1
|
Fu F, Yu Y, Zou B, Long Y, Wu L, Yin J, Zhou Q. Role of actin-binding proteins in prostate cancer. Front Cell Dev Biol 2024; 12:1430386. [PMID: 39055653 PMCID: PMC11269120 DOI: 10.3389/fcell.2024.1430386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
The molecular mechanisms driving the onset and metastasis of prostate cancer remain poorly understood. Actin, under the control of actin-binding proteins (ABPs), plays a crucial role in shaping the cellular cytoskeleton, which in turn supports the morphological alterations in normal cells, as well as the invasive spread of tumor cells. Previous research indicates that ABPs of various types serve distinct functions, and any disruptions in their activities could predispose individuals to prostate cancer. These ABPs are intricately implicated in the initiation and advancement of prostate cancer through a complex array of intracellular processes, such as severing, linking, nucleating, inducing branching, assembling, facilitating actin filament elongation, terminating elongation, and promoting actin molecule aggregation. As such, this review synthesizes existing literature on several ABPs linked to prostate cancer, including cofilin, filamin A, and fascin, with the aim of shedding light on the molecular mechanisms through which ABPs influence prostate cancer development and identifying potential therapeutic targets. Ultimately, this comprehensive examination seeks to contribute to the understanding and management of prostate diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qing Zhou
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
2
|
Gui J, Zhou H, Wan H, Yang D, Liu Q, Zhu L, Mi Y. The Role of Vasodilator-stimulated Phosphoproteins in the Development of Malignant Tumors. Curr Cancer Drug Targets 2024; 24:477-489. [PMID: 37962042 PMCID: PMC11092557 DOI: 10.2174/0115680096262439231023110106] [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] [Received: 05/15/2023] [Revised: 08/06/2023] [Accepted: 09/06/2023] [Indexed: 11/15/2023]
Abstract
Vasodilator-stimulated phosphoprotein (VASP) is an actin-binding protein that includes three structural domains: Enabled/VASP homolog1 (EVH1), EVH2, and proline-rich (PRR). VASP plays an important role in various cellular behaviors related to cytoskeletal regulation. More importantly, VASP plays a key role in the progression of several malignant tumors and is associated with malignant cell proliferation, invasion, and metastasis. Here, we have summarized current studies on the impact of VASP on the development of several malignant tumors and their mechanisms. This study provides a new theoretical basis for clinical molecular diagnosis and molecular targeted therapy.
Collapse
Affiliation(s)
- Jiandong Gui
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Hangsheng Zhou
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Hongyuan Wan
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Dongjie Yang
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Qing Liu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
- Huadong Sanatorium, 67 Dajishan, Wuxi 214122, Jiangsu Province, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| |
Collapse
|
3
|
Wang C, Zhao X, Zhao L, Wang Y, Jia Y, Zhang X, Ma W. PKCζ phosphorylates VASP to mediate chemotaxis in breast cancer cells. Exp Cell Res 2023; 433:113823. [PMID: 37890607 DOI: 10.1016/j.yexcr.2023.113823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/17/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023]
Abstract
Breast carcinoma (BC) is one of the most common malignant cancers in females, and metastasis remains the leading cause of death in these patients. Chemotaxis plays an important role in cancer cell metastasis and the mechanism of breast cancer chemotaxis has become a central issue in contemporary research. PKCζ, a member of the atypical PKC family, has been reported to be an essential component of the EGF-stimulated chemotactic signaling pathway. However, the molecular mechanism through which PKCζ regulates chemotaxis remains unclear. Here, we used a proteomic approach to identify PKCζ-interacting proteins in breast cancer cells and identified VASP as a potential binding partner. Intriguingly, stimulation with EGF enhanced this interaction and induced the translocalization of PKCζ and VASP to the cell membrane. Further experiments showed that PKCζ catalyzes the phosphorylation of VASP at Ser157, which is critical for the biological function of VASP in regulating chemotaxis and actin polymerization in breast cancer cells. Furthermore, in PKCζ knockdown BC cells, the enrichment of VASP at the leading edge was reduced, and its interaction with profilin1 was attenuated, thereby reducing the chemotaxis and overall motility of breast cancer cells after EGF treatment. In functional assays, PKCζ promoted chemotaxis and motility of BC cells through VASP. Our findings demonstrate that PKCζ, a new kinase of VASP, plays an important role in promoting breast cancer metastasis and provides a theoretical basis for expanding new approaches to tumor biotherapy.
Collapse
Affiliation(s)
- Chunqing Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine China
| | - Xiaoqing Zhao
- Department of Clinical Laboratory Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong China
| | - Liqing Zhao
- Department of Pediatrics, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277000, China
| | - Yunqiu Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine China
| | - Yan Jia
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Xiaofang Zhang
- Department of Clinical Laboratory Medicine, Tianjin Medical University General Hospital, Tianjin, China.
| | - Wanshan Ma
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine China.
| |
Collapse
|
4
|
Pan W, Tian Y, Zheng Q, Yang Z, Qiang Y, Zhang Z, Zhang N, Xiong J, Zhu X, Wei L, Li F. Oncogenic BRAF noncanonically promotes tumor metastasis by mediating VASP phosphorylation and filopodia formation. Oncogene 2023; 42:3194-3205. [PMID: 37689827 DOI: 10.1038/s41388-023-02829-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023]
Abstract
BRAF is frequently mutated in various cancer types and contributes to tumorigenesis and metastasis. As an important switch in RAS signaling pathway, BRAF typically enables the activation of MEK and ERK, and its mutation significantly promotes metastasis. However, whether BRAF could stimulate metastasis via a distinct manner is still unknown. Herein, we found that a portion of the BRAF protein localized at the plasma membrane and that the BRAFV600E mutation led to abundant formation of filopodia, which is a hallmark of invasive cancer cells. Mechanistically, BRAF physically interacts with the pseudopod formation-related protein Vasodilator-stimulated phosphoprotein (VASP), and BRAF specifically catalyzes VASP phosphorylation at Ser157. VASP depletion or disruption of Ser157 phosphorylation preferentially reduced the motility, invasion and metastasis of tumor cells harboring oncogenic BRAF or KRAS. Moreover, in clinical cancer tissues, BRAFV600E was positively correlated with the extent of invasion, and tissues with BRAFV600E expression exhibited elevated levels of VASP Ser157 phosphorylation. Our study therefor reveals a noncanonical mechanism by which oncogenic BRAF or KRAS promotes metastasis, suggests that VASP Ser157 phosphorylation might serve as a valuable therapeutic target in BRAF or KRAS mutant cancers.
Collapse
Affiliation(s)
- Wenting Pan
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yihao Tian
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qian Zheng
- Department of Medical Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zelin Yang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yulong Qiang
- Department of Medical Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zun Zhang
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Nan Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jie Xiong
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| | - Xin Zhu
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Lei Wei
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| | - Feng Li
- Department of Medical Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
- Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan, China.
| |
Collapse
|
5
|
Wang S, Chen J, Guo XZ. KAI1/CD82 gene and autotaxin-lysophosphatidic acid axis in gastrointestinal cancers. World J Gastrointest Oncol 2022; 14:1388-1405. [PMID: 36160748 PMCID: PMC9412925 DOI: 10.4251/wjgo.v14.i8.1388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/06/2022] [Accepted: 07/22/2022] [Indexed: 02/05/2023] Open
Abstract
The KAI1/CD82 gene inhibits the metastasis of most tumors and is remarkably correlated with tumor invasion and prognosis. Cell metabolism dysregulation is an important cause of tumor occurrence, development, and metastasis. As one of the important characteristics of tumors, cell metabolism dysregulation is attracting increasing research attention. Phospholipids are an indispensable substance in the metabolism in various tumor cells. Phospholipid metabolites have become important cell signaling molecules. The pathological role of lysophosphatidic acid (LPA) in tumors was identified in the early 1990s. Currently, LPA inhibitors have entered clinical trials but are not yet used in clinical treatment. Autotaxin (ATX) has lysophospholipase D (lysoPLD) activity and can regulate LPA levels in vivo. The LPA receptor family and ATX/lysoPLD are abnormally expressed in various gastrointestinal tumors. According to our recent pre-experimental results, KAI1/CD82 might inhibit the migration and metastasis of cancer cells by regulating the ATX-LPA axis. However, no relevant research has been reported. Clarifying the mechanism of ATX-LPA in the inhibition of cancer metastasis by KAI1/CD82 will provide an important theoretical basis for targeted cancer therapy. In this paper, the molecular compositions of the KAI1/CD82 gene and the ATX-LPA axis, their physiological functions in tumors, and their roles in gastrointestinal cancers and target therapy are reviewed.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang 110840, Liaoning Province, China
| | - Jiang Chen
- Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang 110840, Liaoning Province, China
| | - Xiao-Zhong Guo
- Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang 110840, Liaoning Province, China
| |
Collapse
|
6
|
Tarvainen I, Nunn RC, Tuominen RK, Jäntti MH, Talman V. Protein Kinase A-Mediated Effects of Protein Kinase C Partial Agonist 5-(Hydroxymethyl)Isophthalate 1a3 in Colorectal Cancer Cells. J Pharmacol Exp Ther 2022; 380:54-62. [PMID: 34697230 DOI: 10.1124/jpet.121.000848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/14/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer is the third most commonly occurring cancer in men and the second in women. The global burden of colorectal cancer is projected to increase to over 2 million new cases with over 1 million deaths within the next 10 years, and there is a great need for new compounds with novel mechanisms of action. Our group has developed protein kinase C (PKC)-modulating isophthalic acid derivatives that induce cytotoxicity toward human cervical and prostate cancer cell lines. In this study, we investigated the effects of 5-(hydroxymethyl)isophthalate 1a3 (HMI-1a3) on colorectal cancer cell lines (Caco-2, Colo205, and HT29). HMI-1a3 inhibited cell proliferation, decreased cell viability, and induced an apoptotic response in all studied cell lines. These effects, however, were independent of PKC. Using serine/threonine kinome profiling and pharmacological kinase inhibitors, we identified activation of the cAMP/PKA pathway as a new mechanism of action for HMI-1a3-induced anticancer activity in colorectal cancer cell lines. Our current results strengthen the hypothesis for HMI-1a3 as a potential anticancer agent against various malignancies. SIGNIFICANCE STATEMENT: Colorectal cancer (CRC) is a common solid organ malignancy. This study demonstrates that the protein kinase C (PKC)-C1 domain-targeted isophthalatic acid derivative 5-(hydroxymethyl)isophthalate 1a3 (HMI-1a3) has anticancer activity on CRC cell lines independently of PKC. We identified PKA activation as a mechanism of HMI-1a3-induced anticancer effects. The results reveal a new anticancer mechanism of action for the partial PKC agonist HMI-1a3 and thus provide new insights for the development of PKC and PKA modulators for cancer therapy.
Collapse
Affiliation(s)
- Ilari Tarvainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| | - Rebecca C Nunn
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| | - Raimo K Tuominen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| | - Maria H Jäntti
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| |
Collapse
|
7
|
Lysophosphatidic Acid Signaling in Cancer Cells: What Makes LPA So Special? Cells 2021; 10:cells10082059. [PMID: 34440828 PMCID: PMC8394178 DOI: 10.3390/cells10082059] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/13/2022] Open
Abstract
Lysophosphatidic acid (LPA) refers to a family of simple phospholipids that act as ligands for G protein-coupled receptors. While LPA exerts effects throughout the body in normal physiological circumstances, its pathological role in cancer is of great interest from a therapeutic viewpoint. The numerous LPA receptors (LPARs) are coupled to a variety of G proteins, and more than one LPAR is typically expressed on any given cell. While the individual receptors signal through conventional GPCR pathways, LPA is particularly efficacious in stimulating cancer cell proliferation and migration. This review addresses the mechanistic aspects underlying these pro-tumorigenic effects. We provide examples of LPA signaling responses in various types of cancers, with an emphasis on those where roles have been identified for specific LPARs. While providing an overview of LPAR signaling, these examples also reveal gaps in our knowledge regarding the mechanisms of LPA action at the receptor level. The current understanding of the LPAR structure and the roles of LPAR interactions with other receptors are discussed. Overall, LPARs provide insight into the potential molecular mechanisms that underlie the ability of individual GPCRs (or combinations of GPCRs) to elicit a unique spectrum of responses from their agonist ligands. Further knowledge of these mechanisms will inform drug discovery, since GPCRs are promising therapeutic targets for cancer.
Collapse
|
8
|
Xie Y, Wang Y, Xiang W, Wang Q, Cao Y. Molecular Mechanisms of the Action of Myricetin in Cancer. Mini Rev Med Chem 2020; 20:123-133. [PMID: 31648635 DOI: 10.2174/1389557519666191018112756] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/31/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023]
Abstract
Natural compounds, such as paclitaxel and camptothecin, have great effects on the treatment of tumors. Such natural chemicals often achieve anti-tumor effects through a variety of mechanisms. Therefore, it is of great significance to conduct further studies on the anticancer mechanism of natural anticancer agents to lay a solid foundation for the development of new drugs. Myricetin, originally isolated from Myrica nagi, is a natural pigment of flavonoids that can inhibit the growth of cancer cells (such as liver cancer, rectal cancer, skin cancer and lung cancer, etc.). It can regulate many intracellular activities (such as anti-inflammatory and blood lipids regulation) and can even be bacteriostatic. The purpose of this paper is to outline the molecular pathways of the anticancer effects of myricetin, including the effect on cancer cell death, proliferation, angiogenesis, metastasis and cell signaling pathway.
Collapse
Affiliation(s)
- Yutao Xie
- Department of Pharmacy, Nanchong Center Hospital, The Second Clinical Medical College, North Sichuan Medical College (University), Nanchong, 637000, Sichuan, China
| | - Yunlong Wang
- Department of Pharmacy, Nanchong Center Hospital, The Second Clinical Medical College, North Sichuan Medical College (University), Nanchong, 637000, Sichuan, China
| | - Wei Xiang
- Department of Pharmacy, Nanchong Center Hospital, The Second Clinical Medical College, North Sichuan Medical College (University), Nanchong, 637000, Sichuan, China
| | - Qiaoying Wang
- Department of Cardiothoracic Surgery, Nanchong Center Hospital, The Second Clinical Medical College, North Sichuan Medical College (University), Nanchong, 637000, Sichuan, China
| | - Yajun Cao
- Department of Pharmacy, Nanchong Center Hospital, The Second Clinical Medical College, North Sichuan Medical College (University), Nanchong, 637000, Sichuan, China
| |
Collapse
|
9
|
Mir SS, Bhat HF, Bhat ZF. Dynamic actin remodeling in response to lysophosphatidic acid. J Biomol Struct Dyn 2020; 38:5253-5265. [PMID: 31920158 DOI: 10.1080/07391102.2019.1696230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Lysophosphatidic acid (LPA) is a multifunctional regulator of actin cytoskeleton that exerts a dramatic impact on the actin cytoskeleton to build a platform for diverse cellular processes including growth cone guidance, neurite retraction and cell motility. It has been implicated in the formation and dissociation of complexes between actin and actin binding proteins, supporting its role in actin remodeling. Several studies point towards its ability to facilitate formation of special cellular structures including focal adhesions and actin stress fibres by phosphoregulation of several actin associated proteins and their multiple regulatory kinases and phosphatases. In addition, multiple levels of crosstalk among the signaling cascades activated by LPA, affect actin cytoskeleton-mediated cell migration and chemotaxis which in turn play a crucial role in cancer metastasis. In the current review, we have attempted to highlight the role of LPA as an actin modulator which functions by controlling activities of specific cellular proteins that underlie mechanisms employed in cytoskeletal and pathophysiological events within the cell. Further studies on the actin affecting/remodeling activity of LPA in different cell types will no doubt throw up many surprises essential to gain a full understanding of its contribution in physiological processes as well as in diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saima S Mir
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu And Kashmir, India.,Division of Animal Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, Jammu And Kashmir, India
| | - Hina F Bhat
- Division of Animal Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, Jammu And Kashmir, India
| | - Zuhaib F Bhat
- Department of Wine, Food & Molecular Biosciences, Lincoln University, Lincoln, New Zealand.,Division of Livestock Products and Technology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu (SKUAST-J), R.S. Pora, Jammu And Kashmir, India
| |
Collapse
|
10
|
Khong ZJ, Lai SK, Koh CG, Geifman-Shochat S, Li HY. A novel function of AAA-ATPase p97/VCP in the regulation of cell motility. Oncotarget 2020; 11:74-85. [PMID: 32002125 PMCID: PMC6967774 DOI: 10.18632/oncotarget.27419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/21/2019] [Indexed: 11/25/2022] Open
Abstract
High level of the multifunctional AAA-ATPase p97/VCP is often correlated to the development of cancer; however, the underlying mechanism is not understood completely. Here, we report a novel function of p97/VCP in actin regulation and cell motility. We found that loss of p97/VCP promotes stabilization of F-actin, which cannot be reversed by actin-destabilizing agent, Cytochalasin D. Live-cell imaging demonstrated reduced actin dynamics in p97/VCP-knockdown cells, leading to compromised cell motility. We further examined the underlying mechanism and found elevated RhoA protein levels along with increased phosphorylation of its downstream effectors, ROCK, LIMK, and MLC upon the knockdown of p97/VCP. Since p97/VCP is indispensable in the ubiquitination-dependent protein degradation pathway, we investigated if the loss of p97/VCP hinders the protein degradation of RhoA. Knockdown of p97/VCP resulted in a higher amount of ubiquitinated RhoA, suggesting p97/VCP involvement in the proteasome-dependent protein degradation pathway. Finally, we found that p97/VCP interacts with FBXL19, a molecular chaperone known to guide ubiquitinated RhoA for proteasomal degradation. Reduction of p97/VCP may result in the accumulation of RhoA which, in turn, enhances cytoplasmic F-actin formation. In summary, our study uncovered a novel function of p97/VCP in actin regulation and cell motility via the Rho-ROCK dependent pathway which provides fundamental insights into how p97/VCP is involved in cancer development.
Collapse
Affiliation(s)
- Zi-Jia Khong
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Soak-Kuan Lai
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Cheng-Gee Koh
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Susana Geifman-Shochat
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Hoi-Yeung Li
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| |
Collapse
|
11
|
Hypoxia Downregulates LPP3 and Promotes the Spatial Segregation of ATX and LPP1 During Cancer Cell Invasion. Cancers (Basel) 2019; 11:cancers11091403. [PMID: 31546971 PMCID: PMC6769543 DOI: 10.3390/cancers11091403] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/07/2019] [Accepted: 09/12/2019] [Indexed: 12/16/2022] Open
Abstract
Hypoxia is a common characteristic of advanced solid tumors and a potent driver of tumor invasion and metastasis. Recent evidence suggests the involvement of autotaxin (ATX) and lysophosphatidic acid receptors (LPARs) in cancer cell invasion promoted by the hypoxic tumor microenvironment; however, the transcriptional and/or spatiotemporal control of this process remain unexplored. Herein, we investigated whether hypoxia promotes cell invasion by affecting the main enzymes involved in its production (ATX) and degradation (lipid phosphate phosphatases, LPP1 and LPP3). We report that hypoxia not only modulates the expression levels of lysophosphatidic acid (LPA) regulatory enzymes but also induces their significant spatial segregation in a variety of cancers. While LPP3 expression was downregulated by hypoxia, ATX and LPP1 were asymmetrically redistributed to the leading edge and to the trailing edge, respectively. This was associated with the opposing roles of ATX and LPPs in cell invasion. The regulated expression and compartmentalization of these enzymes of opposing function can provide an effective way to control the generation of an LPA gradient that drives cellular invasion and migration in the hypoxic zones of tumors.
Collapse
|
12
|
Lin YC, Chen CC, Chen WM, Lu KY, Shen TL, Jou YC, Shen CH, Ohbayashi N, Kanaho Y, Huang YL, Lee H. LPA 1/3 signaling mediates tumor lymphangiogenesis through promoting CRT expression in prostate cancer. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1305-1315. [PMID: 30053596 DOI: 10.1016/j.bbalip.2018.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/13/2018] [Accepted: 07/20/2018] [Indexed: 12/15/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid growth factor which is present in high levels in serum and platelets. LPA binds to its specific G-protein-coupled receptors, including LPA1 to LPA6, thereby regulating various physiological functions, including cancer growth, angiogenesis, and lymphangiogenesis. Our previous study showed that LPA promotes the expression of the lymphangiogenic factor vascular endothelial growth factor (VEGF)-C in prostate cancer (PCa) cells. Interestingly, LPA has been shown to regulate the expression of calreticulin (CRT), a multifunctional chaperone protein, but the roles of CRT in PCa progression remain unclear. Here we investigated the involvement of CRT in LPA-mediated VEGF-C expression and lymphangiogenesis in PCa. Knockdown of CRT significantly reduced LPA-induced VEGF-C expression in PC-3 cells. Moreover, LPA promoted CRT expression through LPA receptors LPA1 and LPA3, reactive oxygen species (ROS) production, and phosphorylation of eukaryotic translation initiation factor 2α (eIF2α). Tumor-xenografted mouse experiments further showed that CRT knockdown suppressed tumor growth and lymphangiogenesis. Notably, clinical evidence indicated that the LPA-producing enzyme autotaxin (ATX) is related to CRT and that CRT level is highly associated with lymphatic vessel density and VEGF-C expression. Interestingly, the pharmacological antagonist of LPA receptors significantly reduced the lymphatic vessel density in tumor and lymph node metastasis in tumor-bearing nude mice. Together, our results demonstrated that CRT is critical in PCa progression through the mediation of LPA-induced VEGF-C expression, implying that targeting the LPA signaling axis is a potential therapeutic strategy for PCa.
Collapse
Affiliation(s)
- Yueh-Chien Lin
- Department of Life Sciences, National Taiwan University, Taipei 10617, Taiwan; Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Chien-Chin Chen
- Department of Pathology, Chia-Yi Christian Hospital, Chiayi 600, Taiwan; Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Wei-Min Chen
- Department of Life Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Kuan-Ying Lu
- Department of Life Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 10617, Taiwan
| | - Yeong-Chin Jou
- Department of Urology, Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Cheng-Huang Shen
- Department of Urology, Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Norihiko Ohbayashi
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yasunori Kanaho
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuan-Li Huang
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan.
| | - Hsinyu Lee
- Department of Life Sciences, National Taiwan University, Taipei 10617, Taiwan; Department of Electrical Engineering, National Taiwan University, Taipei 10617, Taiwan; Institute of Biomedical Electronic and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan; Center for Biotechnology, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
13
|
Tan HT, Chung MCM. Label-Free Quantitative Phosphoproteomics Reveals Regulation of Vasodilator-Stimulated Phosphoprotein upon Stathmin-1 Silencing in a Pair of Isogenic Colorectal Cancer Cell Lines. Proteomics 2018; 18:e1700242. [PMID: 29460479 DOI: 10.1002/pmic.201700242] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 02/10/2018] [Indexed: 02/06/2023]
Abstract
In this communication, we present the phosphoproteome changes in an isogenic pair of colorectal cancer cell lines, viz., the poorly metastatic HCT-116 and the highly metastatic derivative E1, upon stathmin-1 (STMN1) knockdown. The aim was to better understand how the alterations of the phosphoproteins in these cells are involved in cancer metastasis. After the phosphopeptides were enriched using the TiO2 HAMMOC approach, comparative proteomics analysis was carried out using sequential window acquisition of all theoretical mass spectra-MS. Following bioinformatics analysis using MarkerView and OneOmics platforms, we identified a list of regulated phosphoproteins that may play a potential role in signaling, maintenance of cytoskeletal structure, and focal adhesion. Among these phosphoproteins, was the actin cytoskeleton regulator protein, vasodilator-stimulated phosphoprotein (VASP), where its change in phosphorylation status was found to be concomitant with STMN1-associated roles in metastasis. We further showed that silencing of stathmin-1 altered the expression, subcellular localization and phosphorylation status of VASP, which suggested that it might be associated with remodeling of the cell cytoskeleton in colorectal cancer metastasis.
Collapse
Affiliation(s)
- Hwee Tong Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Maxey Ching Ming Chung
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
14
|
New cGMP analogues restrain proliferation and migration of melanoma cells. Oncotarget 2017; 9:5301-5320. [PMID: 29435180 PMCID: PMC5797051 DOI: 10.18632/oncotarget.23685] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022] Open
Abstract
Melanoma is one of the most aggressive cancers and displays high resistance to conventional chemotherapy underlining the need for new therapeutic strategies. The cGMP/PKG signaling pathway was detected in melanoma cells and shown to reduce migration, proliferation and to increase apoptosis in different cancer types. In this study, we evaluated the effects on cell viability, cell death, proliferation and migration of novel dimeric cGMP analogues in two melanoma cell lines (MNT1 and SkMel28). These new dimeric cGMP analogues, by activating PKG with limited effects on PKA, significantly reduced proliferation, migration and increased cell death. No decrease in cell viability was observed in non-tumor cells suggesting a tumor-specific effect. These effects observed in melanoma are possibly mediated by PKG2 activation based on the decreased toxic effects in tumor cell lines not expressing PKG2. Finally, PKG-associated phosphorylation of vasodilator-stimulated-phosphoprotein (VASP), linked to cell death, proliferation and migration was found increased and with a change of subcellular localization. Increased phosphorylation of RhoA induced by activation of PKG may also contribute to reduced migration ability of the SkMel28 melanoma cell line when treated with cGMP analogues. These findings suggest that the cGMP/PKG pathway can be envisaged as a therapeutic target of novel dimeric cGMP analogues for the treatment of melanoma.
Collapse
|
15
|
Döppler H, Bastea L, Borges S, Geiger X, Storz P. The phosphorylation status of VASP at serine 322 can be predictive for aggressiveness of invasive ductal carcinoma. Oncotarget 2016; 6:29740-52. [PMID: 26336132 PMCID: PMC4745759 DOI: 10.18632/oncotarget.4965] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 07/31/2015] [Indexed: 11/25/2022] Open
Abstract
Vasodilator-stimulated phosphoprotein (VASP) signaling is critical for dynamic actin reorganization processes that define the motile phenotype of cells. Here we show that VASP is generally highly expressed in normal breast tissue and breast cancer. We also show that the phosphorylation status of VASP at S322 can be predictive for breast cancer progression to an aggressive phenotype. Our data indicate that phosphorylation at S322 is gradually decreased from normal breast to DCIS, luminal/ER+, HER2+ and basal-like/TN phenotypes. Similarly, the expression levels of PKD2, the kinase that phosphorylates VASP at this site, are decreased in invasive ductal carcinoma samples of all three groups. Overall, the phosphorylation status of this residue may serve as an indicator of aggressiveness of breast tumors.
Collapse
Affiliation(s)
- Heike Döppler
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ligia Bastea
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Sahra Borges
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Peter Storz
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
16
|
Hwang YS, Lee J, Zhang X, Lindholm PF. Lysophosphatidic acid activates the RhoA and NF-κB through Akt/IκBα signaling and promotes prostate cancer invasion and progression by enhancing functional invadopodia formation. Tumour Biol 2015; 37:6775-85. [PMID: 26662305 DOI: 10.1007/s13277-015-4549-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/30/2015] [Indexed: 12/31/2022] Open
Abstract
We have demonstrated previously that increased RhoA and nuclear factor (NF)-κB activities are associated with increased PC-3 prostate cancer cell invasion and that lysophosphatidic acid (LPA) significantly increases cancer invasion through RhoA and NF-κB activation. In this study, we identified the intermediate signaling molecules and specialized cell structures which are activated by LPA, resulting in enhanced cellular invasion. LPA-induced Akt and IκBα signaling pathways were necessary for RhoA and NF-κB activation, and these LPA effects were abolished by RhoA inhibition. Mice injected with PC-3 cells expressing dominant-negative RhoA N19 developed significantly less tumor growth compared with those injected with control (pcDNA 3.1). In addition, LPA treatment increased functional invadopodia formation. Activation of RhoA and NF-κB through the Akt and IκBα signaling pathway was required for LPA-stimulated gelatin degradation activity. LPA administration increased tumor growth and osteolytic lesions in a mouse xenograft model. These results indicate that LPA promotes PC-3 cell invasion by increasing functional invadopodia formation via upregulating RhoA and NF-κB signaling which contributes to prostate cancer progression. Therefore, the LPA and RhoA-NF-κB signaling axis may represent key molecular targets to inhibit prostate cancer invasion and progression.
Collapse
Affiliation(s)
- Young Sun Hwang
- Department of Dental Hygiene, College of Health Science, Eulji University, Seongnam, Republic of Korea
| | - Jongsung Lee
- Department of Genetic Engineering, Sungkyunkwan University, 2066, Seobu-Ro, Jangan Gu, Suwon City, Gyunggi Do, 164-19, Republic of Korea
| | - Xianglan Zhang
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Republic of Korea.,Department of Pathology, Yanbian University Hospital, Yanji City, Jilin Province, China
| | - Paul F Lindholm
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
17
|
Abstract
The G12 family of heterotrimeric G proteins is defined by their α-subunits,
Gα12 and Gα13. These α-subunits
regulate cellular homeostasis, cell migration, and oncogenesis in a
context-specific manner primarily through their interactions with distinct
proteins partners that include diverse effector molecules and scaffold proteins.
With a focus on identifying any other novel regulatory protein(s) that can
directly interact with Gα13, we subjected Gα13
to tandem affinity purification-coupled mass spectrometric analysis. Our results
from such analysis indicate that Gα13 potently interacts with
mammalian Ric-8A. Our mass spectrometric analysis data also indicates that
Ric-8A, which was tandem affinity purified along with Gα13, is
phosphorylated at Ser-436, Thr-441, Thr-443 and Tyr-435. Using a serial deletion
approach, we have defined that the C-terminus of Gα13 containing
the guanine-ring interaction site is essential and sufficient for its
interaction with Ric-8A. Evaluation of Gα13-specific signaling
pathways in SKOV3 or HeyA8 ovarian cancer cell lines indicate that Ric-8A
potentiates Gα13-mediated activation of RhoA, Cdc42, and the
downstream p38MAPK. We also establish that the tyrosine phosphorylation of
Ric-8A, thus far unidentified, is potently stimulated by Gα13.
Our results also indicate that the stimulation of tyrosine-phosphorylation of
Ric-8A by Gα13 is partially sensitive to inhibitors of
Src-family of kinases, namely PP2 and SI. Furthermore, we demonstrate that
Gα13 promotes the translocation of Ric-8A to plasma membrane
and this translocation is attenuated by the Src-inhibitors, SI1 and PP2. Thus,
our results demonstrate for the first time that Gα13 stimulates
the tyrosine phosphorylation of Ric-8A and Gα13-mediated
tyrosine-phosphorylation plays a critical role in the translocation of Ric-8A to
plasma membrane.
Collapse
|
18
|
Guo L, He P, No YR, Yun CC. Krüppel-like factor 5 incorporates into the β-catenin/TCF complex in response to LPA in colon cancer cells. Cell Signal 2015; 27:961-8. [PMID: 25683913 DOI: 10.1016/j.cellsig.2015.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/26/2015] [Accepted: 02/07/2015] [Indexed: 11/24/2022]
Abstract
Lysophosphatidic acid (LPA) is a simple phospholipid with potent mitogenic effects on various cells including colon cancer cells. LPA stimulates proliferation of colon cancer cells by activation of β-catenin or Krüppel-like factor 5 (KLF5), but the functional relationship between these two transcription factors is not clear. Hence, we sought to investigate the mechanism of β-catenin activation by LPA and the role of KLF5 in the regulation of β-catenin by LPA. We found that LPA and Wnt3 additively activated the β-catenin/TCF (T cell factor) reporter activity in HCT116 cells. In addition to phosphorylating glycogen synthase kinase 3β (GSK-3β) at Ser9, LPA resulted in phosphorylation of β-catenin at Ser552 and Ser675. Mutation of Ser552 and Ser675 ablated LPA-induced β-catenin/TCF transcriptional activity. Knockdown of KLF5 significantly attenuated activation of β-catenin/TCF reporter activity by LPA but not by Wnt3. However, nuclear accumulation of β-catenin by LPA was not altered by knockdown of KLF5. β-catenin, TCF, and KLF5 were present in a 250-300kDa macro-complex, and their presence was enhanced by LPA. LPA simulated the interaction of β-catenin with TCF4, and depletion of KLF5 decreased β-catenin-TCF4 association and the transcriptional activity. In summary, LPA activates β-catenin by multiple pathways involving phosphorylation of GSK-3 and β-catenin, and enhancing β-catenin interaction with TCF4. KLF5 plays a critical role in β-catenin activation by increasing the β-catenin-TCF4 interaction.
Collapse
Affiliation(s)
- Leilei Guo
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yi Ran No
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - C Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
19
|
Ren A, Moon C, Zhang W, Sinha C, Yarlagadda S, Arora K, Wang X, Yue J, Parthasarathi K, Heil-Chapdelaine R, Tigyi G, Naren AP. Asymmetrical macromolecular complex formation of lysophosphatidic acid receptor 2 (LPA2) mediates gradient sensing in fibroblasts. J Biol Chem 2014; 289:35757-69. [PMID: 25542932 DOI: 10.1074/jbc.m114.595512] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chemotactic migration of fibroblasts toward growth factors relies on their capacity to sense minute extracellular gradients and respond to spatially confined receptor-mediated signals. Currently, mechanisms underlying the gradient sensing of fibroblasts remain poorly understood. Using single-particle tracking methodology, we determined that a lysophosphatidic acid (LPA) gradient induces a spatiotemporally restricted decrease in the mobility of LPA receptor 2 (LPA2) on chemotactic fibroblasts. The onset of decreased LPA2 mobility correlates to the spatial recruitment and coupling to LPA2-interacting proteins that anchor the complex to the cytoskeleton. These localized PDZ motif-mediated macromolecular complexes of LPA2 trigger a Ca(2+) puff gradient that governs gradient sensing and directional migration in response to LPA. Disruption of the PDZ motif-mediated assembly of the macromolecular complex of LPA2 disorganizes the gradient of Ca(2+) puffs, disrupts gradient sensing, and reduces the directional migration of fibroblasts toward LPA. Our findings illustrate that the asymmetric macromolecular complex formation of chemoattractant receptors mediates gradient sensing and provides a new mechanistic basis for models to describe gradient sensing of fibroblasts.
Collapse
Affiliation(s)
- Aixia Ren
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Changsuk Moon
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Weiqiang Zhang
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Chandrima Sinha
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Sunitha Yarlagadda
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Kavisha Arora
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Xusheng Wang
- the Department of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, and
| | - Junming Yue
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Kaushik Parthasarathi
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | | | - Gabor Tigyi
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Anjaparavanda P Naren
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229,
| |
Collapse
|
20
|
Ward JD, Ha JH, Jayaraman M, Dhanasekaran DN. LPA-mediated migration of ovarian cancer cells involves translocalization of Gαi2 to invadopodia and association with Src and β-pix. Cancer Lett 2014; 356:382-91. [PMID: 25451317 DOI: 10.1016/j.canlet.2014.09.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/12/2014] [Accepted: 09/13/2014] [Indexed: 12/23/2022]
Abstract
Lysophosphatidic acid (LPA) plays a critical role in the migration and invasion of ovarian cancer cells. However, the downstream spatiotemporal signaling events involving specific G protein(s) underlying this process are largely unknown. In this report, we demonstrate that LPA signaling causes the translocation of Gαi2 into the invadopodia leading to its interaction with the tyrosine kinase Src and the Rac/CDC42-specific guanine nucleotide exchange factor, β-pix. Our results establish that Gαi2 activates Rac1 through a p130Cas-dependent pathway in ovarian cancer cells. Moreover, our report reveals that knockdown of Gαi2 leads to loss of β-pix and active-Rac association in the invadopodia. We also show that knockdown of Gαi2 leads to the complete loss of translocation to p130Cas to focal adhesions. Finally, when Gαi2 is knocked down, this led to the total distribution of Src being shifted primarily from invadopodia and the leading edge of the cells to the perinuclear region, suggesting that Src is inactive in the absence of Gαi2. Overall, our report provides tantalizing evidence that Gαi2 is a critical signaling component of a large signaling complex in the invadopodia that if disrupted could serve as an excellent target for therapy in ovarian and potentially other cancers.
Collapse
Affiliation(s)
- Jeremy D Ward
- Stephenson Cancer Center, Department of Cell Biology, The University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA
| | - Ji Hee Ha
- Stephenson Cancer Center, Department of Cell Biology, The University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA
| | - Muralidharan Jayaraman
- Stephenson Cancer Center, Department of Cell Biology, The University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, Department of Cell Biology, The University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA.
| |
Collapse
|
21
|
Döppler H, Storz P. Regulation of VASP by phosphorylation: consequences for cell migration. Cell Adh Migr 2013; 7:482-6. [PMID: 24401601 DOI: 10.4161/cam.27351] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Phosphorylations control all aspects of vasodilator-stimulated phospho-protein (VASP) function. Mapped phosphorylation sites include Y39, S157, S239, T278, and S322, and multiple kinases have been shown to mediate their phosphorylation. Recently, Protein Kinase D1 (PKD1) as a direct kinase for S157 and S322 joined this group. While S157 phosphorylation generally seems to serve as a signal for membrane localization, phosphorylations at S322 or at S239 and T278 have opposite effects on F-actin accumulation. In migrating cells, S322 phosphorylation increases filopodia numbers and length, while S239/T278 phosphorylations decrease these and also disrupt formation of focal adhesions. Therefore, the kinases mediating these phosphorylations can be seen as switches needed to facilitate cell motility.
Collapse
Affiliation(s)
- Heike Döppler
- Department of Cancer Biology; Mayo Clinic Comprehensive Cancer Center; Mayo Clinic; Jacksonville, FL USA
| | - Peter Storz
- Department of Cancer Biology; Mayo Clinic Comprehensive Cancer Center; Mayo Clinic; Jacksonville, FL USA
| |
Collapse
|
22
|
Lysophosphatidic acid induces reactive oxygen species generation by activating protein kinase C in PC-3 human prostate cancer cells. Biochem Biophys Res Commun 2013; 440:564-9. [DOI: 10.1016/j.bbrc.2013.09.104] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 09/20/2013] [Indexed: 11/21/2022]
|
23
|
Ochiai S, Furuta D, Sugita K, Taniura H, Fujita N. GPR87 mediates lysophosphatidic acid-induced colony dispersal in A431 cells. Eur J Pharmacol 2013; 715:15-20. [PMID: 23831392 DOI: 10.1016/j.ejphar.2013.06.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 06/06/2013] [Accepted: 06/21/2013] [Indexed: 01/17/2023]
Abstract
We have previously reported that an orphan G protein-coupled receptor GPR87 was activated by lysophosphatidic acid (LPA) and that it induced an increase in the intracellular Ca(2+) levels in the CHO cells genetically engineered to express GPR87-Gα16 fusion protein. Because the Ca(2+) response was blocked by the LPA receptor antagonist Ki16425, GPR87 was suggested to be a putative LPA receptor. However, further studies are required to confirm whether GPR87 is an LPA receptor. A previous study showed that colonies of A431 cells treated with LPA showed rapid and synchronized dissociation. Because A431 cells have been shown to express GPR87, we used these cells to examine whether GPR87 acted as an LPA receptor. When A431 cells were treated with gpr87-specific siRNA, the expression of GPR87 was decreased and LPA-induced colony dispersal was significantly reduced. Treatment of the cells with lpa1 siRNA had an additive effect in decrease in the colony dispersal. Studies on the LPA-mediated signaling pathway in A431 cells indicated that transactivation of the epidermal growth factor receptor (EGFR) by LPA led to cell scattering. PD153035, an inhibitor of tyrosine-kinase of EGFR, and BB94, an inhibitor of metalloprotease which produces a ligand for EGFR, significantly prevented the LPA-induced scattering of A431 cells pretreated with lpa1 or gpr87-siRNA. These results strongly suggested that GPR87 acts as an LPA receptor and induces colony dispersal via the transactivation of EGFR in A431 cells.
Collapse
Affiliation(s)
- Shoichi Ochiai
- Laboratory of Pharmacoinformitcs, Graduate School of Ritsumeikan University, and School of Pharmacy, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
| | | | | | | | | |
Collapse
|
24
|
Härmä V, Knuuttila M, Virtanen J, Mirtti T, Kohonen P, Kovanen P, Happonen A, Kaewphan S, Ahonen I, Kallioniemi O, Grafström R, Lötjönen J, Nees M. Lysophosphatidic acid and sphingosine-1-phosphate promote morphogenesis and block invasion of prostate cancer cells in three-dimensional organotypic models. Oncogene 2012; 31:2075-89. [PMID: 21996742 PMCID: PMC3330266 DOI: 10.1038/onc.2011.396] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 06/26/2011] [Accepted: 07/28/2011] [Indexed: 12/12/2022]
Abstract
Normal prostate and some malignant prostate cancer (PrCa) cell lines undergo acinar differentiation and form spheroids in three-dimensional (3-D) organotypic culture. Acini formed by PC-3 and PC-3M, less pronounced also in other PrCa cell lines, spontaneously undergo an invasive switch, leading to the disintegration of epithelial structures and the basal lamina, and formation of invadopodia. This demonstrates the highly dynamic nature of epithelial plasticity, balancing epithelial-to-mesenchymal transition against metastable acinar differentiation. This study assessed the role of lipid metabolites on epithelial maturation. PC-3 cells completely failed to form acinar structures in delipidated serum. Adding back lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) rescued acinar morphogenesis and repressed invasion effectively. Blocking LPA receptor 1 (LPAR1) functions by siRNA (small interference RNA) or the specific LPAR1 inhibitor Ki16425 promoted invasion, while silencing of other G-protein-coupled receptors responsive to LPA or S1P mainly caused growth arrest or had no effects. The G-proteins Gα(12/13) and Gα(i) were identified as key mediators of LPA signalling via stimulation of RhoA and Rho kinases ROCK1 and 2, activating Rac1, while inhibition of adenylate cyclase and accumulation of cAMP may be secondary. Interfering with these pathways specifically impeded epithelial polarization in transformed cells. In contrast, blocking the same pathways in non-transformed, normal cells promoted differentiation. We conclude that LPA and LPAR1 effectively promote epithelial maturation and block invasion of PrCa cells in 3-D culture. The analysis of clinical transcriptome data confirmed reduced expression of LPAR1 in a subset of PrCa's. Our study demonstrates a metastasis-suppressor function for LPAR1 and Gα(12/13) signalling, regulating cell motility and invasion versus epithelial maturation.
Collapse
Affiliation(s)
- V Härmä
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
| | - M Knuuttila
- Biotechnology Centre, University of Turku, Turku, Finland
| | - J Virtanen
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Biotechnology Centre, University of Turku, Turku, Finland
| | - T Mirtti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - P Kohonen
- Biotechnology Centre, University of Turku, Turku, Finland
| | - P Kovanen
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - A Happonen
- Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - S Kaewphan
- Department of Information Technology, University of Turku, Turku, Finland
| | - I Ahonen
- Biotechnology Centre, University of Turku, Turku, Finland
| | - O Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - R Grafström
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Laboratory for Toxicology, Karolinska Institute, Stockholm, Sweden
| | - J Lötjönen
- Knowledge Intensive Services, VTT Technical Research Centre of Finland, Tampere, Finland
| | - M Nees
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
| |
Collapse
|
25
|
Jia W, Eneh JO, Ratnaparkhe S, Altman MK, Murph MM. MicroRNA-30c-2* expressed in ovarian cancer cells suppresses growth factor-induced cellular proliferation and downregulates the oncogene BCL9. Mol Cancer Res 2011; 9:1732-45. [PMID: 22024689 DOI: 10.1158/1541-7786.mcr-11-0245] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that function as master regulators of posttranscriptional gene expression with each miRNA negatively regulating hundreds of genes. Lysophosphatidic acid (LPA) is a mitogenic lipid present within the ovarian tumor microenvironment and induces LPA receptor activation and intracellular signaling cascades like ERK/MAPK, leading to enhanced cellular proliferation. Here, we show that in SKOV-3 and OVCAR-3 cells, LPA stimulation at concentrations ranging from 1 nmol/L to 20 μmol/L for 30 to 60 minutes increases miR-30c-2*, and this effect is mediated through a combination of receptors because knock down of multiple LPA receptors is required for inhibition. The epidermal growth factor and platelet-derived growth factor also increase miR-30c-2* transcript expression, suggesting a broader responsive role for miR-30c-2*. Thus, we investigated the functional role of miR-30c-2* through ectopic expression of synthetic miRNA precursors of mature miRNA or antagomir transfection and observed that microRNA-30c-2* reduces, and the antagomir enhances, cell proliferation and viability in OVCAR-3, cisplatin-insensitive SKOV-3 and chemoresistant HeyA8-MDR cells. Ectopic expression of miR-30c-2* reduces BCL9 mRNA transcript abundance and BCL9 protein. Consistent with this observation, miR-30c-2* ectopic expression also reduced BCL9 luciferase reporter gene expression. In comparison with IOSE cells, all cancer cells examined showed increased BCL9 expression, which is consistent with its role in tumor progression. Taken together, this suggest that growth factor induced proliferation mediates a neutralizing response by significantly increasing miR-30c-2* which reduces BCL9 expression and cell proliferation in SKOV-3 and OVCAR-3 cells, likely as a mechanism to regulate signal transduction downstream.
Collapse
Affiliation(s)
- Wei Jia
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, The University of Georgia, Athens, GA 30602, USA
| | | | | | | | | |
Collapse
|
26
|
Li H, Zhang H, Wei G, Cai Q, Yan L, Xu Y. Tumor cell group via phospholipase A₂ is involved in prostate cancer development. Prostate 2011; 71:373-84. [PMID: 20812222 DOI: 10.1002/pros.21251] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 07/22/2010] [Indexed: 11/11/2022]
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most common malignancies among men in the United States. Further understanding of the molecular mechanisms underlying PCa tumorigenic development is critical for advancing treatment strategies for PCa. The role of Group VIA phospholipase A₂β (iPLA₂β) in cancers has recently emerged. However, the biological functions of iPLA₂β in PCa development have been minimally investigated and only in vitro studies have been reported. METHODS We tested the role of iPLA₂β in host cells using an iPLA₂β deficient mouse model and the role of iPLA₂β in tumor cells by comparing the proliferation, migration, and invasion in vitro and tumorigenesis in vivo. CONCLUSIONS iPLA₂β deficiency did not affect tumor development in C57BL/6 mice injected with syngeneic PCa cell line TRAMP-C1P3 in any of three models (subcutaneous, orthotopic, or intratibia injection) tested, suggesting that host cell iPLA₂β is not required for PCa tumorigenesis and metastasis. In contrast, when iPLA₂β was down-regulated in TRAMP-C1P3 cells, cell proliferation was reduced in vitro and tumor growth was suppressed in vivo compared to control cells. In particular, iPLA₂β was required for lysophosphatidic acid (LPA)-induced migration and invasion in TRAMP-C1P3 cells. We compared human and mouse PCa cells and showed that they shared high similarities in LPA-stimulated effects and signaling pathways. LPA stimulated cell migration and/or invasion via a PI3K-dependent pathway. Together, our results suggest that the tumor cell iPLA₂β-LPA axis may represent a novel target for PCa.
Collapse
Affiliation(s)
- Hui Li
- Department of Obstetrics and Gynecology, Indiana University, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
The invasion of cancer cells into the surrounding tissue is a prerequisite and initial step in metastasis, which is the leading cause of death from cancer. Invasive cell migration requires the formation of various structures, such as invadopodia and pseudopodia, which require actin assembly that is regulated by specialized actin nucleation factors. There is a large variety of different actin nucleators in human cells, such as formins, spire and Arp2/3-regulating proteins, and the list is likely to grow. Studies of the mechanisms of various actin nucleation factors that are involved in cancer cell function may ultimately provide new treatments for invasive and metastatic disease.
Collapse
Affiliation(s)
- Alexander Nürnberg
- Institute of Pharmacology, University of Marburg, Karl-von-Frisch-Str. 1, 35032 Marburg, Germany
| | | | | |
Collapse
|
28
|
Yu H, Li Q, Kolosov VP, Perelman JM, Zhou X. Interleukin-13 induces mucin 5AC production involving STAT6/SPDEF in human airway epithelial cells. ACTA ACUST UNITED AC 2011; 17:83-92. [PMID: 21275604 DOI: 10.3109/15419061.2010.551682] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mucus hypersecretion is commonly observed in many chronic airway inflammatory diseases. Mucin 5AC (MUC5AC) is a major airway mucin because of its high expression in goblet cells. Here, the authors identified a gene called SAM domain-containing prostate-derived Ets factor (SPDEF) that was induced by interleukin (IL)-13. Their results showed that specific knockdown of SPDEF reduced IL-13-induced MUC5AC expression in human airway epithelial cells. This finding was associated with decreased expression of anterior gradient 2 (AGR2) and Ca(2+)-activated Cl(-) channel (CLCA1), which regulate IL-13-mediated MUC5AC overproduction. Furthermore, transfection with SPDEF siRNA enhanced expression of forkhead box a2 (Foxa2), a key transcription factor that is known to prevent mucus production. The authors also demonstrated that the repression of STAT6 inhibited expression of SPDEF and MUC5AC induced by IL-13. These results show that SPDEF plays a critical role in regulating a transcriptional network mediating IL-13-induced MUC5AC synthesis dependent on STAT6.
Collapse
Affiliation(s)
- Hongmei Yu
- Division of Respiratory Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
29
|
Hooks SB, Callihan P, Altman MK, Hurst JH, Ali MW, Murph MM. Regulators of G-Protein signaling RGS10 and RGS17 regulate chemoresistance in ovarian cancer cells. Mol Cancer 2010; 9:289. [PMID: 21044322 PMCID: PMC2988731 DOI: 10.1186/1476-4598-9-289] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Accepted: 11/02/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A critical therapeutic challenge in epithelial ovarian carcinoma is the development of chemoresistance among tumor cells following exposure to first line chemotherapeutics. The molecular and genetic changes that drive the development of chemoresistance are unknown, and this lack of mechanistic insight is a major obstacle in preventing and predicting the occurrence of refractory disease. We have recently shown that Regulators of G-protein Signaling (RGS) proteins negatively regulate signaling by lysophosphatidic acid (LPA), a growth factor elevated in malignant ascites fluid that triggers oncogenic growth and survival signaling in ovarian cancer cells. The goal of this study was to determine the role of RGS protein expression in ovarian cancer chemoresistance. RESULTS In this study, we find that RGS2, RGS5, RGS10 and RGS17 transcripts are expressed at significantly lower levels in cells resistant to chemotherapy compared with parental, chemo-sensitive cells in gene expression datasets of multiple models of chemoresistance. Further, exposure of SKOV-3 cells to cytotoxic chemotherapy causes acute, persistent downregulation of RGS10 and RGS17 transcript expression. Direct inhibition of RGS10 or RGS17 expression using siRNA knock-down significantly reduces chemotherapy-induced cell toxicity. The effects of cisplatin, vincristine, and docetaxel are inhibited following RGS10 and RGS17 knock-down in cell viability assays and phosphatidyl serine externalization assays in SKOV-3 cells and MDR-HeyA8 cells. We further show that AKT activation is higher following RGS10 knock-down and RGS 10 and RGS17 overexpression blocked LPA mediated activation of AKT, suggesting that RGS proteins may blunt AKT survival pathways. CONCLUSIONS Taken together, our data suggest that chemotherapy exposure triggers loss of RGS10 and RGS17 expression in ovarian cancer cells, and that loss of expression contributes to the development of chemoresistance, possibly through amplification of endogenous AKT signals. Our results establish RGS10 and RGS17 as novel regulators of cell survival and chemoresistance in ovarian cancer cells and suggest that their reduced expression may be diagnostic of chemoresistance.
Collapse
Affiliation(s)
- Shelley B Hooks
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, GA, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Altman MK, Gopal V, Jia W, Yu S, Hall H, Mills GB, McGinnis AC, Bartlett MG, Jiang G, Madan D, Prestwich GD, Xu Y, Davies MA, Murph MM. Targeting melanoma growth and viability reveals dualistic functionality of the phosphonothionate analogue of carba cyclic phosphatidic acid. Mol Cancer 2010; 9:140. [PMID: 20529378 PMCID: PMC2895597 DOI: 10.1186/1476-4598-9-140] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 06/09/2010] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Although the incidence of melanoma in the U.S. is rising faster than any other cancer, the FDA-approved chemotherapies lack efficacy for advanced disease, which results in poor overall survival. Lysophosphatidic acid (LPA), autotaxin (ATX), the enzyme that produces LPA, and the LPA receptors represent an emerging group of therapeutic targets in cancer, although it is not known which of these is most effective. RESULTS Herein we demonstrate that thio-ccPA 18:1, a stabilized phosphonothionate analogue of carba cyclic phosphatidic acid, ATX inhibitor and LPA1/3 receptor antagonist, induced a marked reduction in the viability of B16F10 metastatic melanoma cells compared with PBS-treated control by 80-100%. Exogenous LPA 18:1 or D-sn-1-O-oleoyl-2-O-methylglyceryl-3-phosphothioate did not reverse the effect of thio-ccPA 18:1. The reduction in viability mediated by thio-ccPA 18:1 was also observed in A375 and MeWo melanoma cell lines, suggesting that the effects are generalizable. Interestingly, siRNA to LPA3 (siLPA3) but not other LPA receptors recapitulated the effects of thio-ccPA 18:1 on viability, suggesting that inhibition of the LPA3 receptor is an important dualistic function of the compound. In addition, siLPA3 reduced proliferation, plasma membrane integrity and altered morphology of A375 cells. Another experimental compound designed to antagonize the LPA1/3 receptors significantly reduced viability in MeWo cells, which predominantly express the LPA3 receptor. CONCLUSIONS Thus the ability of thio-ccPA 18:1 to inhibit the LPA3 receptor and ATX are key to its molecular mechanism, particularly in melanoma cells that predominantly express the LPA3 receptor. These observations necessitate further exploration and exploitation of these targets in melanoma.
Collapse
Affiliation(s)
- Molly K Altman
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, College of Pharmacy, 250 W. Green Street, Athens, Georgia 30602, USA
| | - Vashisht Gopal
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 7455 Fannin, 1 SCRB 2.3019, Houston, TX 77054, USA
| | - Wei Jia
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, College of Pharmacy, 250 W. Green Street, Athens, Georgia 30602, USA
| | - Shuangxing Yu
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Hassan Hall
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Gordon B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - A Cary McGinnis
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, College of Pharmacy, 250 W. Green Street, Athens, Georgia 30602, USA
| | - Michael G Bartlett
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, College of Pharmacy, 250 W. Green Street, Athens, Georgia 30602, USA
| | - Guowei Jiang
- Department of Medicinal Chemistry, The University of Utah, 419 Wakara Way, Suite 205, Salt Lake City, UT 84108, USA
| | - Damian Madan
- Echelon Biosciences Inc., 675 Arapeen Dr., Suite 302, Salt Lake City, UT 84108, USA
| | - Glenn D Prestwich
- Department of Medicinal Chemistry, The University of Utah, 419 Wakara Way, Suite 205, Salt Lake City, UT 84108, USA
| | - Yong Xu
- Department of Medicinal Chemistry, The University of Utah, 419 Wakara Way, Suite 205, Salt Lake City, UT 84108, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 7455 Fannin, 1 SCRB 2.3019, Houston, TX 77054, USA
| | - Mandi M Murph
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, College of Pharmacy, 250 W. Green Street, Athens, Georgia 30602, USA
| |
Collapse
|
31
|
Zhang D, Ouyang J, Wang N, Zhang Y, Bie J, Zhang Y. Promotion of PDGF-induced endothelial cell migration by phosphorylated VASP depends on PKA anchoring via AKAP. Mol Cell Biochem 2009; 335:1-11. [DOI: 10.1007/s11010-009-0234-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 08/13/2009] [Indexed: 12/20/2022]
|
32
|
Hasegawa Y, Murph M, Yu S, Tigyi G, Mills GB. Lysophosphatidic acid (LPA)-induced vasodilator-stimulated phosphoprotein mediates lamellipodia formation to initiate motility in PC-3 prostate cancer cells. Mol Oncol 2009; 2:54-69. [PMID: 19081821 DOI: 10.1016/j.molonc.2008.03.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Prostate cancer remains the most frequently diagnosed malignancy and the second leading cause of cancer mortality among men in the United States. Hormone refractory, metastatic disease has no molecular therapeutics to date and survival is poor. Lysophosphatidic acid (LPA) is a bioactive lipid exhibiting motility, invasive, growth, proliferative and survival effects in multiple cancer cell lineages. Cells express different combinations of LPA-specific G protein-coupled receptors, LPA(1), LPA(2) LPA(3), and LPA(4) as well as other LPA receptors, which bind LPA and thereby regulate lipid signaling. The role of specific LPA receptors in functional outcomes of lysolipid signaling remains to be fully elucidated in prostate cancer. We hypothesized that LPA can initiate cell migration through specific LPA receptors by activating actin-associating proteins involved in motility, including the vasodilator-stimulated phosphoprotein (VASP). In the present study, we demonstrate that LPA-induced lamellipodia formation in cells is dependent on LPA receptor-mediated phosphorylation of VASP, demonstrating a previously unknown regulation by LPA. LPA induces phosphorylation of VASP at Ser(157), through protein kinase A (PKA) since the stimulation was abrogated by PKA inhibition. In addition, we found the effects of LPA-induced lamellipodia formation and migration were reduced by knockdown of either VASP or LPA receptor expression, suggesting that LPA receptor-induced VASP phosphorylation is a critical mediator of migration initiation. Thus the LPA(2) and LPA(3) receptors, in addition to the previously implicated LPA(1) receptor, play a role in cellular motility potentially contributing to invasion and metastases. Emerging drugs targeting the LPA pathway may be beneficial for the treatment of metastatic progression in prostate cancer.
Collapse
Affiliation(s)
- Yutaka Hasegawa
- Department of Systems Biology, The University of Texas M. D. Anderson Cancer Center, 7435 Fannin Street, Houston, TX 77054, USA
| | | | | | | | | |
Collapse
|
33
|
Tanyi J, Rigó Jr. J. Lysophosphatidic acid as a potential target for treatment and molecular diagnosis of epithelial ovarian cancers. Orv Hetil 2009; 150:1109-18. [DOI: 10.1556/oh.2009.28631] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Az ováriumtumorok mortalitása a legmagasabb a nőgyógyászati tumorok között. Ez egyrészt a késői diagnózisnak, másrészt a hatásos terápia hiányának következménye. Az ováriumtumorok karcinogenezise és metasztázisképzése egy komplikált genetikai, molekuláris és biokémiai folyamatsor eredménye. A lizofoszfátsav (LPA) termelésének, receptorstátusának és szignáltranszdukciós útvonalának abnormalitása gyakran megtalálható az ováriumtumorokban, ami azt sejteti, hogy az LPA nagyon fontos szerepet játszik ennek a betegségnek a kialakulásában és patofiziológiájában. Így jogosan feltételezhetjük, hogy az LPA-szignálkaszkád számos célpontot szolgáltat a molekuláris kezelési módok kialakítására és jó példát mutat arra, hogyan lehet új diagnosztikus és terápiás módszereket kialakítani egyes betegségek ellen. Az LPA-t lebontó és termelő enzimcsaládoknak csak a közelmúltban történt felfedezése és a receptorspecifikus molekulák kifejlesztése új fejezetet nyithat e potenciálisan halálos betegség kezelésében. Ebben az összefoglaló tanulmányban ismertetjük, hogy a tumorsejtekben lévő LPA-t lebontó enzimek aktivitása csökkent, és ez hozzájárul a tumor progressziójához. Ugyanezen enzimek mesterségesen létrehozott, fokozott aktivitása csökkenti a tumorsejtek növekedését és elősegíti a fiziológiás viszonyok helyreállását. Bemutatjuk azokat az irodalmi adatokat, amelyek egyértelműen bizonyítják, hogy a lipidfoszfát-foszfatáz enzimek hatásukat a sejten kívüli LPA lebontásával érik el. Minthogy ez a lebontás extracellulárisan történik, ez megmagyarázza a „bystander-effect” előfordulását, amit szintén ismertetünk. Az LPA-lebontás és -termelés enzimjei, illetve az LPA-t kötő receptorok kitűnő célpontok új molekuláris terápia kidolgozására. A különböző LPA-izoformák és más lizofoszfolipidek szintváltozásainak korai detektálása segíthet a tumor korai diagnosztizálásában, illetve később a kezelés hatékonyságának követésében. A közelmúlt jelentős LPA-szignálkaszkáddal kapcsolatos kutatási eredményei azt sejtetik, hogy azok jelentős szerepet fognak játszani ennek a még mindig halálos betegségnek a kezelésében, de további kutatások szükségesek a részletek pontos megértéséhez.
Collapse
Affiliation(s)
- János Tanyi
- 1 University of Pennsylvania Health System Department of Gynecologic Oncology Philadelphia
| | - János Rigó Jr.
- 2 Semmelweis Egyetem, Általános Orvostudományi Kar I. Szülészeti és Nőgyógyászati Klinika Budapest Baross u. 27. 1088
| |
Collapse
|
34
|
Murph MM, Liu W, Yu S, Lu Y, Hall H, Hennessy BT, Lahad J, Schaner M, Helland A, Kristensen G, Børresen-Dale AL, Mills GB. Lysophosphatidic acid-induced transcriptional profile represents serous epithelial ovarian carcinoma and worsened prognosis. PLoS One 2009; 4:e5583. [PMID: 19440550 PMCID: PMC2679144 DOI: 10.1371/journal.pone.0005583] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 04/18/2009] [Indexed: 01/03/2023] Open
Abstract
Background Lysophosphatidic acid (LPA) governs a number of physiologic and pathophysiological processes. Malignant ascites fluid is rich in LPA, and LPA receptors are aberrantly expressed by ovarian cancer cells, implicating LPA in the initiation and progression of ovarian cancer. However, there is an absence of systematic data critically analyzing the transcriptional changes induced by LPA in ovarian cancer. Methodology and Principal Findings In this study, gene expression profiling was used to examine LPA-mediated transcription by exogenously adding LPA to human epithelial ovarian cancer cells for 24 h to mimic long-term stimulation in the tumor microenvironment. The resultant transcriptional profile comprised a 39-gene signature that closely correlated to serous epithelial ovarian carcinoma. Hierarchical clustering of ovarian cancer patient specimens demonstrated that the signature is associated with worsened prognosis. Patients with LPA-signature-positive ovarian tumors have reduced disease-specific and progression-free survival times. They have a higher frequency of stage IIIc serous carcinoma and a greater proportion is deceased. Among the 39-gene signature, a group of seven genes associated with cell adhesion recapitulated the results. Out of those seven, claudin-1, an adhesion molecule and phenotypic epithelial marker, is the only independent biomarker of serous epithelial ovarian carcinoma. Knockdown of claudin-1 expression in ovarian cancer cells reduces LPA-mediated cellular adhesion, enhances suspended cells and reduces LPA-mediated migration. Conclusions The data suggest that transcriptional events mediated by LPA in the tumor microenvironment influence tumor progression through modulation of cell adhesion molecules like claudin-1 and, for the first time, report an LPA-mediated expression signature in ovarian cancer that predicts a worse prognosis.
Collapse
Affiliation(s)
- Mandi M Murph
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|