1
|
Mi-ichi F, Tsugawa H, Yoshida H, Arita M. Unique features of Entamoeba histolytica glycerophospholipid metabolism; has the E. histolytica lipid metabolism network evolved through gene loss and gain to enable parasitic life cycle adaptation? mSphere 2023; 8:e0017423. [PMID: 37584599 PMCID: PMC10597341 DOI: 10.1128/msphere.00174-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/05/2023] [Indexed: 08/17/2023] Open
Abstract
Entamoeba histolytica, a protozoan parasite, causes amoebiasis, which is a global public health problem. During the life cycle of this parasite, the properties of the cell membrane are changed markedly. To clarify the mechanism of membrane lipid changes, we exploited state-of-the-art untargeted lipidomic analysis, and atypical features of glycerophospholipids, lysoglycerophospholipids, and sphingolipids were observed compared with human equivalents. Here, we overview an entire E. histolytica glycerophospholipid metabolic pathway based on re-evaluated whole lipidome and genome along with the results of metabolic labeling experiments. We also discuss whether the E. histolytica lipid metabolism network, including the glycerophospholipid metabolic pathway, has unique features necessary for parasitic life cycle adaptation through gene loss and/or gain, and raise important questions involving biochemistry, molecular cell biology, and physiology underlying this network. Answering these questions will advance the understanding of Entamoeba physiology and will provide potential targets to develop new anti-amoebiasis drugs.
Collapse
Affiliation(s)
- Fumika Mi-ichi
- Central Laboratory, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, Japan
| | - Hiroshi Tsugawa
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, Japan
| |
Collapse
|
2
|
Identification of glycerophospholipids using self-built recognition software based on positive and negative ion high-resolution mass spectrometric fragmentation experiments. Talanta 2022; 238:123006. [PMID: 34857339 DOI: 10.1016/j.talanta.2021.123006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 11/22/2022]
Abstract
Glycerophospholipids (GPs) have a wide variety and complex structure, which makes their identification challenging. Our software affords a novel tool for the automated identification of non-target GPs in biological mixtures. Here, we explored the multi-stage fragmentation processes of GPs in positive and negative ion modes, and then constructed multi-stage fragment ion databases. This database includes 8214 simulated GP molecules from a random combination of fatty acids corresponding to 42,439 self-built predicted multi-stage fragment ions in positive ion mode and 31,487 self-built predicted multi-stage fragment ions in negative ion mode (MS ≤ 3). The automatic GP identification (AGPI) software can screen out GP candidates utilizing the MS1 accurate mass. The isomers of fatty acid chains and the phosphoryl head group can be distinguished using the MS2 and MS3 fragment spectra in positive-ion and negative-ion modes. All of the selected 45 GP standards were putatively identified using AGPI software; however, there were false positives because the software cannot distinguish positional isomers of fatty acids. Therefore, the AGPI software could be applied to identify GPs in samples, such as cancer cells; we successfully identified 41 GPs in cancer cells.
Collapse
|
3
|
Lusk H, Burdette JE, Sanchez LM. Models for measuring metabolic chemical changes in the metastasis of high grade serous ovarian cancer: fallopian tube, ovary, and omentum. Mol Omics 2021; 17:819-832. [PMID: 34338690 PMCID: PMC8649074 DOI: 10.1039/d1mo00074h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ovarian cancer (OC) is the most lethal gynecologic malignancy and high grade serous ovarian cancer (HGSOC) is the most common and deadly subtype, accounting for 70-80% of OC deaths. HGSOC has a distinct pattern of metastasis as many believe it originates in the fallopian tube and then it metastasizes first to the ovary, and later to the adipose-rich omentum. Metabolomics has been heavily utilized to investigate metabolite changes in HGSOC tumors and metastasis. Generally, metabolomics studies have traditionally been applied to biospecimens from patients or animal models; a number of recent studies have combined metabolomics with innovative cell-culture techniques to model the HGSOC metastatic microenvironment for the investigation of cell-to-cell communication. The purpose of this review is to serve as a tool for researchers aiming to model the metastasis of HGSOC for metabolomics analyses. It will provide a comprehensive overview of current knowledge on the origin and pattern of metastasis of HGSOC and discuss the advantages and limitations of different model systems to help investigators choose the best model for their research goals, with a special emphasis on compatibility with different metabolomics modalities. It will also examine what is presently known about the role of small molecules in the origin and metastasis of HGSOC.
Collapse
Affiliation(s)
- Hannah Lusk
- Department of Chemistry and Biochemistry, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA.
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 900 S Ashland Ave., Chicago, IL, 60607, USA
| | - Laura M Sanchez
- Department of Chemistry and Biochemistry, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA.
| |
Collapse
|
4
|
Liu W, Hopkins AM, Hou J. The development of modulators for lysophosphatidic acid receptors: A comprehensive review. Bioorg Chem 2021; 117:105386. [PMID: 34695732 DOI: 10.1016/j.bioorg.2021.105386] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/03/2021] [Accepted: 09/25/2021] [Indexed: 12/23/2022]
Abstract
Lysophosphatidic acids (LPAs) are bioactive phospholipids implicated in a wide range of cellular activities that regulate a diverse array of biological functions. They recognize two types of G protein-coupled receptors (LPARs): LPA1-3 receptors and LPA4-6 receptors that belong to the endothelial gene (EDG) family and non-endothelial gene family, respectively. In recent years, the LPA signaling pathway has captured an increasing amount of attention because of its involvement in various diseases, such as idiopathic pulmonary fibrosis, cancers, cardiovascular diseases and neuropathic pain, making it a promising target for drug development. While no drugs targeting LPARs have been approved by the FDA thus far, at least three antagonists have entered phase Ⅱ clinical trials for idiopathic pulmonary fibrosis (BMS-986020 and BMS-986278) and systemic sclerosis (SAR100842), and one radioligand (BMT-136088/18F-BMS-986327) has entered phase Ⅰ clinical trials for positron emission tomography (PET) imaging of idiopathic pulmonary fibrosis. This article provides an extensive review on the current status of ligand development targeting LPA receptors to modulate LPA signaling and their therapeutic potential in various diseases.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Chemistry, Lakehead University and Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON P7B 6V4, Canada
| | - Austin M Hopkins
- Department of Chemistry, Lakehead University and Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON P7B 6V4, Canada
| | - Jinqiang Hou
- Department of Chemistry, Lakehead University and Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON P7B 6V4, Canada.
| |
Collapse
|
5
|
Zhao J, Stephens T, Zhao Y. Molecular Regulation of Lysophosphatidic Acid Receptor 1 Maturation and Desensitization. Cell Biochem Biophys 2021; 79:477-483. [PMID: 34032994 PMCID: PMC8887818 DOI: 10.1007/s12013-021-00999-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2021] [Indexed: 10/21/2022]
Abstract
Lysophosphatidic acid receptor 1 (LPA1) belongs to the G protein-coupled receptor family. The ligand for LPA1 is LPA, the simplest lysophospholipid. LPA is considered a growth factor and induces cell proliferation, anti-apoptosis, and cell migration. The pro-inflammatory and pro-fibrotic roles of LPA have also been well-demonstrated. Most of the biological functions of LPA are mostly executed through LPA1. The mature form of LPA1 is glycosylated and localized on the plasma membrane. LPA1 is bound to heterotrimetric G proteins and transduces intracellular signaling in response to ligation to LPA. Desensitization of LPA1 negatively regulates LPA1-mediated signaling and the resulting biological functions. Phosphorylation and ubiquitination are well-demonstrated posttranslational modifications of GPCR. In this review, we will discuss our knowledge of LPA1 glycosylation, maturation, and trafficking from the endoplasmic reticulum (ER)/Golgi to the plasma membrane. Moreover, in light of recent findings, we will also discuss molecular regulation of LPA1 internalization and stability.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Physiology and Cell Biology, the Ohio State University, Columbus, OH, USA
| | - Thomas Stephens
- Department of Physiology and Cell Biology, the Ohio State University, Columbus, OH, USA
| | - Yutong Zhao
- Department of Physiology and Cell Biology, the Ohio State University, Columbus, OH, USA.
| |
Collapse
|
6
|
Zhang M, Li P, Wang F, Zhang S, Li H, Zhang Y, Wang X, Liu K, Li X. Separation, identification and cardiovascular activities of phospholipid classes from the head of Penaeus vannamei by lipidomics and zebrafish models. Food Funct 2021; 12:2282-2291. [DOI: 10.1039/d0fo03017a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Five phospholipid classes of Penaeus vannamei head were separated, analyzed and quantified. They had different cardiovascular activities evaluated in zebrafish models, which may provide a research basis for pharmaceutical use of marine phospholipids.
Collapse
Affiliation(s)
- Mengqi Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province
- Key Laboratory for Biosensor of Shandong Province
- Biology Institute
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
| | - Peihai Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province
- Key Laboratory for Biosensor of Shandong Province
- Biology Institute
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
| | - Fengxia Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province
- Key Laboratory for Biosensor of Shandong Province
- Biology Institute
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
| | - Shanshan Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province
- Key Laboratory for Biosensor of Shandong Province
- Biology Institute
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
| | - Haonan Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province
- Key Laboratory for Biosensor of Shandong Province
- Biology Institute
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
| | - Yun Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province
- Key Laboratory for Biosensor of Shandong Province
- Biology Institute
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
| | - Ximin Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province
- Key Laboratory for Biosensor of Shandong Province
- Biology Institute
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
| | - Kechun Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province
- Key Laboratory for Biosensor of Shandong Province
- Biology Institute
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
| | - Xiaobin Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province
- Key Laboratory for Biosensor of Shandong Province
- Biology Institute
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
| |
Collapse
|
7
|
Feng Y, Xiao M, Zhang Z, Cui R, Jiang X, Wang S, Bai H, Liu C, Zhang Z. Potential interaction between lysophosphatidic acid and tumor-associated macrophages in ovarian carcinoma. JOURNAL OF INFLAMMATION-LONDON 2020; 17:23. [PMID: 32774171 PMCID: PMC7405460 DOI: 10.1186/s12950-020-00254-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
Ovarian carcinoma is the deadliest type of gynecological cancer. The unique tumor microenvironment enables specific and efficient metastasis, weakens immunological monitoring, and mediates drug resistance. Tumor associated macrophages (TAMs) are a crucial part of the TME and are involved in various aspects of tumor behavior. Lysophosphatidic acid (LPA) is elevated in the blood of ovarian carcinoma patients, as well as in the tumor tissues and ascites, which make it a useful biomarker and a potential therapeutic target. Recent studies have shown that LPA transforms monocytes into macrophages and regulates the formation of macrophages through the AKT/mTOR pathway, and PPAR γ is a major regulator of LPA-derived macrophages. In addition, TAMs synthesize and secrete LPA and express LPA receptor (LPAR) on the surface. With these data in mind, we hypothesize that LPA can convert monocytes directly into TAMs in the microenvironment of ovarian cancer. LPA may mediate TAM formation by activating the PI3K/AKT/mTOR signaling pathway through LPAR on the cell surface, which may also affect the function of PPAR γ, leading to increased LPA production by TAMs. Thus, LPA and TAMs form a vicious circle that affects the malignant behavior of ovarian cancer.
Collapse
Affiliation(s)
- Ying Feng
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Meizhu Xiao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Zihan Zhang
- Department of Gynecology and Obstetrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ran Cui
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Xuan Jiang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Shuzhen Wang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Huimin Bai
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Chongdong Liu
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| |
Collapse
|
8
|
Hwang HJ, Park T, Kim M, Shin HS, Hwang W, Min YK, Song SG, Park D, Lee CH. A Novel Therapeutic Reagent, KA-1002 for Alleviating Lysophosphatidic Acid-Mediated Inflammation Related Gene Expression in Swine Macrophages. Animals (Basel) 2020; 10:ani10030534. [PMID: 32210054 PMCID: PMC7142756 DOI: 10.3390/ani10030534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Inflammatory diseases are a key factor reducing the productivity of animals in a livestock industrial environment. We have identified a novel lysophosphatidic acid signaling antagonist, KA-1002, which alleviates lysophosphatidic acid-mediated a broad range of inflammation related gene expression in swine macrophages. Specifically, we found that KA-1002 significantly alleviated LPA-induced genes related with inflammation such as a role of macrophages, fibroblasts and endothelial cells in rheumatoid arthritis and STAT3 signal pathway. Taken together, KA-1002 could be considered a novel therapeutic reagent candidate for swine inflammatory diseases. Abstract Stresses and various infectious reagents caused multiple inflammatory diseases in swine in a livestock industrial environment. Therefore, there is a need for an effective therapeutic or preventive agent that could alleviate chronic and acute inflammation. We found that lysophosphatidic acid (LPA), a stress-induced potent endogenous inflammatory molecule, causes a broad range-regulation of inflammation related genes inflammation in swine macrophages. We further investigated the genome scaled transcriptional regulatory effect of a novel LPA-signaling antagonist, KA-1002 on swine macrophages, inducing the alleviated LPA-mediated inflammation related gene expression. Therefore, KA-1002 could potentially serve as a novel therapeutic or preventive agent to maintain physiologically healthy and balanced conditions of pigs.
Collapse
Affiliation(s)
- Hyeon-Jeong Hwang
- Bio and Drug Discovery Division, Center for Information-Based Drug Research, Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (H.-J.H.); (M.K.); (H.-s.S.); (W.H.); (Y.K.M.)
| | - Tamina Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea;
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Korea
| | - Miok Kim
- Bio and Drug Discovery Division, Center for Information-Based Drug Research, Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (H.-J.H.); (M.K.); (H.-s.S.); (W.H.); (Y.K.M.)
- Chungnam National University School of Medicine, Daejeon 34137, Korea
| | - Hee-su Shin
- Bio and Drug Discovery Division, Center for Information-Based Drug Research, Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (H.-J.H.); (M.K.); (H.-s.S.); (W.H.); (Y.K.M.)
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Wooyeon Hwang
- Bio and Drug Discovery Division, Center for Information-Based Drug Research, Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (H.-J.H.); (M.K.); (H.-s.S.); (W.H.); (Y.K.M.)
- Department of Pharmaceutical Science, Kyunghee University, Seoul 02447, Korea
| | - Yong Ki Min
- Bio and Drug Discovery Division, Center for Information-Based Drug Research, Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (H.-J.H.); (M.K.); (H.-s.S.); (W.H.); (Y.K.M.)
| | - Suk-gil Song
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Korea;
| | - Daeui Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea;
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Korea
- Correspondence: (D.P.); (C.H.L.); Tel.: +82-42-610-8251 (D.P.); +82-42-860-7414 (C.H.L.)
| | - Chang Hoon Lee
- Bio and Drug Discovery Division, Center for Information-Based Drug Research, Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (H.-J.H.); (M.K.); (H.-s.S.); (W.H.); (Y.K.M.)
- Correspondence: (D.P.); (C.H.L.); Tel.: +82-42-610-8251 (D.P.); +82-42-860-7414 (C.H.L.)
| |
Collapse
|
9
|
KA-1002, a Novel Lysophosphatidic Acid Signaling Antagonist, Alleviates Bovine Tracheal Cell Disruption and Inflammation. Animals (Basel) 2020; 10:ani10020295. [PMID: 32069887 PMCID: PMC7070416 DOI: 10.3390/ani10020295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 02/08/2020] [Indexed: 12/23/2022] Open
Abstract
Abstract: The industrial livestock environment can cause stress and weakened immunity in cattle, leading to microbial infections which reduce productivity. As such, there is a need for an effective therapeutic agent that can alleviate uncontrolled destructive respiratory inflammation. We found that lysophosphatidic acid (LPA), a potent endogenous stress-induced inflammatory agent, causes respiratory tissue damage and triggers inflammation in bovine bronchial cells. LPA also inflames pulmonary bovine blood vessel cells to produce inflammatory cytokines. These findings strongly suggest that LPA is a highly important endogenous material exacerbating bovine respiratory diseases. We further identified a novel LPA-signaling antagonist, KA-1002, and showed that it alleviated LPA-mediated bovine tracheal cell disruption and inflammation. Therefore, KA-1002 could potentially serve as a novel therapeutic agent to maintain physiologically healthy and balanced conditions in bovine respiratory tracts.
Collapse
|
10
|
Xu Y. Targeting Lysophosphatidic Acid in Cancer: The Issues in Moving from Bench to Bedside. Cancers (Basel) 2019; 11:E1523. [PMID: 31658655 PMCID: PMC6826372 DOI: 10.3390/cancers11101523] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/02/2019] [Accepted: 10/08/2019] [Indexed: 12/16/2022] Open
Abstract
Since the clear demonstration of lysophosphatidic acid (LPA)'s pathological roles in cancer in the mid-1990s, more than 1000 papers relating LPA to various types of cancer were published. Through these studies, LPA was established as a target for cancer. Although LPA-related inhibitors entered clinical trials for fibrosis, the concept of targeting LPA is yet to be moved to clinical cancer treatment. The major challenges that we are facing in moving LPA application from bench to bedside include the intrinsic and complicated metabolic, functional, and signaling properties of LPA, as well as technical issues, which are discussed in this review. Potential strategies and perspectives to improve the translational progress are suggested. Despite these challenges, we are optimistic that LPA blockage, particularly in combination with other agents, is on the horizon to be incorporated into clinical applications.
Collapse
Affiliation(s)
- Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 950 W. Walnut Street R2-E380, Indianapolis, IN 46202, USA.
| |
Collapse
|
11
|
Zhang Q, Xu H, Liu R, Gao P, Yang X, Jin W, Zhang Y, Bi K, Li Q. A Novel Strategy for Targeted Lipidomics Based on LC-Tandem-MS Parameters Prediction, Quantification, and Multiple Statistical Data Mining: Evaluation of Lysophosphatidylcholines as Potential Cancer Biomarkers. Anal Chem 2019; 91:3389-3396. [DOI: 10.1021/acs.analchem.8b04715] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Qian Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Huarong Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Ran Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Peng Gao
- Metabolomics Core Facility of RHLCCC, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Xiao Yang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Wei Jin
- Urumqi Traditional Chinese Medicine Hospital, 590 Youhao South Road, Urumqi 830000, China
| | - Yiwen Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Qing Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
12
|
Lysophospholipid Signaling in the Epithelial Ovarian Cancer Tumor Microenvironment. Cancers (Basel) 2018; 10:cancers10070227. [PMID: 29987226 PMCID: PMC6071084 DOI: 10.3390/cancers10070227] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/03/2018] [Accepted: 07/05/2018] [Indexed: 12/12/2022] Open
Abstract
As one of the important cancer hallmarks, metabolism reprogramming, including lipid metabolism alterations, occurs in tumor cells and the tumor microenvironment (TME). It plays an important role in tumorigenesis, progression, and metastasis. Lipids, and several lysophospholipids in particular, are elevated in the blood, ascites, and/or epithelial ovarian cancer (EOC) tissues, making them not only useful biomarkers, but also potential therapeutic targets. While the roles and signaling of these lipids in tumor cells are extensively studied, there is a significant gap in our understanding of their regulations and functions in the context of the microenvironment. This review focuses on the recent study development in several oncolipids, including lysophosphatidic acid and sphingosine-1-phosphate, with emphasis on TME in ovarian cancer.
Collapse
|
13
|
Lysophosphatidic acid (LPA) as a pro-fibrotic and pro-oncogenic factor: a pivotal target to improve the radiotherapy therapeutic index. Oncotarget 2018; 8:43543-43554. [PMID: 28402936 PMCID: PMC5522168 DOI: 10.18632/oncotarget.16672] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/08/2017] [Indexed: 12/21/2022] Open
Abstract
Radiation-induced fibrosis is widely considered as a common but forsaken phenomenon that can lead to clinical sequela and possibly vital impairments. Lysophosphatidic acid is a bioactive lipid involved in fibrosis and probably in radiation-induced fibrosis as suggested in recent studies. Lysophosphatidic acid is also a well-described pro-oncogenic factor, involved in carcinogenesis processes (proliferation, survival, angiogenesis, invasion, migration). The present review highlights and summarizes the links between lysophosphatidic acid and radiation-induced fibrosis, lysophosphatidic acid and radioresistance, and proposes lysophosphatidic acid as a potential central actor of the radiotherapy therapeutic index. Besides, we hypothesize that following radiotherapy, the newly formed tumour micro-environment, with increased extracellular matrix and increased lysophosphatidic acid levels, is a favourable ground to metastasis development. Lysophosphatidic acid could therefore be an exciting therapeutic target, minimizing radio-toxicities and radio-resistance effects.
Collapse
|
14
|
Fan Q, Cai Q, Li P, Wang W, Wang J, Gerry E, Wang TL, Shih IM, Nephew KP, Xu Y. The novel ZIP4 regulation and its role in ovarian cancer. Oncotarget 2017; 8:90090-90107. [PMID: 29163813 PMCID: PMC5685734 DOI: 10.18632/oncotarget.21435] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/26/2017] [Indexed: 12/20/2022] Open
Abstract
Our RNAseq analyses revealed that ZIP4 is a top gene up-regulated in more aggressive ovarian cancer cells. ZIP4's role in cancer stem cells has not been reported in any type of cancer. In addition, the role and regulation of ZIP4, a zinc transporter, have been studied in the context of extracellular zinc transporting. Factors other than zinc with ZIP4 regulatory effects are essentially unknown. ZIP4 expression and its regulation in epithelial ovarian cancer cells was assessed by immunoblotting, quantitative PCR, or immunohistochemistry staining in human ovarian tissues. Cancer stem cell-related activities were examined to evaluate the role of ZIP4 in human high-grade serous ovarian cancer cells in vitro and in vivo. RNAi and CRISPR techniques were used to knockdown or knockout ZIP4 and related genes. Ovarian cancer tissues overexpressed ZIP4 when compared with normal and benign tissues. ZIP4 knockout significantly reduced several cancer stem cell-related activities in EOC cells, including proliferation, anoikis-resistance, colony-formation, spheroid-formation, drug-resistance, and side-population in vitro. ZIP4-expressing side-population highly expressed known CSC markers ALDH1 and OCT4. ZIP4 knockout dramatically reduced tumorigenesis and ZIP4 overexpression increased tumorigenesis in vivo. In addition, the ZIP4-expressing side-population had the tumor initiating activity. Moreover, the oncolipid lysophosphatic acid effectively up-regulated ZIP4 expression via the nuclear receptor peroxisome proliferator-activated receptor gamma and lysophosphatic acid 's promoting effects in cancer stem cell-related activities in HGSOC cells was at least partially mediated by ZIP4 in an extracellular zinc-independent manner. Our critical data imply that ZIP4 is a new and important cancer stem cell regulator in ovarian cancer. Our data also provide an innovative interpretation for the apparent disconnection between low levels of zinc and up-regulation of ZIP4 in ovarian cancer tissues.
Collapse
Affiliation(s)
- Qipeng Fan
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qingchun Cai
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Pengfei Li
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Pharmaceutical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, P.R. China
| | - Wenyan Wang
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei City, 230601, P.R. China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- MASDINO (Beijing) Medical Research Co. Ltd., Beijing, 100123, P.R. China
| | - Emily Gerry
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Tian-Li Wang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Ie-Ming Shih
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Kenneth P. Nephew
- Medical Sciences, Indiana University School of Medicine, Jordan Hall 302, Bloomington, IN 47405, USA
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
15
|
Ray U, Roy SS. Aberrant lipid metabolism in cancer cells - the role of oncolipid-activated signaling. FEBS J 2017; 285:432-443. [PMID: 28971574 DOI: 10.1111/febs.14281] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/25/2017] [Accepted: 09/26/2017] [Indexed: 12/27/2022]
Abstract
Metabolic activity of malignant cells is very different from that of their nontransformed equivalents, which establishes metabolic reprogramming as an important hallmark of every transformed cell. In particular, the current arena of research in this field aims to understand the regulatory effect of oncogenic signaling on metabolic rewiring in transformed cells in order to exploit this for therapeutic benefit. Alterations in lipid metabolism are one of the main aspects of metabolic rewiring of transformed cells. Up-regulation of several lipogenic enzymes has been reported to be a characteristic of various cancer types. Lysophosphatidic acid (LPA), a simple byproduct of the lipid biosynthesis pathway, has gained immense importance due to its elevated level in several cancers and associated growth-promoting activity. Importantly, a current study revealed its role in increased de novo lipid synthesis through up-regulation of sterol regulatory element-binding protein 1, a master regulator of lipid metabolism. This review summarizes the recent insights in the field of oncolipid LPA-mediated signaling in regard to lipid metabolism in cancers. Future work in this domain is required to understand the up-regulation of the de novo synthesis pathway and the role of its end products in malignant cells. This will open a new arena of research toward the development of specific metabolic inhibitors that can add to the pre-existing chemotherapeutics in order to increase the efficacy of clinical output in cancer patients.
Collapse
Affiliation(s)
- Upasana Ray
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| | - Sib Sankar Roy
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| |
Collapse
|
16
|
Ray U, Roy Chowdhury S, Vasudevan M, Bankar K, Roychoudhury S, Roy SS. Gene regulatory networking reveals the molecular cue to lysophosphatidic acid-induced metabolic adaptations in ovarian cancer cells. Mol Oncol 2017; 11:491-516. [PMID: 28236660 PMCID: PMC5527468 DOI: 10.1002/1878-0261.12046] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/25/2017] [Accepted: 02/15/2017] [Indexed: 12/15/2022] Open
Abstract
Extravasation and metastatic progression are two main reasons for the high mortality rate associated with cancer. The metastatic potential of cancer cells depends on a plethora of metabolic challenges prevailing within the tumor microenvironment. To achieve higher rates of proliferation, cancer cells reprogram their metabolism, increasing glycolysis and biosynthetic activities. Just why this metabolic reprogramming predisposes cells towards increased oncogenesis remains elusive. The accumulation of myriad oncolipids in the tumor microenvironment has been shown to promote the invasiveness of cancer cells, with lysophosphatidic acid (LPA) being one such critical factor enriched in ovarian cancer patients. Cellular bioenergetic studies confirm that oxidative phosphorylation is suppressed and glycolysis is increased with long exposure to LPA in ovarian cancer cells compared with non‐transformed epithelial cells. We sought to uncover the regulatory complexity underlying this oncolipid‐induced metabolic perturbation. Gene regulatory networking using RNA‐Seq analysis identified the oncogene ETS‐1 as a critical mediator of LPA‐induced metabolic alterations for the maintenance of invasive phenotype. Moreover, LPA receptor‐2 specific PtdIns3K‐AKT signaling induces ETS‐1 and its target matrix metalloproteases. Abrogation of ETS‐1 restores cellular bioenergetics towards increased oxidative phosphorylation and reduced glycolysis, and this effect was reversed by the presence of LPA. Furthermore, the bioenergetic status of LPA‐treated ovarian cancer cells mimics hypoxia through induction of hypoxia‐inducible factor‐1α, which was found to transactivate ets‐1. Studies in primary tumors generated in syngeneic mice corroborated the in vitro findings. Thus, our study highlights the phenotypic changes induced by the pro‐metastatic factor ETS‐1 in ovarian cancer cells. The relationship between enhanced invasiveness and metabolic plasticity further illustrates the critical role of metabolic adaptation of cancer cells as a driver of tumor progression. These findings reveal oncolipid‐induced metabolic predisposition as a new mechanism of tumorigenesis and propose metabolic inhibitors as a potential approach for future management of aggressive ovarian cancer.
Collapse
Affiliation(s)
- Upasana Ray
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| | - Shreya Roy Chowdhury
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| | | | - Kiran Bankar
- Bionivid Technology Private Limited, Bangalore, India
| | | | - Sib Sankar Roy
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| |
Collapse
|
17
|
Ray U, Roy SS, Chowdhury SR. Lysophosphatidic Acid Promotes Epithelial to Mesenchymal Transition in Ovarian Cancer Cells by Repressing SIRT1. Cell Physiol Biochem 2017; 41:795-805. [PMID: 28214851 DOI: 10.1159/000458744] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/21/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND/AIMS Epithelial-to-mesenchymal transition (EMT) plays an essential role in the transition from early to invasive phenotype, however the underlying mechanisms still remain elusive. Herein, we propose a mechanism through which the class-III deacetylase SIRT1 regulates EMT in ovarian cancer (OC) cells. METHODS Expression analysis was performed using Q-PCR, western blot, immunofluorescence and fluorescence-IHC study. Matrigel invasion assay was used for the invasion study. Morphological alterations were observed by phalloidin-staining. Co-immunoprecipitation study was performed to analyze protein-protein interaction. RESULTS Overexpression of SIRT1-WT as well as Resveratrol-mediated SIRT1 activation antagonized the invasion of OC cells by suppressing EMT. SIRT1 deacetylates HIF1α, to inactivate its transcriptional activity. To further validate HIF1α inactivation, its target gene, i.e. ZEB1, an EMT-inducing factor was found to attenuate upon SIRT1 activation. To uncover the regulatory factor governing SIRT1 expression, lysophosphatidic acid (LPA), a highly enriched oncolipid in ascites/serum of OC patients, was found to down-regulate SIRT1 expression. Importantly, LPA was found to induce the mesenchymal switch in OC cells through suppression of SIRT1. Decreased level of SIRT1 was further validated in ovarian tissue samples of OC patients. CONCLUSION We have identified a mechanism that relates SIRT1 down-regulation to LPA-induced EMT in OC cells and may open new arenas on developing novel anti-cancer therapeutics.
Collapse
|
18
|
Lorke DE, Stegmeier-Petroianu A, Petroianu GA. Biologic activity of cyclic and caged phosphates: a review. J Appl Toxicol 2016; 37:13-22. [DOI: 10.1002/jat.3369] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 07/01/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Dietrich E. Lorke
- Department of Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine; Florida International University; Miami Florida USA
| | - Anka Stegmeier-Petroianu
- Mannheim Institute of Public Health, Social and Preventive Medicine; Medical Faculty Mannheim, Heidelberg University; D-68167 Mannheim Germany
| | - Georg A. Petroianu
- Department of Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine; Florida International University; Miami Florida USA
| |
Collapse
|
19
|
Fingolimod promotes peripheral nerve regeneration via modulation of lysophospholipid signaling. J Neuroinflammation 2016; 13:143. [PMID: 27283020 PMCID: PMC4901498 DOI: 10.1186/s12974-016-0612-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 06/06/2016] [Indexed: 12/26/2022] Open
Abstract
Background The lysophospholipids sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) are pleiotropic signaling molecules with a broad range of physiological functions. Targeting the S1P1 receptor on lymphocytes with the immunomodulatory drug fingolimod has proven effective in the treatment of multiple sclerosis. An emerging body of experimental evidence points to additional direct effects on cells of the central and peripheral nervous system. Furthermore, fingolimod has been reported to reduce LPA synthesis via inhibition of the lysophospholipase autotaxin. Here we investigated whether modulation of particular signaling aspects of S1P as well as LPA by fingolimod might propagate peripheral nerve regeneration in vivo and independent of its anti-inflammatory potency. Methods Sciatic nerve crush was performed in wildtype C57BL/6, in immunodeficient Rag1−/− and Foxn1−/− mice. Analyses were based on walking track analysis and electrophysiology, histology, and cAMP formation. Quantification of different LPA species was performed by liquid chromatography coupled to tandem mass spectrometry. Furthermore, functional consequences of autotaxin inhibition by the specific inhibitor PF-8380 and the impact of fingolimod on early cytokine release in the injured sciatic nerve were investigated. Results Clinical and electrophysiological measures indicated an improvement of nerve regeneration under fingolimod treatment that is partly independent of its anti-inflammatory properties. Fingolimod treatment correlated with a significant elevation of axonal cAMP, a crucial factor for axonal outgrowth. Additionally, fingolimod significantly reduced LPA levels in the injured nerve. PF-8380 treatment correlated with improved myelin thickness. Sciatic nerve cytokine levels were not found to be significantly altered by fingolimod treatment. Conclusions Our findings provide in vivo evidence for direct effects of fingolimod on cells of the peripheral nervous system that may propagate nerve regeneration via a dual mode of action, differentially affecting axonal outgrowth and myelination by modulating relevant aspects of S1P and LPA signaling.
Collapse
|
20
|
Cai H, Xu Y. The role of LPA and YAP signaling in long-term migration of human ovarian cancer cells. Cell Commun Signal 2013; 11:31. [PMID: 23618389 PMCID: PMC3655373 DOI: 10.1186/1478-811x-11-31] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/09/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The Hippo-YAP signaling pathway is altered and implicated as oncogenic in many human cancers. However, extracellular signals that regulate the mammalian Hippo pathway have remained elusive until very recently when it was shown that the Hippo pathway is regulated by G-protein-coupled receptor (GPCR) ligands including lysophosphatidic acid (LPA) and sphingosine 1-phosphophate (S1P). LPA inhibits Lats kinase activity in HEK293 cells, but the potential involvement of a protein phosphatase was not investigated. The extracellular regulators of YAP dephosphorylation (dpYAP) and nuclear translocation in epithelial ovarian cancer (EOC) are essentially unknown. RESULTS We showed here that LPA dose- and time-dependently induced dpYAP in human EOC cell lines OVCA433, OVCAR5, CAOV3, and Monty-1, accompanied by increased YAP nuclear translocation. YAP was involved in LPA-induced migration and invasion of EOC cells and LPA3 was a major LPA receptor mediating the migratory effect. We demonstrated that G13, but not or to a lesser extent G12, Gi or Gq, was necessary for LPA-induced dpYAP and its nuclear translocation and that RhoA-ROCK, but not RhoB, RhoC, Rac1, cdc42, PI3K, ERK, or AKT, were required for the LPA-dpYAP effect. In contrast to results in HEK293 cells, LPA did not inhibit Mst and Lats kinase in OVCA433 EOC cells. Instead, protein phosphatase 1A (PP1A) acted down-stream of RhoA in LPA-induction of dpYAP. In addition, we identified that amphiregulin (AREG), a down-stream target of YAP which activated EGF receptors (EGFR), mediated an LPA-stimulated and EGFR-dependent long-term (16 hr) cell migration. This process was transcription- and translation-dependent and was distinct from a transcription- and YAP-independent short-term (4 hr) cell migration. EOC tissues had reduced pYAP levels compared to normal and benign ovarian tissues, implying the involvement of dpYAP in EOC pathogenesis, as well as its potential marker and/or target values. CONCLUSIONS A novel LPA-LPA3-G13-RhoA-ROCK-PP1A-dpYAP-AREG-EGFR signaling pathway was linked to LPA-induced migration of EOC cells. Reduced pYAP levels were demonstrated in human EOC tumors as compared to both normal ovarian tissues and benign gynecologic masses. Our findings support that YAP is a potential marker and target for developing novel therapeutic strategies against EOC.
Collapse
Affiliation(s)
- Hui Cai
- First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 975 W. Walnut St. IB355A, Indianapolis, IN 46202, USA
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 975 W. Walnut St. IB355A, Indianapolis, IN 46202, USA
| |
Collapse
|
21
|
Korkina O, Dong Z, Marullo A, Warshaw G, Symons M, Ruggieri R. The MLK-related kinase (MRK) is a novel RhoC effector that mediates lysophosphatidic acid (LPA)-stimulated tumor cell invasion. J Biol Chem 2013; 288:5364-73. [PMID: 23319595 DOI: 10.1074/jbc.m112.414060] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The small GTPase RhoC is overexpressed in many invasive tumors and is essential for metastasis. Despite its high structural homology to RhoA, RhoC appears to perform functions that are different from those controlled by RhoA. The identity of the signaling components that are differentially regulated by these two GTPases is only beginning to emerge. Here, we show that the MAP3K protein MRK directly binds to the GTP-bound forms of both RhoA and RhoC in vitro. However, siRNA-mediated depletion of MRK in cells phenocopies depletion of RhoC, rather than that of RhoA. MRK depletion, like that of RhoC, inhibits LPA-stimulated cell invasion, while depletion of RhoA increases invasion. We also show that active MRK enhances LPA-stimulated invasion, further supporting a role for MRK in the regulation of invasion. Depletion of either RhoC or MRK causes sustained myosin light chain phosphorylation after LPA stimulation. In addition, activation of MRK causes a reduction in myosin light chain phosphorylation. In contrast, as expected, depletion of RhoA inhibits myosin light chain phosphorylation. We also present evidence that both RhoC and MRK are required for LPA-induced stimulation of the p38 and ERK MAP kinases. In conclusion, we have identified MRK as a novel RhoC effector that controls LPA-stimulated cell invasion at least in part by regulating myosin dynamics, ERK and p38.
Collapse
Affiliation(s)
- Olga Korkina
- Oncology and Cell Biology Center, The Feinstein Institute for Medical Research, Manhasset, New York 11030, USA
| | | | | | | | | | | |
Collapse
|
22
|
Zhu C, Dane A, Spijksma G, Wang M, van der Greef J, Luo G, Hankemeier T, Vreeken RJ. An efficient hydrophilic interaction liquid chromatography separation of 7 phospholipid classes based on a diol column. J Chromatogr A 2012; 1220:26-34. [DOI: 10.1016/j.chroma.2011.11.034] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Revised: 11/08/2011] [Accepted: 11/18/2011] [Indexed: 11/26/2022]
|
23
|
Li L, Tam L, Liu L, Jin T, Ng DS. Wnt-signaling mediates the anti-adipogenic action of lysophosphatidic acid through cross talking with the Rho/Rho associated kinase (ROCK) pathway. Biochem Cell Biol 2011; 89:515-21. [PMID: 21999426 DOI: 10.1139/o11-048] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid with a diverse range of biological activities including the modulation of adipogenesis. Treatment of 3T3-L1 cells and 3T3F44A cells with LPA inhibits adipogenesis and reduces expression of PPARγ through activation of RhoGTPase and its downstream Rho associated kinase (ROCK). The mechanism of suppression of PPARγ expression by Rho/ROCK is poorly understood. By treating the differentiating 3T3-L1 cells with various combinations of LPA and ROCK inhibitors, Y-27632 and fasudil, we observed that LPA treatment resulted in attenuation of adipogenesis and a significant reduction in PPARγ mRNA as early as 3 d post-induction. LPA treatment also resulted in significant but delayed upregulation of components of the canonical Wnt signaling, namely Wnt10b mRNA, β-catenin protein, and mRNA expression of β-catenin target genes, detectable at day 7, but not day 3. Treatment of the 3T3-L1 cells with ROCK inhibitors Y-27632 and fasudil revealed a tonic activation of β-catenin/target genes by ROCK. This study identified the existence of a novel cross talk between the Rho/ROCK pathway and the Wnt-signaling pathway. The LPA/Rho/ROCK pathway inhibits expression of PPARγ and adipogenesis in part through a delayed activation of the canonical Wnt-signaling pathway based on increased Wnt10b expression and β-catenin induction.
Collapse
Affiliation(s)
- L Li
- Keenan Research Centre, LiKaShing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | | | | | | | | |
Collapse
|
24
|
Li H, Zhao Z, Antalis C, Zhao Z, Emerson R, Wei G, Zhang S, Zhang ZY, Xu Y. Combination therapy of an inhibitor of group VIA phospholipase A2 with paclitaxel is highly effective in blocking ovarian cancer development. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:452-61. [PMID: 21703423 DOI: 10.1016/j.ajpath.2011.03.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 03/03/2011] [Accepted: 03/21/2011] [Indexed: 02/04/2023]
Abstract
We and others have shown that calcium-independent phospholipase A(2) (iPLA(2)) is involved in epithelial ovarian cancer (EOC). Hence, we propose that iPLA(2) is a potential effective and novel target for EOC. We tested this concept and found that bromoenol lactone (BEL), a selective inhibitor of iPLA(2), significantly inhibited EOC metastatic tumor growth in mouse xenograft models using human SKOV3 and HEY ovarian cancer cells. Moreover, the combination of BEL with paclitaxel (PTX), one of the most commonly used therapeutic agents in EOC, almost completely blocked tumor development in the xenograft mouse model. BEL showed no detectable cytotoxic effects in mice. Another iPLA(2) inhibitor, FKGK11, also inhibited tumor development in the xenograft mouse model, supporting that the major target of action was iPLA(2). The additional effects of BEL with PTX in vivo likely stem from their distinct cellular effects. BEL and FKGK11 reduced adhesion, migration, and invasion of EOC cells in vitro; the reduced ability to adhere, migrate, and invade seems to increase the vulnerability of tumor cells to PTX. These results provide an important basis for the development of new treatment modalities for EOC.
Collapse
Affiliation(s)
- Hui Li
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Li H, Zhao Z, Wei G, Yan L, Wang D, Zhang H, Sandusky GE, Turk J, Xu Y. Group VIA phospholipase A2 in both host and tumor cells is involved in ovarian cancer development. FASEB J 2010; 24:4103-16. [PMID: 20530749 PMCID: PMC2996900 DOI: 10.1096/fj.10-161356] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2010] [Accepted: 05/20/2010] [Indexed: 11/11/2022]
Abstract
Host-tumor cell interactions are recognized to be critical in tumor development. We have shown that group VIA phospholipase A(2) [calcium-independent phospholipase A(2)β (iPLA(2)β)] is important in regulating extracellular lysophosphatidic acid (LPA) levels around human epithelial ovarian cancer (EOC) cells. To explore the role of iPLA(2)β in host-tumor cell interactions, we have used immunocompetent iPLA(2)β knockout (iPLA(2)β(-/-)) mice and the mouse EOC cell line ID8. Tumorigenesis and ascites formation were reduced in iPLA(2)β(-/-) mice compared with wild-type (WT) mice by more >50% and were reduced further when ID8 cell iPLA(2)β levels were lowered (by>95%) with shRNA. LPA and lysophosphatidylcholine (LPC) levels in the tumor microenvironment were reduced to ∼80% of WT levels in iPLA(2)β(-/-) mice. LPA, but not LPC, stimulated ID8 cell migration and invasion with cells in which iPLA(2)β expression had been down-regulated in vitro. LPA, but not LPC, also enhanced in vivo ascites formation (by ∼5-fold) and tumorigenesis in iPLA(2)β(-/-) mice. This is the first demonstration of a role for host cell iPLA(2)β in cancer, and these findings suggest that iPLA(2)β is a potential target for developing novel antineoplastic therapeutic strategies.
Collapse
Affiliation(s)
- Hui Li
- Department of Obstetrics and Gynecology and
| | | | - Gang Wei
- Department of Obstetrics and Gynecology and
| | - Libo Yan
- Department of Obstetrics and Gynecology and
| | | | - Hong Zhang
- Department of Obstetrics and Gynecology and
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana, USA
- Department of General Surgery, Second Hospital of Jilin University, Changchun, Jilin, China; and
| | - George Earl Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana, USA
- Department of General Surgery, Second Hospital of Jilin University, Changchun, Jilin, China; and
| | - John Turk
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yan Xu
- Department of Obstetrics and Gynecology and
| |
Collapse
|
26
|
Immunohistochemical Detection of Autotaxin (ATX)/Lysophospholipase D (lysoPLD) in Submucosal Invasive Colorectal Cancer. J Gastrointest Cancer 2010; 42:204-11. [DOI: 10.1007/s12029-010-9186-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
27
|
Wang C, Michener CM, Belinson JL, Vaziri S, Ganapathi R, Sengupta S. Role of the 18:1 lysophosphatidic acid-ovarian cancer immunoreactive antigen domain containing 1 (OCIAD1)-integrin axis in generating late-stage ovarian cancer. Mol Cancer Ther 2010; 9:1709-18. [PMID: 20515946 DOI: 10.1158/1535-7163.mct-09-1024] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chemotherapy resistance in ovarian cancer remains an unsolved problem in caring for women with this disease. We now show that ovarian cancer immunoreactive antigen domain containing 1 (OCIAD1) has higher expression in chemoresistant compared with chemosensitive ovarian cancer cell lines. We have designed a novel secondary cell homing assay (SCHA) to test the ability of cells to withstand chemotherapy and form secondary colonies that could form recurrent disease. OCIAD1 upregulated cells had significantly higher secondary colony-forming ability than had OCIAD1 downregulated cells following treatment with paclitaxel. Additionally, 18:1 lysophosphatidic acid (LPA) increases OCIAD1 expression in a time- and dose-dependent manner. LPA stimulates OCIAD1 serine phosphorylation within two hours of stimulation. Transfection of MKK6 increases OCIAD1 expression but nuclear translocation is inhibited. Inhibition of p38 mitogen-activated protein kinase blocks LPA-induced OCIAD1 expression. Cycloheximide treatment of MKK6-transfected cells does not inhibit OCIAD1 expression, suggesting that MKK6 upregulation is not translationally controlled. OCIAD1 downregulation knocks down LPA-induced cell adhesion to collagen I and laminin 10/11 and specifically inhibits cell attachment to alpha2, alpha5, alphaV, and beta1 integrins. Proteomic studies indicate that OCIAD1 is physically attached to alpha actin 4 and beta actin. Thus, OCIAD1 may play a role in cytoskeletal function which can alter sensitivity to paclitaxel. This is the first study to indicate that OCIAD1 is a key player in generating ovarian cancer recurrence; it is functionally controlled by LPA and MKK6 signaling, and inhibition of OCIAD1 could be an important strategy in the management of recurrent ovarian cancer.
Collapse
Affiliation(s)
- Chunyan Wang
- Gynecology Oncology Section, Department of Obstetrics and Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
28
|
Müller R, Berliner C, Leptin J, Pörtner D, Bialecki W, Kleuser B, Schumacher U, Milićević NM. Expression of sphingosine-1-phosphate receptors and lysophosphatidic acid receptors on cultured and xenografted human colon, breast, melanoma, and lung tumor cells. Tumour Biol 2010; 31:341-9. [PMID: 20480410 DOI: 10.1007/s13277-010-0043-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Accepted: 04/21/2010] [Indexed: 12/25/2022] Open
Abstract
The lysophospholipids sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) are small lipid molecules with a variety of physiological roles. Additionally, their involvement in the initiation and progression of malignant tumors has been increasingly recognized in recent years. However, the data on the expression of S1P and LPA receptors on different cancer cells are very few. Real-time polymerase chain reaction was used for the analysis of mRNA expression of five S1P((1-5)) and three LPA((1-3)) receptors on a large panel of 13 colon, breast, melanoma, and lung cancer cell lines. Furthermore, the modulation of S1P and LPA receptor mRNA expression was studied upon xenotransplantation of tumor cells into severe combined immunodeficient (SCID) mice. The S1P and LPA receptors were expressed to a variable degree on all tumor cell lines tested (with exception of colon cancer SW480). Most notably, tumor cell lines in vitro expressed S1P(2) mRNA that was down-regulated upon xenotransplantation, whereas LPA(2) receptor mRNA was strongly expressed both in vitro and in vivo (except by breast cancer cells). The latter was especially distinctive for small cell lung tumor cells. The S1P and LPA receptors are differentially expressed on tumor cell lines in vitro. Their expression is modulated upon xenografting into SCID mice in vivo.
Collapse
Affiliation(s)
- Reinhard Müller
- Center for Experimental Medicine, Institute of Anatomy II: Experimental Morphology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
D'Arrigo P, Servi S. Synthesis of lysophospholipids. Molecules 2010; 15:1354-77. [PMID: 20335986 PMCID: PMC6257299 DOI: 10.3390/molecules15031354] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 03/04/2010] [Accepted: 03/05/2010] [Indexed: 12/01/2022] Open
Abstract
New synthetic methods for the preparation of biologically active phospholipids and lysophospholipids (LPLs) are very important in solving problems of membrane-chemistry and biochemistry. Traditionally considered just as second-messenger molecules regulating intracellular signalling pathways, LPLs have recently shown to be involved in many physiological and pathological processes such as inflammation, reproduction, angiogenesis, tumorogenesis, atherosclerosis and nervous system regulation. Elucidation of the mechanistic details involved in the enzymological, cell-biological and membrane-biophysical roles of LPLs relies obviously on the availability of structurally diverse compounds. A variety of chemical and enzymatic routes have been reported in the literature for the synthesis of LPLs: the enzymatic transformation of natural glycerophospholipids (GPLs) using regiospecific enzymes such as phospholipases A1 (PLA1), A2 (PLA2) phospholipase D (PLD) and different lipases, the coupling of enzymatic processes with chemical transformations, the complete chemical synthesis of LPLs starting from glycerol or derivatives. In this review, chemo-enzymatic procedures leading to 1- and 2-LPLs will be described.
Collapse
Affiliation(s)
- Paola D'Arrigo
- Dipartimento di Chimica, Materiali ed Ingegneria Chimica Giulio Natta, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy. paola.d'
| | | |
Collapse
|
30
|
Tomar A, George SP, Mathew S, Khurana S. Differential effects of lysophosphatidic acid and phosphatidylinositol 4,5-bisphosphate on actin dynamics by direct association with the actin-binding protein villin. J Biol Chem 2010; 284:35278-82. [PMID: 19808673 DOI: 10.1074/jbc.c109.060830] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
We have previously reported that the epithelial cell-specific actin-binding protein villin directly associates with phosphatidylinositol 4,5-bisphosphate (PIP(2)) through three binding sites that overlap with actin-binding sites in villin. As a result, association of villin with PIP(2) inhibits actin depolymerization and enhances actin cross-linking by villin. In this study, we demonstrate that these three PIP(2)-binding sites also bind the more hydrophilic phospholipid, lysophosphatidic acid (LPA) but with a higher affinity than PIP(2) (dissociation constant (K(d)) of 22 mum versus 39.5 mum for PIP(2)). More interestingly, unlike PIP(2), the association of villin with LPA inhibits all actin regulatory functions of villin. In addition, unlike PIP(2), LPA dramatically stimulates the tyrosine phosphorylation of villin by c-Src kinase. These studies suggest that in cells, selective interaction of villin with either PIP(2) or LPA could have dramatically different outcomes on actin reorganization as well as phospholipid-regulated cell signaling. These studies provide a novel regulatory mechanism for phospholipid-induced changes in the microfilament structure and cell function and suggest that LPA could be an intracellular regulator of the actin cytoskeleton.
Collapse
Affiliation(s)
- Alok Tomar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | |
Collapse
|
31
|
Zhao Z, Xu Y. An extremely simple method for extraction of lysophospholipids and phospholipids from blood samples. J Lipid Res 2009; 51:652-9. [PMID: 19783525 DOI: 10.1194/jlr.d001503] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lipids, lysophospholipids and phospholipids in particular, have been shown to be biomarkers and potential therapeutic targets for human diseases. While many extraction and analytical methods have been developed for quantitative analyses of these molecules, most of them are laborious and time-consuming, with associated issues of poor reproducibility. This becomes one of the critical bottle-necks to move lipid markers to clinics. In the current work, we have developed an extremely simple method for lysophospholipids and phospholipids extraction from human plasma or serum samples, which only utilizes a single methanol (MeOH) solvent and involves a single step of centrifugation. This method has been subjected to strict validation by comparing it with classical lipid extraction methods. This simple method will be extremely useful for the lipidomic, diseases marker, and lipid biochemistry fields not only for its potential wide applications associated with its simplicity and reproducibility, but also for its impact in moving lipid markers into clinics through high-throughput processing.
Collapse
Affiliation(s)
- Zhenwen Zhao
- Department of Obstetrics and Gynecology, Indiana University Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
32
|
Li H, Wang D, Zhang H, Kirmani K, Zhao Z, Steinmetz R, Xu Y. Lysophosphatidic acid stimulates cell migration, invasion, and colony formation as well as tumorigenesis/metastasis of mouse ovarian cancer in immunocompetent mice. Mol Cancer Ther 2009; 8:1692-701. [PMID: 19509252 DOI: 10.1158/1535-7163.mct-08-1106] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have already established human xenographic models for the effect of lysophosphatidic acid (LPA) on tumor metastasis in vivo. The purpose of this work is to establish a preclinical LPA effect model in immunocompetent mice. We first characterized the mouse epithelial ovarian cancer (EOC) cell line ID8 for its responsiveness to LPA in cell proliferation, migration, and invasion and compared these properties with those of human EOC. The signaling pathways related to cell migration were further investigated using pharmacologic and genetic approaches. The effects of LPA on the tumorigenesis of ID8 cells and mouse survival were then examined using two different mouse models (i.p. and orthotopic injections). LPA stimulated cell proliferation, migration, and invasion of mouse EOC ID8 cells in a manner closely resembling its activity in human EOC cells. The signaling pathways involved in LPA-induced cell migration in ID8 cells were also similar to those identified in human EOC cells. We have identified cyclooxygenase-1 and 15-lipoxygenase as two new signaling molecules involved in LPA-induced cell migration in both human and mouse EOC cells. In addition, LPA enhanced the tumorigenesis/metastasis of ID8 cell in vivo as assessed by increased tumor size, early onset of ascites formation, and reduced animal survival. We have established the first LPA-EOC preclinical model in immunocompetent mice. Because ID8 cells respond to LPA similar to human EOC cells, this model is very valuable in developing and testing therapeutic reagents targeting LPA in EOC.
Collapse
Affiliation(s)
- Hui Li
- Indiana University-Purdue University Indianapolis, 975 West Walnut Street, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Affiliation(s)
- Yan Xu
- Department of Obstetrics and Gynecology, Indiana University, 975 W. Walnut St., IB355A, Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
34
|
Kalari S, Zhao Y, Spannhake EW, Berdyshev EV, Natarajan V. Role of acylglycerol kinase in LPA-induced IL-8 secretion and transactivation of epidermal growth factor-receptor in human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2008; 296:L328-36. [PMID: 19112101 DOI: 10.1152/ajplung.90431.2008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
LPA (lysophosphatidic acid) is a potent bioactive phospholipid, which regulates a number of diverse cellular responses through G protein-coupled LPA receptors. Intracellular LPA is generated by the phosphorylation of monoacylglycerol by acylglycerol kinase (AGK); however, the role of intracellular LPA in signaling and cellular responses remains to be elucidated. Here, we investigated signaling pathways of IL-8 secretion mediated by AGK and intracellular LPA in human bronchial epithelial cells (HBEpCs). Expression of AGK in HBEpCs was detected by real-time PCR, and overexpressed AGK was mainly localized in mitochondria as determined by immunofluorescence and confocal microscopy. Overexpression of lentiviral AGK wild type increased intracellular LPA production ( approximately 1.8-fold), enhanced LPA-mediated IL-8 secretion, and stimulated tyrosine phosphorylation epidermal growth factor-receptor (EGF-R). Furthermore, downregulation of native AGK by AGK small interfering RNA decreased intracellular LPA levels ( approximately 2-fold) and attenuated LPA-induced p38 MAPK, JNK, and NF-kappaB activation, tyrosine phosphorylation of EGF-R, and IL-8 secretion. These results suggest that native AGK regulates LPA-mediated IL-8 secretion involving MAPKs, NF-kappaB, and transactivation of EGF-R. Thus AGK may play an important role in innate immunity and airway remodeling during inflammation.
Collapse
Affiliation(s)
- Satish Kalari
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, USA
| | | | | | | | | |
Collapse
|
35
|
Cohen A, Sagron R, Somech E, Segal-Hayoun Y, Zilberberg N. Pain-associated signals, acidosis and lysophosphatidic acid, modulate the neuronal K(2P)2.1 channel. Mol Cell Neurosci 2008; 40:382-9. [PMID: 19130888 DOI: 10.1016/j.mcn.2008.12.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 11/28/2008] [Accepted: 12/11/2008] [Indexed: 11/27/2022] Open
Abstract
Pain is a physiological state promoting protective responses to harmful episodes. However, pain can become pathophysiological and become a chronic disruptive condition, damaging quality of life. The mammalian K(2P)2.1 (KCNK2, TREK-1) channel, expressed in sensory neurons of the dorsal root ganglia, was previously identified as a polymodal molecular sensor involved in pain perception. Here, we report that two pain-associated signals, external acidosis and lysophosphatidic acid (LPA), known to rise during injury, inflammation and cancer, profoundly down-modulate human K(2P)2.1 activity. The pH regulatory effect was mediated by activation of proton-sensitive G-protein coupled receptors and phospholipase C. Physiological concentrations of LPA overcame the effects of known K(2P)2.1 activators, such as arachidonic acid, lysophosphatidylcholine and temperature, by activating cell-surface receptors stimulating the G(q) pathway. Furthermore, we identified three K(2P)2.1 carboxy-terminal residues that mediate both pH and LPA regulatory effects. Our results highlight the important role of K(2P)2.1 channels as receptors for mediators known to cause nociception.
Collapse
Affiliation(s)
- Asi Cohen
- Department of Life Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | |
Collapse
|
36
|
Yea K, Kim J, Lim S, Kwon T, Park HS, Park KS, Suh PG, Ryu SH. Lysophosphatidylserine regulates blood glucose by enhancing glucose transport in myotubes and adipocytes. Biochem Biophys Res Commun 2008; 378:783-8. [PMID: 19063864 DOI: 10.1016/j.bbrc.2008.11.122] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Accepted: 11/23/2008] [Indexed: 12/27/2022]
Abstract
Lysophosphatidylserine (LPS) is known to have diverse cellular effects, but although LPS is present in many biological fluids, its in vivo effects have not been elucidated. In the present study, we investigated the effects of LPS on glucose metabolism in vivo, and how skeletal muscle cells respond to LPS stimulation. LPS enhanced glucose uptake in a dose- and time-dependent manner in L6 GLUT4myc myotubes, and this effect of LPS on glucose uptake was mediated by a Galpha(i) and PI 3-kinase dependent signal pathway. LPS increased the level of GLUT4 on the cell surface of L6 GLUT4myc myotubes, and enhanced glucose uptake in 3T3-L1 adipocytes. In line with its cellular functions, LPS lowered blood glucose levels in normal mice, while leaving insulin secretion unaffected. LPS also had a glucose-lowering effect in STZ-treated type 1 diabetic mice and in obese db/db type 2 diabetic mice. This study shows that LPS-stimulated glucose transport both in skeletal muscle cells and adipocytes, and significantly lowered blood glucose levels both in type 1 and 2 diabetic mice. Our results suggest that LPS is involved in the regulation of glucose homeostasis in skeletal muscle and adipose tissue.
Collapse
Affiliation(s)
- Kyungmoo Yea
- Division of Molecular and Life Science, Pohang University of Science and Technology, Pohang, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Gustin C, Van Steenbrugge M, Raes M. LPA modulates monocyte migration directly and via LPA-stimulated endothelial cells. Am J Physiol Cell Physiol 2008; 295:C905-14. [PMID: 18632732 DOI: 10.1152/ajpcell.00544.2007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lysophospholipid ligand present in oxidized low-density lipoprotein. The effects of LPA were investigated, first separately on endothelial cells (EC) and monocytes. Using Ki16425 (an LPA(1) and LPA(3) receptor antagonist), GW9662 [a peroxisome proliferator-activator receptor (PPARgamma) antagonist], and pertussis toxin (that inhibits G(i/o)), we demonstrate that LPA enhances IL-8 and monocyte chemoattractant protein-1 expression through a LPA(1)-, LPA(3)-, G(i/o)- and PPARgamma-dependent manner in the EAhy926 cells. The effect of LPA on chemokine overexpression was confirmed in human umbilical vein endothelial cells. LPA was able to enhance monocyte migration at concentrations <1 microM and to inhibit their migration at LPA concentrations >1 microM, as demonstrated by using a chemotaxis assay. We then investigated the effects of LPA on the cross-talk between EC and monocytes by evaluating the chemotactic activity in the supernatants of LPA-treated EC. At 1 microM LPA, both cell types respond cooperatively, favoring monocyte migration. At higher LPA concentration (25 microM), the chemotactic response varies as a function of time. After 4 h, the chemotactic effect of the cytokines secreted by the EC is counteracted by the direct inhibitory effect of LPA on monocytes. For longer periods of time (24 h), we observe a monocyte migration, probably due to lowered concentrations of bioactive LPA, given the induction of lipid phosphate phosphatase-2 in monocytes that may inactivate LPA. These results suggest that LPA activates EC to secrete chemokines that in combination with LPA itself might favor or not favor interactions between endothelium and circulating monocytes.
Collapse
Affiliation(s)
- Cindy Gustin
- Laboratory of Biochemistry and Cellular Biology, Unit of Research on Cellular Biology (URBC), Facultés Universitaires Notre-Dame de la Paix, University of Namur, Namur, Belgium.
| | | | | |
Collapse
|
38
|
Ookoshi T, Hasegawa K, Ohhashi Y, Kimura H, Takahashi N, Yoshida H, Miyazaki R, Goto Y, Naiki H. Lysophospholipids induce the nucleation and extension of 2-microglobulin-related amyloid fibrils at a neutral pH. Nephrol Dial Transplant 2008; 23:3247-55. [DOI: 10.1093/ndt/gfn231] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
39
|
A lysophosphatidic acid receptor lacking the PDZ-binding domain is constitutively active and stimulates cell proliferation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1783:748-59. [PMID: 18157949 DOI: 10.1016/j.bbamcr.2007.11.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 11/13/2007] [Accepted: 11/28/2007] [Indexed: 11/22/2022]
Abstract
Lysophosphatidic acid (LPA) is an extracellular signaling lipid that regulates cell proliferation, survival, and motility of normal and cancer cells. These effects are produced through G protein-coupled LPA receptors, LPA(1) to LPA(5). We generated an LPA(1) mutant lacking the SerValVal sequence of the C-terminal PDZ-binding domain to examine the role of this domain in intracellular signaling and other cellular functions. B103 neuroblastoma cells expressing the mutant LPA(1) showed rapid cell proliferation and tended to form colonies under serum-free conditions. The enhanced cell proliferation of the mutant cells was inhibited by exogenous expression of the plasmids inhibiting G proteins including G(betagamma), G(alphai) and G(alphaq) or G(alpha12/13), or treatment with pertussis toxin, phosphoinositide 3-kinase (PI3K) inhibitors or a Rho inhibitor. We confirmed that the PI3K-Akt and Rho pathways were intrinsically activated in mutant cells by detecting increases in phosphorylated Akt in western blot analyses or by directly measuring Rho activity. Interestingly, expression of the mutant LPA(1) in non-tumor mouse fibroblasts induced colony formation in a clonogenic soft agar assay, indicating that oncogenic pathways were activated. Taken together, these observations suggest that the mutant LPA(1) constitutively activates the G protein signaling leading to PI3K-Akt and Rho pathways, resulting in enhanced cell proliferation.
Collapse
|
40
|
Chen J, Baydoun AR, Xu R, Deng L, Liu X, Zhu W, Shi L, Cong X, Hu S, Chen X. Lysophosphatidic acid protects mesenchymal stem cells against hypoxia and serum deprivation-induced apoptosis. Stem Cells 2007; 26:135-45. [PMID: 17932426 DOI: 10.1634/stemcells.2007-0098] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) have shown great promise for cardiac repair. However, poor viability of transplanted MSCs within the ischemic heart has limited their therapeutic potential. Our previous studies have documented that hypoxia and serum deprivation (hypoxia/SD), induced MSCs apoptosis through the mitochondrial apoptotic pathway. Since serum lysophosphatidic acid (LPA) levels are known to be significantly elevated after acute myocardial infarction and that LPA enhanced survival of other cell systems, we embarked on determining whether LPA protects MSCs against hypoxia/SD-induced apoptosis. We have also investigated the potential mechanism(s) that may mediate such actions of LPA. All experiments were carried out on rat bone marrow MSCs. Apoptosis was induced by exposure of cells to hypoxia/SD in a sealed GENbox hypoxic chamber. Effects of LPA were investigated in the absence and presence of inhibitors that target either G(i)proteins, the mitogen activated protein kinases ERK1/2, or phosphoinositide 3-kinase (PI3K). The data obtained showed that hypoxia/SD-induced apoptosis was significantly attenuated by LPA through Gi-coupled LPA(1) receptors linked to the downstream ERK1/2 and PI3K/Akt signaling pathways that function in parallel. Additional studies have demonstrated that hypoxia/SD-induced activation of mitochondrial dysfunction was virtually abolished by LPA treatment and that inhibition of the LPA(1) receptor, Gi proteins, the PI3K/Akt pathway, or ERKs effectively reversed this protective action of LPA. Taken together, our findings indicate that LPA is a novel, potent survival factor for MSCs and this may prove to be of considerable therapeutic significance in terms of exploiting MSC-based therapy in the infracted myocardium.
Collapse
Affiliation(s)
- Jinghai Chen
- Research Center for Cardiovascular Regenerative Medicine, The Ministry of Health of China, Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yea K, Kim J, Lim S, Park HS, Park KS, Suh PG, Ryu SH. Lysophosphatidic acid regulates blood glucose by stimulating myotube and adipocyte glucose uptake. J Mol Med (Berl) 2007; 86:211-20. [PMID: 17924084 DOI: 10.1007/s00109-007-0269-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 09/06/2007] [Accepted: 09/13/2007] [Indexed: 01/06/2023]
Abstract
Lysophosphatidic acid (LPA) is known to have diverse cellular effects, but although LPA is present in many biological fluids, including blood, its effects on glucose metabolism have not been elucidated. In this study, we investigated whether LPA stimulation is related to glucose regulation. LPA was found to enhance glucose uptake in a dose-dependent manner both in L6 GLUT4myc myotubes and 3T3-L1 adipocytes by triggering GLUT4 translocation to the plasma membrane. Moreover, the effect of LPA on glucose uptake was completely inhibited by pretreating both cells with LPA receptor antagonist Ki16425 and Gi inhibitor pertussis toxin. In addition, LPA increased the phosphorylation of AKT-1 with no effects on IRS-1, and LPA-induced glucose uptake was abrogated by pretreatment with the PI 3-kinase inhibitor LY294002. When low concentration of insulin and LPA were treated simultaneously, an additive effect on glucose uptake was observed in both cell types. In line with its cellular functions, LPA significantly lowered blood glucose levels in normal mice but did not affect insulin secretion. LPA also had a glucose-lowering effect in streptozotocin-treated type 1 diabetic mice. In combination, these results suggest that LPA is involved in the regulation of glucose homeostasis in muscle and adipose tissues.
Collapse
Affiliation(s)
- Kyungmoo Yea
- Division of Molecular and Life Science, Pohang University of Science and Technology, San 31 Hyojadong, Pohang, 790-784, South Korea
| | | | | | | | | | | | | |
Collapse
|
42
|
Watanabe K, Bianco C, Strizzi L, Hamada S, Mancino M, Bailly V, Mo W, Wen D, Miatkowski K, Gonzales M, Sanicola M, Seno M, Salomon DS. Growth factor induction of Cripto-1 shedding by glycosylphosphatidylinositol-phospholipase D and enhancement of endothelial cell migration. J Biol Chem 2007; 282:31643-55. [PMID: 17720976 DOI: 10.1074/jbc.m702713200] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cripto-1 (CR-1) is a glycosylphosphatidylinositol (GPI)-anchored membrane glycoprotein that has been shown to play an important role in embryogenesis and cellular transformation. CR-1 is reported to function as a membrane-bound co-receptor and as a soluble ligand. Although a number of studies implicate the role of CR-1 as a soluble ligand in tumor progression, it is unclear how transition from the membrane-bound to the soluble form is physiologically regulated and whether differences in biological activity exist between these forms. Here, we demonstrate that CR-1 protein is secreted from tumor cells into the conditioned medium after treatment with serum, epidermal growth factor, or lysophosphatidic acid, and this soluble form of CR-1 exhibits the ability to promote endothelial cell migration as a paracrine chemoattractant. On the other hand, membrane-bound CR-1 can stimulate endothelial cell sprouting through direct cell-cell interaction. Shedding of CR-1 occurs at the GPI-anchorage site by the activity of GPI-phospholipase D (GPI-PLD), because CR-1 shedding was suppressed by siRNA knockdown of GPI-PLD and enhanced by overexpression of GPI-PLD. These findings describe a novel molecular mechanism of CR-1 shedding, which may contribute to endothelial cell migration and possibly tumor angiogenesis.
Collapse
Affiliation(s)
- Kazuhide Watanabe
- Tumor Growth Factor Section, Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhao Z, Xiao Y, Elson P, Tan H, Plummer SJ, Berk M, Aung PP, Lavery IC, Achkar JP, Li L, Casey G, Xu Y. Plasma lysophosphatidylcholine levels: potential biomarkers for colorectal cancer. J Clin Oncol 2007; 25:2696-701. [PMID: 17602074 DOI: 10.1200/jco.2006.08.5571] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE Plasma levels of lysophospholipids were evaluated as potential biomarkers for colorectal cancer (CRC), where a highly reliable and minimally invasive blood test is lacking. PATIENTS AND METHODS Patients with CRC (n = 133) and control subjects (n = 125) were recruited through the Cleveland Clinic. Preoperative plasma samples were analyzed for lysophospholipid levels using liquid chromatography mass spectrometry in a blinded fashion. Participants were randomly divided in a 2:1 ratio into a "training set" (TS) and a "validation set" (VS). Logistic regression models were used in the TS to identify markers that best discriminated between CRC and controls. A cutoff point for the final discriminating model was developed using the receiver operating characteristic curve to achieve 95% specificity. All analyses were then independently validated in the VS. RESULTS Plasma levels of several lysophosphatidylcholines (LPCs), including 18:1- and 18:2-LPC, were significantly decreased in CRC patients compared with controls (P < .001). A model based on total saturated LPC and the difference between the proportional amounts of 18:2-LPC and 18:1-LPC in the unsaturated LPC fraction was derived from the TS. This model achieved a sensitivity and specificity of 82% and 93%, respectively, in the VS. Overall, 118 (94%) of 125 control subjects and 113 (85%) of 133 CRC cases were correctly identified, including eight (89%) of nine CRC cases with stage T1 disease. CONCLUSION Percentage of 18:1-LPC or 18:2-LPC plasma levels compared with total saturated LPC levels, either individually or in combination, may represent potential biomarkers for CRC.
Collapse
Affiliation(s)
- Zhenwen Zhao
- Department of Cancer Biology, Lerner Research Institute, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mori K, Kitayama J, Aoki J, Kishi Y, Shida D, Yamashita H, Arai H, Nagawa H. Submucosal connective tissue-type mast cells contribute to the production of lysophosphatidic acid (LPA) in the gastrointestinal tract through the secretion of autotaxin (ATX)/lysophospholipase D (lysoPLD). Virchows Arch 2007; 451:47-56. [PMID: 17554559 DOI: 10.1007/s00428-007-0425-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Revised: 04/11/2007] [Accepted: 04/25/2007] [Indexed: 01/21/2023]
Abstract
Lysophosphatidic acid (LPA) is involved in a broad spectrum of biological activities, including wound healing and cancer metastasis. Autotaxin (ATX), originally isolated from a melanoma supernatant as a tumor cell motility-stimulating factor, has been shown to be molecularly identical to lysophospholipase D (lysoPLD), which is the main enzyme in the production of LPA. Although ATX/lysoPLD is known to be widely expressed in normal human tissues, the exact distribution of ATX-producing cells has not been fully investigated. In this study, we evaluated ATX/lysoPLD expression by immunohistochemical staining using a rat anti-ATX mAb in the human gastrointestinal tract and found that submucosal mast cells (MC) highly expressed this enzyme. This was confirmed by immunofluorescent double staining using mAbs to tryptase and chymase. Then, we isolated MC from human gastric tissue by an immunomagnetic method using CD117-microbeads and showed that a subpopulation of CD203c-positive MC showed positive staining for intracellular ATX/lysoPLD on flowcytometry. This was confirmed by Western blotting of the isolated cells. Moreover, a significant level of ATX/lysoPLD release could be detected in the culture supernatants of human MC by Western blot analysis. Our data suggest that submucosal MC play significant roles in various aspects of pathophysiology in the gastrointestinal tract by locally providing bioactive LPA through the production of ATX/lysoPLD.
Collapse
Affiliation(s)
- Ken Mori
- Department of Surgical Oncology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Tokumura A, Kume T, Fukuzawa K, Tahara M, Tasaka K, Aoki J, Arai H, Yasuda K, Kanzaki H. Peritoneal fluids from patients with certain gynecologic tumor contain elevated levels of bioactive lysophospholipase D activity. Life Sci 2007; 80:1641-9. [PMID: 17367815 DOI: 10.1016/j.lfs.2006.12.041] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Revised: 11/13/2006] [Accepted: 12/12/2006] [Indexed: 11/28/2022]
Abstract
Levels of lysophosphatidic acid (LPA), an important phospholipid mediator, in serum and ascitic fluid from ovarian cancer patients were shown to be higher than those from healthy women and from patients with other type of cancer, respectively. Although LPA in human serum seems mainly to be generated by lysophospholipase D (lysoPLD), the source and pathway for LPA in the ascitic fluid remain still obscure. In this study, we examined whether lysoPLD activity producing bioactive LPA in human peritoneal fluid was significantly elevated under pathological statuses. Lysophospholipase D activity in human peritoneal fluids was measured by quantifying choline released from exogenous lysophosphatidylcholine on their incubation at 37 degrees C. We also compared the activity of lysoPLD in sera from patients with different gynecologic diseases. We found relatively high lysoPLD activity in peritoneal fluids from patients with ovarian cancer, dermoid cyst or mucinous cystadenoma, whereas there were no significant differences in the serum lysoPLD activity among clinical groups and healthy subjects. The lysoPLD in the peritoneal fluid was found to have similar substrate specificity and metal ion requirement to those of serum lysoPLD, that has been identified as autotaxin, a tumor cell-motility stimulating protein. Our results suggest that increased lysoPLD activity in peritoneal fluid from patients with certain gynecologic tumors might be relevant to its potential of tumor progression.
Collapse
Affiliation(s)
- Akira Tokumura
- Department of Health Chemistry, Institute of Health Biosciences, University of Tokushima Graduate School, 1-78-1 Shomachi, Tokushima, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Koh E, Clair T, Hermansen R, Bandle RW, Schiffmann E, Roberts DD, Stracke ML. Sphingosine-1-phosphate initiates rapid retraction of pseudopodia by localized RhoA activation. Cell Signal 2007; 19:1328-38. [PMID: 17307336 DOI: 10.1016/j.cellsig.2007.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Revised: 01/09/2007] [Accepted: 01/09/2007] [Indexed: 01/17/2023]
Abstract
Lysophosphatidic acid (LPA) stimulates sphingosine-1-phosphate (S1P)-sensitive motility in NIH3T3 clone7 cells. S1P inhibits motility only when added to the bottom well of the Boyden chamber, suggesting that pseudopodia can respond to their microenvironment. In order to study and localize this effect, we utilized a Transwell insert system to isolate pseudopodia. LPA stimulates protrusion of pseudopodia that are enriched in RhoA compared to cell bodies. Removal of LPA results in slow retraction with loss of vinculin-rich adhesion complexes and prolonged activation of RhoA. However, RhoA, ROCK and mDia are not required for this process. In contrast, rapid retraction, induced by adding S1P to the bottom well, is associated with a quick spike of activated RhoA and coalescence of adhesion complexes that colocalize with the ends of stress fibers. S1P-induced retraction requires RhoA and ROCK but is only delayed by inhibition of mDia. These data indicate that pseudopodia sense and integrate signals initiated by localized bioactive lipids, affecting both cellular polarity and their own function in motility.
Collapse
Affiliation(s)
- Eunjin Koh
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 2A33, Bethesda, MD 20892-1500, United States
| | | | | | | | | | | | | |
Collapse
|
47
|
Koh E, Bandle R, Clair T, Roberts DD, Stracke ML. Trichostatin A and 5-aza-2'-deoxycytidine switch S1P from an inhibitor to a stimulator of motility through epigenetic regulation of S1P receptors. Cancer Lett 2006; 250:53-62. [PMID: 17189669 DOI: 10.1016/j.canlet.2006.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 09/11/2006] [Accepted: 09/22/2006] [Indexed: 12/31/2022]
Abstract
The histone deacetylase inhibitor, trichostatin A (TSA), and the DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (Aza-dC), induced epigenetic regulation of sphingosine-1-phosphate (S1P) receptors in human melanoma cells, switching S1P from motility inhibitor to stimulator. Quantitative PCR revealed increased expression of S1P(1) and S1P(3), associated with S1P-induced chemotaxis, and decreased expression of S1P(2), associated with motility inhibition. Expression of lysophosphatidic acid (LPA) receptors was less affected. The TSA effect was reversible suggesting no mutational change, and Aza-dC treatment resulted in demethylation of a putative S1P(1) promoter. S1P receptors, therefore, appear to be susceptible to epigenetic regulation, accompanied by altered cellular functionality.
Collapse
Affiliation(s)
- Eunjin Koh
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1500, USA.
| | | | | | | | | |
Collapse
|
48
|
|
49
|
Sengupta S, Kim KS, Berk MP, Oates R, Escobar P, Belinson J, Li W, Lindner DJ, Williams B, Xu Y. Lysophosphatidic acid downregulates tissue inhibitor of metalloproteinases, which are negatively involved in lysophosphatidic acid-induced cell invasion. Oncogene 2006; 26:2894-901. [PMID: 17130843 DOI: 10.1038/sj.onc.1210093] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ovarian cancer is a highly metastatic disease. Lysophosphatidic acid (LPA) levels are elevated in ascites from ovarian cancer patients, but its potential role in ovarian cancer metastasis has just begun to be revealed. In this work, we show that LPA stimulates invasion of primary ovarian cancer cells, but not ovarian epithelial or borderline ovarian tumor cells, although these benign cells indeed respond to LPA in cell migration. We have found that LPA downregulates tissue inhibitor of metalloproteinases (TIMPs). TIMP2 and TIMP3 play functional role in LPA-induced invasion as negative regulators. G(i) protein, phosphatidylinositol-3 kinase (PI3K), p38 mitogen-activated protein kinase (MAPK), cytosolic phospholipase A(2) and urokinase type plasminogen activator (uPA) are required for LPA-induced cells invasion. TIMP3 may affect two independent downstream targets, vascular endothelial growth factor receptor and p38 MAPK. In vivo, LPA stimulates tumor metastasis in an orthotopic ovarian tumor model, which can be inhibited by a PI3K inhibitor, LY294002. In summary, LPA is likely a key component for promoting ovarian metastasis in vivo. LPA downregulates TIMP3, which may have targets other than metalloproteinases. Our in vivo metastasis mouse model is useful for studying the efficacy of therapeutic regimes of ovarian cancer.
Collapse
Affiliation(s)
- S Sengupta
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kim KS, Sengupta S, Berk M, Kwak YG, Escobar PF, Belinson J, Mok SC, Xu Y. Hypoxia Enhances Lysophosphatidic Acid Responsiveness in Ovarian Cancer Cells and Lysophosphatidic Acid Induces Ovarian Tumor MetastasisIn vivo. Cancer Res 2006; 66:7983-90. [PMID: 16912173 DOI: 10.1158/0008-5472.can-05-4381] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lysophosphatidic acid (LPA) is elevated in ascites of ovarian cancer patients and stimulates growth and other activities of ovarian cancer cells in vitro. Tissue hypoxia is a critical factor for tumor aggressiveness and metastasis in cancers. We tested whether the ascites of ovarian cancer is hypoxic and whether hypoxia influences the effects of LPA on ovarian cancer cells. We found that ovarian ascitic fluids were hypoxic in vivo. Enhanced cellular responsiveness to LPA, including migration and/or invasion of ovarian cancer cells, was observed under hypoxic conditions. This enhancement could be completely blocked by geldanamycin or a small interfering RNA targeting hypoxia-inducible factor 1 alpha (HIF1 alpha). LPA-induced cell migration required cytosolic phospholipase A(2) (cPLA(2)) and LPA stimulates cPLA(2) phosphorylation in a HIF1 alpha-dependent manner under hypoxia conditions. Furthermore, we show for the first time that exogenous LPA enhances tumor metastasis in an orthotopic ovarian cancer model and HIF alpha expression in tumors. 17-Dimethylaminoethylamino-17-demethoxygeldanamycin (an inhibitor of the heat shock protein 90) effectively blocked LPA-induced tumor metastasis in vivo. Together, our data indicate that hypoxic conditions are likely to be pathologically important for ovarian cancer development. HIF1 alpha plays a critical role in enhancing and/or sensitizing the role of LPA on cell migration and invasion under hypoxic conditions, where cPLA(2) is required for LPA-induced cell migration.
Collapse
Affiliation(s)
- Kwan-Sik Kim
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | |
Collapse
|