was to establish the impact of the receptor on the invasive properties of liver cells. METHODS Reconstitution experiments were performed by expression of wild type and mutant M6P/IGF2R in receptor-deficient FRL14 fetal rat liver cells. RNA interference was used to induce M6P/IGF2R downregulation in receptor-positive MIM-1-4 mouse hepatocytes. RESULTS We show that the M6P/IGF2R status exerts a strong impact on the invasiveness of tumorigenic rodent liver cells. M6P/IGF2R-deficient fetal rat liver cells hypersecrete lysosomal cathepsins and penetrate extracellular matrix barriers in a cathepsin-dependent manner. Forced expression of M6P/IGF2R restores intracellular transport of cathepsins to lysosomes and concomitantly reduces the tumorigenicity and invasive potential of these cells. Conversely, M6P/IGF2R knock-down in receptor-positive mouse hepatocytes causes increased cathepsin secretion as well as enhanced cell motility and invasiveness. We also demonstrate that functional M6P-binding sites are important for the anti-invasive properties of M6P/IGF2R, whereas the capacity to bind IGF-II is dispensable for the anti-invasive activity of the receptor in liver cells. CONCLUSIONS M6P/IGF2R restricts liver cell invasion by preventing the pericellular action of M6P-modified proteins.
Collapse
Key Words
- bsa, bovine serum albumin
- ecm, extracellular matrix
- fbs, fetal bovine serum
- hcc, hepatocellular carcinoma
- hgf, hepatocyte growth factor
- igf-ii, insulin-like growth factor ii
- m6p, mannose 6-phosphate
- m6p/igf2r, mannose 6-phosphate/insulin-like growth factor ii receptor
- mpr46, 46-kda mannose 6-phosphate receptor
- rnai, rna interference
- shrna, short hairpin rna
- sirna, short interfering rna
- cathepsin
- hepatocellular carcinoma
- lysosome
- matrix degradation
- cell invasion
Collapse
Affiliation(s)
- Verena Puxbaum
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Elisabeth Nimmerfall
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Christine Bäuerl
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Nicole Taub
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Pia-Maria Blaas
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Johannes Wieser
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Mario Mikula
- Department of Internal Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Wolfgang Mikulits
- Department of Internal Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Ken M. Ng
- School of Biomedical, Biomolecular and Chemical Sciences, The University of Western Australia, 35 Stirling Highway, M310, Crawley, WA 6009, Australia
| | - George C.T. Yeoh
- School of Biomedical, Biomolecular and Chemical Sciences, The University of Western Australia, 35 Stirling Highway, M310, Crawley, WA 6009, Australia,Laboratory for Cancer Medicine, Western Australian Institute for Medical Research, 50 Murray Street, Perth, WA 6000, Australia
| | - Lukas Mach
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria,Corresponding author. Tel.: +43 1 47654 6360, fax: +43 1 47654 6392.
| |
Collapse
|
36
|
Rozema E, Binder M, Bulusu M, Bochkov V, Krupitza G, Kopp B. Effects on inflammatory responses by the sphingoid base 4,8-sphingadienine. Int J Mol Med 2012; 30:703-7. [PMID: 22710663 DOI: 10.3892/ijmm.2012.1035] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 05/04/2012] [Indexed: 11/05/2022] Open
Abstract
Sphingolipids (SLs) are a class of lipids, which are structural cell components involved in the regulation of cellular processes such as cell proliferation, differentiation, apoptosis and inflammation. Dietary SLs are enzymatically hydrolyzed in the gut lumen into metabolites, namely ceramides and sphingoid bases. The sphingoid base 4,8-sphingadienine (4,8-SD) is the metabolite of glucocerebrosides derived from plants that are part of the human diet. The present findings provide insight into the effects of 4,8-SD on inflammatory responses that may be of nutritional and therapeutic benefit. We demonstrated that 4,8-SD significantly inhibited tumor necrosis factor-α (TNF-α)- and lipopolysaccharide (LPS)-induced expression of IL-8 and E-selectin in human endothelial cells in a dose-dependent manner. The anti-inflammatory effect was observed at significantly lower concentrations of 4,8-SD compared those affecting cell viability as judged by the LDH and WST-1 assays.
Collapse
Affiliation(s)
- Evelien Rozema
- Department of Pharmacognosy, University of Vienna, A-1090 Vienna, Austria
| | | | | | | | | | | |
Collapse
|
37
|
Wang F, Eric Knabe W, Li L, Jo I, Mani T, Roehm H, Oh K, Li J, Khanna M, Meroueh SO. Design, synthesis, biochemical studies, cellular characterization, and structure-based computational studies of small molecules targeting the urokinase receptor. Bioorg Med Chem 2012; 20:4760-73. [PMID: 22771232 DOI: 10.1016/j.bmc.2012.06.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 05/25/2012] [Accepted: 06/01/2012] [Indexed: 11/26/2022]
Abstract
The urokinase receptor (uPAR) serves as a docking site to the serine protease urokinase-type plasminogen activator (uPA) to promote extracellular matrix (ECM) degradation and tumor invasion and metastasis. Previously, we had reported a small molecule inhibitor of the uPAR·uPA interaction that emerged from structure-based virtual screening. Here, we measure the affinity of a large number of derivatives from commercial sources. Synthesis of additional compounds was carried out to probe the role of various groups on the parent compound. Extensive structure-based computational studies suggested a binding mode for these compounds that led to a structure-activity relationship study. Cellular studies in non-small cell lung cancer (NSCLC) cell lines that include A549, H460 and H1299 showed that compounds blocked invasion, migration and adhesion. The effects on invasion of active compounds were consistent with their inhibition of uPA and MMP proteolytic activity. These compounds showed weak cytotoxicity consistent with the confined role of uPAR to metastasis.
Collapse
Affiliation(s)
- Fang Wang
- Indiana University, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Leksa V, Pfisterer K, Ondrovičová G, Binder B, Lakatošová S, Donner C, Schiller HB, Zwirzitz A, Mrvová K, Pevala V, Kutejová E, Stockinger H. Dissecting mannose 6-phosphate-insulin-like growth factor 2 receptor complexes that control activation and uptake of plasminogen in cells. J Biol Chem 2012; 287:22450-62. [PMID: 22613725 DOI: 10.1074/jbc.m112.339663] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The plasminogen (Plg) activation cascade on the cell surface plays a central role in cell migration and is involved in a plethora of physiological and pathological processes. Its regulation is coordinated by many receptors, in particular the urokinase-type plasminogen activator receptor (uPAR, CD87), receptors that physically interact and functionally cooperate with uPAR, and Plg binding molecules. Here we studied the impact of one of the Plg binding molecules, the mannose 6-phosphate/insulin-like growth factor 2 receptor (M6P-IGF2R, CD222), on cellular Plg activation. By developing both in vitro and in vivo Plg activation assays on size-fractionated lysates of M6P-IGF2R-silenced cells, we identified Plg-associated complexes with M6P-IGF2R as the regulatory factor. Using lipid raft preserving versus dissolving detergents, we found lipid dependence of the Plg regulatory function of these complexes. Furthermore, M6P-IGF2R-silencing in uPAR-positive human cell lines reduced internalization of Plg, resulting in elevated Plg activation. In contrast, the expression of human M6P-IGF2R in mouse embryonic fibroblasts derived from M6P-IGF2R knock-out mice enhanced Plg internalization. Finally, peptide 18-36 derived from the Plg-binding site within M6P-IGF2R enhanced Plg uptake. Thus, by targeting Plg to endocytic pathways, M6P-IGF2R appears to control Plg activation within cells that might be important to restrict plasmin activity to specific sites and substrates.
Collapse
Affiliation(s)
- Vladimir Leksa
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Takahashi Y, Mimori K, Yamamoto K, Watanabe M, Tanaka JI, Kudo SE, Sugihara KI, Hase K, Mochizuki H, Kusunoki M, Yamada K, Shimada Y, Moriya Y, Mori M. Genomic copy number of a carcinogenic single nucleotide polymorphism at 8q24 in non-risk allele colorectal cancer associated with insulin growth factor 2 receptor expression. J Gastroenterol Hepatol 2012; 27 Suppl 3:95-9. [PMID: 22486879 DOI: 10.1111/j.1440-1746.2012.07081.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM The incidence of both diabetes mellitus and hyperlipidemia is increasing and they are risk factors for colorectal cancer (CRC). On the other hand, the carcinogenic significance of the single nucleotide polymorphism (SNP), rs6983267 at 8q24, in CRC has been reported. The association between the SNP genotype and genes associated with diabetes or hyperlipidemia was investigated in cases of CRC. METHODS In 107 cases of CRC diagnosed in eight institutes from 2003 to 2008, array-CGH and cDNA microarray was performed and the data analyzed from two groups subdivided according to SNP genotype. RESULTS In the array-CGH data, we selected 38 genes related to diabetes or fat metabolism, and of these 10 had a correlation coefficient between the genome copy number at 8q24 locus and that of each gene. Of the 10 genes, insulin growth factor 2 receptor (IGF2R) was the only one with an expression level significantly associated with the 8q24 genotype. IGF2R expression was significantly lower in non-risk allele than in risk allele cases (P = 0.012). There was neither a diabetes- nor a fat metabolism-related gene that was significantly associated with CRC cases with the risk allele at 8q24. CONCLUSIONS SNP at 8q24 makes diabetes a risk factor of CRC via IGF2R, especially in genetically non-risk allele cases. We speculate that the risk allele of 8q24 might be risky enough that diabetes is not necessary to worsen the risk for CRC.
Collapse
Affiliation(s)
- Yusuke Takahashi
- Department of Surgical Oncology, Medical Institute of Bioregulation, Kyushu University, Beppu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Dong X, Li Y, Tang H, Chang P, Hess KR, Abbruzzese JL, Li D. Insulin-like growth factor axis gene polymorphisms modify risk of pancreatic cancer. Cancer Epidemiol 2012; 36:206-11. [PMID: 21852217 DOI: 10.1016/j.canep.2011.05.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 05/18/2011] [Accepted: 05/19/2011] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Insulin-like growth factor (IGF)-axis genes plays a critical role in cancer development and progression via their impact on the RAS/MAPK/ERK and PI3K/AKT/mTOR signaling pathways. We hypothesized that IGF-axis genetic variants modify individual susceptibility to pancreatic cancer. METHODS We retrospectively genotyped 41 single-nucleotide polymorphisms of 10 IGF-axis genes (IGF1, IGF2, IGF1R, IGF2R, IGFBP1, IGFBP3, IGFBP5, IRS1, IRS2, and IRS4) in 706 pancreatic cancer patients and 706 cancer-free controls using Sequenom and TaqMan technology. The association between genotype and pancreatic cancer risk was evaluated using multivariate logistic regression. A P value ≤.007 at a false discovery rate of 10% was set as the significance level. RESULTS We observed that the IGF1 *10212C>A and Ex4+2776G>A and IGF1R IVS2-70184A>G and IVS2+46329T>C variant genotypes were significantly associated with decreased pancreatic cancer risk (odds ratio [OR] range, 0.60-0.75) and that IGFBP1 Ex4+111A>G (I253M) was significantly associated with increased pancreatic cancer risk (OR=1.46) after adjusted for other risk factors and multiple comparisons (P≤.007). IGF2R and IGFBP3 variant haplotypes were associated with increased and decreased pancreatic cancer risk, respectively (P<.001). We also observed a weak interaction of the IGF1R IVS2+46329T>C and IGF2R Ex45+11C>T (L2222L) genotypes with diabetes (P(interaction)=.05) and interaction of IGF2R and IRS1 genotypes with alcohol consumption (P(interaction)=.03 and .019, respectively) on increased pancreatic cancer risk. CONCLUSION These findings support our hypothesis that polymorphic variants of IGF-axis genes act alone or jointly with other risk factors to affect susceptibility to pancreatic cancer.
Collapse
Affiliation(s)
- Xiaoqun Dong
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States.
| | | | | | | | | | | | | |
Collapse
|
41
|
Khanna M, Wang F, Jo I, Knabe WE, Wilson SM, Li L, Bum-Erdene K, Li J, W. Sledge G, Khanna R, Meroueh SO. Targeting multiple conformations leads to small molecule inhibitors of the uPAR·uPA protein-protein interaction that block cancer cell invasion. ACS Chem Biol 2011; 6:1232-43. [PMID: 21875078 DOI: 10.1021/cb200180m] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interaction of the urokinase receptor (uPAR) with its binding partners such as the urokinase-type plasminogen activator (uPA) at the cell surface triggers a series of proteolytic and signaling events that promote invasion and metastasis. Here, we report the discovery of a small molecule (IPR-456) and its derivatives that inhibit the tight uPAR·uPA protein-protein interaction. IPR-456 was discovered by virtual screening against multiple conformations of uPAR sampled from explicit-solvent molecular dynamics simulations. Biochemical characterization reveal that the compound binds to uPAR with submicromolar affinity (K(d) = 310 nM) and inhibits the tight protein-protein interaction with an IC(50) of 10 μM. Free energy calculations based on explicit-solvent molecular dynamics simulations suggested the importance of a carboxylate moiety on IPR-456, which was confirmed by the activity of several derivatives including IPR-803. Immunofluorescence imaging showed that IPR-456 inhibited uPA binding to uPAR of breast MDA-MB-231 tumor cells with an IC(50) of 8 μM. The compounds blocked MDA-MB-231 cell invasion, but IPR-456 showed little effect on MDA-MB-231 migration and no effect on adhesion, suggesting that uPAR mediates these processes through its other binding partners.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Khuchtumur Bum-Erdene
- Department of Chemistry and Chemical Biology, Indiana University Purdue University Indianapolis (IUPUI), Indianapolis, Indiana 46202, United States
| | | | | | | | - Samy O. Meroueh
- Department of Chemistry and Chemical Biology, Indiana University Purdue University Indianapolis (IUPUI), Indianapolis, Indiana 46202, United States
| |
Collapse
|
42
|
Prager GW, Poettler M. Angiogenesis in cancer. Basic mechanisms and therapeutic advances. Hamostaseologie 2011; 32:105-14. [PMID: 21837355 DOI: 10.5482/ha-1163] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 07/18/2011] [Indexed: 12/15/2022] Open
Abstract
Etiological concepts on cancer development, malignant growth and tumour propagation have undergone a revolutionary development during recent years: Among other aspects, the discovery of angiogenesis - the growth of new blood vessels from pre-existing vasculature - as a key element in the pathogenesis of malignancy has opened an abundance of biologic insights and subsequent therapeutic options, which have led to improved prognosis in many cancers including those originating from colon, lung, breast and kidney. Thereby, targeting the major pro-angiogenic stimulus vascular endothelial growth factor (VEGF) became the focus for therapeutic interventions. However, the use of VEGF-targeting drugs has been shown to be of limited efficacy, which might lie in the fact that tumor angiogenesis is mediated by a variety of different subcellular systems. This review focuses on the basic mechanisms involved in angiogenesis, which potentially represent novel targets for pharmacological agents in the treatment of malignancies.
Collapse
Affiliation(s)
- G W Prager
- Comprehensive Cancer Center Vienna, Department of Medicine I, Medical University of Vienna, Austria.
| | | |
Collapse
|
43
|
Leksa V, Loewe R, Binder B, Schiller HB, Eckerstorfer P, Forster F, Soler-Cardona A, Ondrovičová G, Kutejová E, Steinhuber E, Breuss J, Drach J, Petzelbauer P, Binder BR, Stockinger H. Soluble M6P/IGF2R Released by TACE Controls Angiogenesis via Blocking Plasminogen Activation. Circ Res 2011; 108:676-85. [DOI: 10.1161/circresaha.110.234732] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Rationale:
The urokinase plasminogen activator (uPA) system is among the most crucial pericellular proteolytic systems associated with the processes of angiogenesis. We previously identified an important regulator of the uPA system in the mannose 6-phosphate/insulin-like growth factor 2 receptor (M6P/IGF2R).
Objective:
Here, we wanted to clarify whether and how did the soluble form of M6P/IGF2R (sM6P/IGF2R) contribute to modulation of the uPA system.
Methods and Results:
By using specific inhibitors and RNA interference, we show that the tumor necrosis factor α convertase (TACE, ADAM-17) mediates the release of the ectodomain of M6P/IGF2R from human endothelial cells. We demonstrate further that sM6P/IGF2R binds plasminogen (Plg) and thereby prevents Plg from binding to the cell surface and uPA, ultimately inhibiting in this manner Plg activation. Furthermore, peptide 18-36 derived from the Plg-binding site of M6P/IGF2R mimics sM6P/IGF2R in the inhibition of Plg activation and blocks cancer cell invasion in vitro, endothelial cell invasion in vivo, and tumor growth in vivo.
Conclusions:
The interaction of sM6P/IGF2R with Plg may be an important regulatory mechanism to inhibit migration of cells using the uPA/uPAR system.
Collapse
Affiliation(s)
- Vladimir Leksa
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Robert Loewe
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Brigitte Binder
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Herbert B. Schiller
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Paul Eckerstorfer
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Florian Forster
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Ana Soler-Cardona
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Gabriela Ondrovičová
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Eva Kutejová
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Eva Steinhuber
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Johannes Breuss
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Johannes Drach
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Peter Petzelbauer
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Bernd R. Binder
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| | - Hannes Stockinger
- From the Molecular Immunology Unit (V.L., B.B., H.B.S., P.E., F.F., E.S., H.S.), Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria; Institute of Molecular Biology (V.L., G.O., E.K.), Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Dermatology (R.L., A.S.-C., P.B.), Medical University of Vienna, Austria; Department of Vascular Biology and Thrombosis Research (J.B., B.R.B.), Center for
| |
Collapse
|
44
|
Dong X, Javle M, Hess KR, Shroff R, Abbruzzese JL, Li D. Insulin-like growth factor axis gene polymorphisms and clinical outcomes in pancreatic cancer. Gastroenterology 2010; 139:464-73, 473.e1-3. [PMID: 20416304 PMCID: PMC2910789 DOI: 10.1053/j.gastro.2010.04.042] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 03/15/2010] [Accepted: 04/12/2010] [Indexed: 01/22/2023]
Abstract
BACKGROUND & AIMS Insulin-like growth factor (IGF)-axis mediated signaling pathways play an important role in pancreatic cancer development and progression. We examined whether IGF-axis gene variants are associated with clinical outcomes in pancreatic cancer. METHODS We retrospectively genotyped 41 single-nucleotide polymorphisms from 10 IGF-axis genes in 333 patients with localized pancreatic adenocarcinoma and validated the findings in 373 patients with advanced disease. Associations between genotype and overall survival (OS) were evaluated using multivariable Cox proportional hazard regression models. RESULTS IGF1 *8470T>C, IGF1R IVS2+46329T>C, IGFBP3 A32G, IRS1 G972R in patients with localized disease; IGF1R IVS20-3431A>G, IGF1R T766T, IGFBP3-202A>C, IRS1 IVS1+4315C>G, IRS1 G972R in patients with advanced disease; and IGF1R T766T, IGF2R L252V, IGFBP3 -202A>C, IRS1 IVS1+4315C>G, IRS1 G972R, IRS2 IVS1+5687T>C in all patients were significantly associated with OS (P<or=.007). Two haplotypes containing the variant allele of either IRS1 G972R or IVS1-10949G>A, and an IRS2 haplotype predicted worse OS (P<or=.002). A significant correlation between increased number of unfavorable genotypes and decreased OS was observed; patients with 0-1 (n=247), 2 (n=237), 3 (n=145), 4 (n=60), and 5-8 (n=17) unfavorable genotypes had median survival time of 24.2, 16.4, 14.4, 9.6, and 7.4 months, respectively (P<.001). Several single-nucleotide polymorphisms of IGF1R, IGF2R, and IRS1 gene were significantly associated with tumor response to therapy and disease stage. CONCLUSIONS These data suggest that individual genetic variations in the IGF axis pathway may predict worse survival in patients with pancreatic cancer. This information may identify population subgroups that could benefit from IGF(1)R-targeted agents.
Collapse
|
45
|
Bower NI, Johnston IA. Transcriptional regulation of the IGF signaling pathway by amino acids and insulin-like growth factors during myogenesis in Atlantic salmon. PLoS One 2010; 5:e11100. [PMID: 20559434 PMCID: PMC2885424 DOI: 10.1371/journal.pone.0011100] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 05/19/2010] [Indexed: 11/19/2022] Open
Abstract
The insulin-like growth factor signalling pathway is an important regulator of skeletal muscle growth. We examined the mRNA expression of components of the insulin-like growth factor (IGF) signalling pathway as well as Fibroblast Growth Factor 2 (FGF2) during maturation of myotubes in primary cell cultures isolated from fast myotomal muscle of Atlantic salmon (Salmo salar). The transcriptional regulation of IGFs and IGFBP expression by amino acids and insulin-like growth factors was also investigated. Proliferation of cells was 15% d−1 at days 2 and 3 of the culture, increasing to 66% d−1 at day 6. Three clusters of elevated gene expression were observed during the maturation of the culture associated with mono-nucleic cells (IGFBP5.1 and 5.2, IGFBP-6, IGFBP-rP1, IGFBP-2.2 and IGF-II), the initial proliferation phase (IGF-I, IGFBP-4, FGF2 and IGF-IRb) and terminal differentiation and myotube production (IGF2R, IGF-IRa). In cells starved of amino acids and serum for 72 h, IGF-I mRNA decreased 10-fold which was reversed by amino acid replacement. Addition of IGF-I and amino acids to starved cells resulted in an 18-fold increase in IGF-I mRNA indicating synergistic effects and the activation of additional pathway(s) leading to IGF-I production via a positive feedback mechanism. IGF-II, IGFBP-5.1 and IGFBP-5.2 expression was unchanged in starved cells, but increased with amino acid replacement. Synergistic increases in expression of IGFBP5.2 and IGFBP-4, but not IGFBP5.1 were observed with addition of IGF-I, IGF-II or insulin and amino acids to the medium. IGF-I and IGF-II directly stimulated IGFBP-6 expression, but not when amino acids were present. These findings indicate that amino acids alone are sufficient to stimulate myogenesis in myoblasts and that IGF-I production is controlled by both endocrine and paracrine pathways. A model depicting the transcriptional regulation of the IGF pathway in Atlantic salmon muscle following feeding is proposed.
Collapse
Affiliation(s)
- Neil I Bower
- Scottish Oceans Institute, School of Biology, University of St Andrews, St Andrews, United Kingdom.
| | | |
Collapse
|
46
|
Abstract
Urokinase-type plasminogen activator receptor (uPAR) expression is elevated during inflammation and tissue remodelling and in many human cancers, in which it frequently indicates poor prognosis. uPAR regulates proteolysis by binding the extracellular protease urokinase-type plasminogen activator (uPA; also known as urokinase) and also activates many intracellular signalling pathways. Coordination of extracellular matrix (ECM) proteolysis and cell signalling by uPAR underlies its important function in cell migration, proliferation and survival and makes it an attractive therapeutic target in cancer and inflammatory diseases. uPAR lacks transmembrane and intracellular domains and so requires transmembrane co-receptors for signalling. Integrins are essential uPAR signalling co-receptors and a second uPAR ligand, the ECM protein vitronectin, is also crucial for this process.
Collapse
Affiliation(s)
- Harvey W Smith
- Goodman Cancer Centre, McGill University, West Montreal, Quebec, H3A 1A3, Canada.
| | | |
Collapse
|