1
|
Blanc-Durand F, Clemence Wei Xian L, Tan DSP. Targeting the immune microenvironment for ovarian cancer therapy. Front Immunol 2023; 14:1328651. [PMID: 38164130 PMCID: PMC10757966 DOI: 10.3389/fimmu.2023.1328651] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
Ovarian cancer (OC) is an aggressive malignancy characterized by a complex immunosuppressive tumor microenvironment (TME). Immune checkpoint inhibitors have emerged as a breakthrough in cancer therapy by reactivating the antitumor immune response suppressed by tumor cells. However, in the case of OC, these inhibitors have failed to demonstrate significant improvements in patient outcomes, and existing biomarkers have not yet identified promising subgroups. Consequently, there remains a pressing need to understand the interplay between OC tumor cells and their surrounding microenvironment to develop effective immunotherapeutic approaches. This review aims to provide an overview of the OC TME and explore its potential as a therapeutic strategy. Tumor-infiltrating lymphocytes (TILs) are major actors in OC TME. Evidence has been accumulating regarding the spontaneous TILS response against OC antigens. Activated T-helpers secrete a wide range of inflammatory cytokines with a supportive action on cytotoxic T-cells. Simultaneously, mature B-cells are recruited and play a significant antitumor role through opsonization of target antigens and T-cell recruitment. Macrophages also form an important subset of innate immunity (M1-macrophages) while participating in the immune-stimulation context. Finally, OC has shown to engage a significant natural-killer-cells immune response, exerting direct cytotoxicity without prior sensitization. Despite this initial cytotoxicity, OC cells develop various strategies to induce an immune-tolerant state. To this end, multiple immunosuppressive molecules are secreted to impair cytotoxic cells, recruit regulatory cells, alter antigen presentation, and effectively evade immune response. Consequently, OC TME is predominantly infiltrated by immunosuppressive cells such as FOXP3+ regulatory T-cells, M2-polarized macrophages and myeloid-derived suppressor cells. Despite this strong immunosuppressive state, PD-1/PD-L1 inhibitors have failed to improve outcomes. Beyond PD-1/PD-L1, OC expresses multiple other immune checkpoints that contribute to immune evasion, and each representing potential immune targets. Novel immunotherapies are attempting to overcome the immunosuppressive state and induce specific immune responses using antibodies adoptive cell therapy or vaccines. Overall, the OC TME presents both opportunities and obstacles. Immunotherapeutic approaches continue to show promise, and next-generation inhibitors offer exciting opportunities. However, tailoring therapies to individual immune characteristics will be critical for the success of these treatments.
Collapse
Affiliation(s)
- Felix Blanc-Durand
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - Lai Clemence Wei Xian
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - David S. P. Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University Centre for Cancer Research (N2CR) and Cancer Science Institute (CSI), National University of Singapore, Singapore, Singapore
| |
Collapse
|
2
|
Wu F, He J, Deng Q, Chen J, Peng M, Xiao J, Zeng Y, Yi L, Li Z, Tian R, Jiang Z. Neuroglobin inhibits pancreatic cancer proliferation and metastasis by targeting the GNAI1/EGFR/AKT/ERK signaling axis. Biochem Biophys Res Commun 2023; 664:108-116. [PMID: 37141638 DOI: 10.1016/j.bbrc.2023.04.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/11/2023] [Accepted: 04/23/2023] [Indexed: 05/06/2023]
Abstract
Pancreatic cancer is an extremely aggressive malignancy with a very disappointing prognosis. Neuroglobin (NGB), a member of the globin family, has been demonstrated to have a significant role in a variety of tumor forms. The possible role of NGB as a tumor suppressor gene in pancreatic cancer was investigated in this work. Information from the public dataset TCGA combined with GTEx was used to analyze the finding that NGB was commonly downregulated in pancreatic cancer cell lines and tissues, correlating with patient age and prognosis. The expression of NGB in pancreatic cancer was investigated via RT-PCR, qRT-PCR, and Western blot experiments. In-vitro and in-vivo assays, NGB elicited cell cycle arrest in the S phase and apoptosis, hindered migration and invasion, reversed the EMT process, and suppressed cell proliferation and development. The mechanism of action of NGB was predicted via bioinformatics analysis and validated using Western blot and co-IP experiments revealed that NGB inhibited the EGFR/AKT/ERK pathway by binding to and reducing expression of GNAI1 and p-EGFR. In addition, pancreatic cancer cells overexpressing NGB showed increased drug sensitivity to gefitinib (EGFR-TKI). In conclusion, NGB inhibits pancreatic cancer progression by specifically targeting the GNAI1/EGFR/AKT/ERK signaling axis.
Collapse
Affiliation(s)
- Fan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jin He
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qianxi Deng
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jun Chen
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Mingyu Peng
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiayi Xiao
- West China School of Medicine and West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, PR China
| | - Yiwei Zeng
- CHINA MEDICAL UNIVERSITY, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Lin Yi
- CHONGQING MEDICAL UNIVERSITY, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, PR China
| | - Zhuoqing Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Rui Tian
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
3
|
Villaseca S, Romero G, Ruiz MJ, Pérez C, Leal JI, Tovar LM, Torrejón M. Gαi protein subunit: A step toward understanding its non-canonical mechanisms. Front Cell Dev Biol 2022; 10:941870. [PMID: 36092739 PMCID: PMC9449497 DOI: 10.3389/fcell.2022.941870] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
The heterotrimeric G protein family plays essential roles during a varied array of cellular events; thus, its deregulation can seriously alter signaling events and the overall state of the cell. Heterotrimeric G-proteins have three subunits (α, β, γ) and are subdivided into four families, Gαi, Gα12/13, Gαq, and Gαs. These proteins cycle between an inactive Gα-GDP state and active Gα-GTP state, triggered canonically by the G-protein coupled receptor (GPCR) and by other accessory proteins receptors independent also known as AGS (Activators of G-protein Signaling). In this review, we summarize research data specific for the Gαi family. This family has the largest number of individual members, including Gαi1, Gαi2, Gαi3, Gαo, Gαt, Gαg, and Gαz, and constitutes the majority of G proteins α subunits expressed in a tissue or cell. Gαi was initially described by its inhibitory function on adenylyl cyclase activity, decreasing cAMP levels. Interestingly, today Gi family G-protein have been reported to be importantly involved in the immune system function. Here, we discuss the impact of Gαi on non-canonical effector proteins, such as c-Src, ERK1/2, phospholipase-C (PLC), and proteins from the Rho GTPase family members, all of them essential signaling pathways regulating a wide range of physiological processes.
Collapse
|
4
|
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
|
5
|
Lysophosphatidic Acid Receptor Antagonists and Cancer: The Current Trends, Clinical Implications, and Trials. Cells 2021; 10:cells10071629. [PMID: 34209775 PMCID: PMC8306951 DOI: 10.3390/cells10071629] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid mediator primarily derived from membrane phospholipids. LPA initiates cellular effects upon binding to a family of G protein-coupled receptors, termed LPA receptors (LPAR1 to LPAR6). LPA signaling drives cell migration and proliferation, cytokine production, thrombosis, fibrosis, angiogenesis, and lymphangiogenesis. Since the expression and function of LPA receptors are critical for cellular effects, selective antagonists may represent a potential treatment for a broad range of illnesses, such as cardiovascular diseases, idiopathic pulmonary fibrosis, voiding dysfunctions, and various types of cancers. More new LPA receptor antagonists have shown their therapeutic potentials, although most are still in the preclinical trial stage. This review provided integrative information and summarized preclinical findings and recent clinical trials of different LPA receptor antagonists in cancer progression and resistance. Targeting LPA receptors can have potential applications in clinical patients with various diseases, including cancer.
Collapse
|
6
|
Abdul Rahman M, Tan ML, Johnson SP, Hollows RJ, Chai WL, Mansell JP, Yap LF, Paterson IC. Deregulation of lysophosphatidic acid metabolism in oral cancer promotes cell migration via the up-regulation of COX-2. PeerJ 2020; 8:e10328. [PMID: 33240646 PMCID: PMC7666559 DOI: 10.7717/peerj.10328] [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: 02/18/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the sixth most common cancer worldwide and accounts for 300,000 new cases yearly. The five-year survival rate is approximately 50% and the major challenges to improving patient prognosis include late presentation, treatment resistance, second primary tumours and the lack of targeted therapies. Therefore, there is a compelling need to develop novel therapeutic strategies. In this study, we have examined the effect of lysophosphatidic acid (LPA) on OSCC cell migration, invasion and response to radiation, and investigated the contribution of cyclooxygenase-2 (COX-2) in mediating the tumour promoting effects of LPA. Using the TCGA data set, we show that the expression of the lipid phosphate phosphatases (LPP), LPP1 and LPP3, was significantly down-regulated in OSCC tissues. There was no significant difference in the expression of the ENPP2 gene, which encodes for the enzyme autotaxin (ATX) that produces LPA, between OSCCs and control tissues but ENPP2 levels were elevated in a subgroup of OSCCs. To explore the phenotypic effects of LPA, we treated OSCC cell lines with LPA and showed that the lipid enhanced migration and invasion as well as suppressed the response of the cells to irradiation. We also show that LPA increased COX-2 mRNA and protein levels in OSCC cell lines and inhibition of COX-2 activity with the COX-2 inhibitor, NS398, attenuated LPA-induced OSCC cell migration. Collectively, our data show for the first time that COX-2 mediates some of the pro-tumorigenic effects of LPA in OSCC and identifies the ATX-LPP-LPA-COX-2 pathway as a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Mariati Abdul Rahman
- Department of Oral and Craniofacial Sciences, University of Malaya, Kuala Lumpur, Malaysia.,Department of Craniofacial Diagnostics and Biosciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - May Leng Tan
- Department of Oral and Craniofacial Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Robert J Hollows
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Wen Lin Chai
- Department of Restorative Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Jason P Mansell
- Department of Applied Sciences, University of the West of England, Bristol, United Kingdom
| | - Lee Fah Yap
- Department of Oral and Craniofacial Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Ian C Paterson
- Department of Oral and Craniofacial Sciences, University of Malaya, Kuala Lumpur, Malaysia.,Oral Cancer Research and Coordinating Centre, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
Wang Z, Shi W, Tian D, Qin H, Vallance BA, Yang H, Yu HB, Yu Q. Autotaxin stimulates LPA2 receptor in macrophages and exacerbates dextran sulfate sodium-induced acute colitis. J Mol Med (Berl) 2020; 98:1781-1794. [PMID: 33128578 DOI: 10.1007/s00109-020-01997-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 10/02/2020] [Accepted: 10/20/2020] [Indexed: 01/12/2023]
Abstract
Autotaxin (ATX) is a secreted enzyme that hydrolyzes lysophosphatidylcholine (LPC) to lysophosphatidic acid (LPA) and choline. ATX has been implicated in multiple chronic inflammatory diseases, but little is known about its role in the development of inflammatory bowel disease (IBD). Here, we investigated how ATX contributed to intestinal inflammation during colitis. We found that ATX expression levels were upregulated in the intestines of ulcerative colitis (UC) patients in acute state as well as in the intestines of dextran sulfate sodium (DSS)-induced colitis mice, which is likely due to increased infiltration of inflammatory cells including macrophages. Intriguingly, the inhibition of ATX activity led to reduced production of inflammatory cytokines, as well as attenuated colitis. These findings suggest that ATX may display strong pro-inflammatory properties. Supporting this, treatment with recombinant mouse ATX (rmATX) increased the production of inflammatory cytokines and enzymes in mouse macrophage cell line RAW264.7 and bone marrow-derived macrophages (BMDM), whereas silencing ATX by siRNA reduced LPS-stimulated production of pro-inflammatory factors. Notably, we found that the levels of LPA2 (an LPA receptor) were dramatically upregulated in rmATX-treated RAW264.7 cells and DSS-treated mice. Gene silencing of lpa2 in RAW264.7 cells by siRNA led to reduced production of inflammatory cytokines. Moreover, adenovirus-mediated delivery of lpa2 short hairpin RNA into DSS-treated mice ameliorated colitis. Collectively, our research suggests that ATX may exacerbate DSS-induced colitis by activating LPA2 receptor in macrophages and represent a promising target for the treatment of IBD. KEY MESSAGES: Increased ATX expression and secretion in colitic colons are likely due to increased infiltration of inflammatory cells including macrophages. Recombinant ATX promotes, but ATX silencing inhibits, the production of inflammatory cytokines in LPS-stimulated RAW264.7 cells and BMDM. •LPA2 mediates the pro-inflammatory effects of ATX on macrophages. Inhibition of ATX and downregulation of LPA2 ameliorate DSS-induced colitis.
Collapse
Affiliation(s)
- Zi Wang
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Avenue, 1095, 430030, Wuhan, People's Republic of China
| | - Wenjie Shi
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Avenue, 1095, 430030, Wuhan, People's Republic of China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Avenue, 1095, 430030, Wuhan, People's Republic of China
| | - Hua Qin
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Avenue, 1095, 430030, Wuhan, People's Republic of China
| | - Bruce A Vallance
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Hyungjun Yang
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Hong B Yu
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada.
| | - Qin Yu
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Avenue, 1095, 430030, Wuhan, People's Republic of China.
| |
Collapse
|
8
|
Xiang H, Lu Y, Shao M, Wu T. Lysophosphatidic Acid Receptors: Biochemical and Clinical Implications in Different Diseases. J Cancer 2020; 11:3519-3535. [PMID: 32284748 PMCID: PMC7150451 DOI: 10.7150/jca.41841] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Lysophosphatidic acid (LPA, 1-acyl-2-hemolytic-sn-glycerol-3-phosphate) extracted from membrane phospholipid is a kind of simple bioactive glycophospholipid, which has many biological functions such as stimulating cell multiplication, cytoskeleton recombination, cell survival, drug-fast, synthesis of DNA and ion transport. Current studies have shown that six G-coupled protein receptors (LPAR1-6) can be activated by LPA. They stimulate a variety of signal transduction pathways through heterotrimeric G-proteins (such as Gα12/13, Gαq/11, Gαi/o and GαS). LPA and its receptors play vital roles in cancers, nervous system diseases, cardiovascular diseases, liver diseases, metabolic diseases, etc. In this article, we discussed the structure of LPA receptors and elucidated their functions in various diseases, in order to better understand them and point out new therapeutic schemes for them.
Collapse
Affiliation(s)
- Hongjiao Xiang
- Center of Chinese Medical Therapy and Systems Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yifei Lu
- Center of Chinese Medical Therapy and Systems Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mingmei Shao
- Center of Chinese Medical Therapy and Systems Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tao Wu
- Center of Chinese Medical Therapy and Systems Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
9
|
Zhou Y, Little PJ, Ta HT, Xu S, Kamato D. Lysophosphatidic acid and its receptors: pharmacology and therapeutic potential in atherosclerosis and vascular disease. Pharmacol Ther 2019; 204:107404. [DOI: 10.1016/j.pharmthera.2019.107404] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
|
10
|
Diverse Effects of Lysophosphatidic Acid Receptors on Ovarian Cancer Signaling Pathways. JOURNAL OF ONCOLOGY 2019; 2019:7547469. [PMID: 31636669 PMCID: PMC6766155 DOI: 10.1155/2019/7547469] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/09/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid with mitogenic and growth factor-like activities affecting cell invasion, cancer progression, and resistance. It is produced mainly by autotaxin and acts on six G-protein-coupled receptors, LPAR1-6. LPA has recently been implicated as a growth factor present in ascites of ovarian cancer patients. However, mitogenic pathways stimulated by LPA via its receptors may involve any novel, thus far uncharacterized, signaling pathway(s). Here we show that three LPA receptors are involved in tumor progression by activation of both the AKT and ERK signaling pathways. CRISPR-edited LPAR2 and LPAR3 knockouts have opposing effects on ERK activation, whereas LPAR6 is involved in the activation of AKT, affecting cell migration and invasion. Our study identifies specific molecular machinery triggered by LPA and its receptors that modulates tumor cells and can serve as therapeutic target in this malignancy.
Collapse
|
11
|
Tang X, McMullen TP, Brindley DN. Increasing the low lipid phosphate phosphatase 1 activity in breast cancer cells decreases transcription by AP-1 and expressions of matrix metalloproteinases and cyclin D1/D3. Am J Cancer Res 2019; 9:6129-6142. [PMID: 31534541 PMCID: PMC6735510 DOI: 10.7150/thno.37094] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023] Open
Abstract
Metastasis is the leading cause of mortality in breast cancer patients and lysophosphatidate (LPA) signaling promotes this process. LPA signaling is attenuated by lipid phosphate phosphatase-1 (LPP1) whose activity is decreased in cancers. Consequently, increasing LPP1 levels suppresses breast tumor growth and metastasis. This study shows that increasing LPP1 in breast cancer cells decreases transcription through cFos and cJun. This decreases production of cyclin D1/D3 and matrix metalloproteinases (MMPs), which provides new insights into the role of LPP1 in controlling tumor growth and metastasis. Methods: Invasiveness was determined by a Matrigel invasion assay. MMP expression was measured by qPCR, multiplex LASER bead technology and gelatin zymography. Levels of cJUN, cFOS, FRA1, cyclin D1, and cyclin D3 were determined by qPCR and western blotting. Collagen was determined by Picro-Sirius Red staining. Results: Increasing LPP1 expression inhibited invasion of MDA-MB-231 breast cancer cells through Matrigel. This was accompanied by decreases in expression of MMP-1, -3, -7, -9, -10, -12 and -13, which are transcriptionally regulated by the AP-1 complex. Increasing LPP1 attenuated the induction of mRNA of MMP-1, -3, cFOS, and cJUN by EGF or TNFα, but increased FRA1. LPP1 expression also decreased the induction of protein levels for cFOS and cJUN in nuclei and cytoplasmic fractions by EGF and TNFα. Protein levels of cyclin D1 and D3 were also decreased by LPP1. Although FRA1 in total cell lysates or cytoplasm was increased by LPP1, nuclear FRA1 was not affected. LPP1-induced decreases in MMPs in mouse tumors created with MDA-MB-231 cells were accompanied by increased collagen in the tumors and fewer lung metastases. Knockdown of LPP1 in MDA-MB-231 cells increased the protein levels of MMP-1 and -3. Human breast tumors also have lower levels of LPP1 and higher levels of cJUN, cFOS, MMP-1, -7, -8, -9, -12, -13, cyclin D1, and cyclin D3 relative to normal breast tissue. Conclusion: This study demonstrated that the low LPP1 expression in breast cancer cells is associated with high levels of cyclin D1/D3 and MMPs as a result of increased transcription by cFOS and cJUN. Increasing LPP1 expression provides a novel approach for decreasing transcription through AP-1, which could provide a strategy for decreasing tumor growth and metastasis.
Collapse
|
12
|
Activity and clinical relevance of autotaxin and lysophosphatidic acid pathways in high-grade serous carcinoma. Virchows Arch 2018; 473:463-470. [PMID: 30032361 DOI: 10.1007/s00428-018-2418-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 07/15/2018] [Indexed: 01/22/2023]
Abstract
The aim of this study was to analyze the expression, biological role and clinical relevance of autotaxin (ATX), the enzyme synthetizing lysophosphatidic acid (LPA), and LPA receptors (LPAR) in high-grade serous carcinoma (HGSC). mRNA expression by qRT-PCR of LPAR1-6 was analyzed in 155 HGSC specimens (88 effusions, 67 solid lesions). ATX mRNA expression was analyzed in 97 specimens. ATX, ERK, and AKT protein expression was studied by Western blotting. LPAR2 mRNA was overexpressed in HGSC cells in effusions compared to solid lesions, with opposite findings for LPAR3 and LPAR6 mRNA and ATX protein. Higher LPAR1 levels were significantly related to longer overall survival (OS) in pre-chemotherapy effusions (p = 0.027). Conversely, higher expression of LPAR1, LPAR2, and LPAR5 in post-chemotherapy effusions was significantly associated with shorter OS (p = 0.037, p = 0.025 and p = 0.021, respectively) and progression-free survival (PFS) (p < 0.001, p = 0.007 and p < 0.001, respectively) in univariate survival analysis. LPAR1 mRNA expression was an independent prognosticator of OS in patients with pre-chemotherapy effusions and PFS in patients with post-chemotherapy effusions (p = 0.013 both). In conclusion, LPAR mRNA and ATX protein levels are anatomic site-dependent in HGSC and the former are informative of disease outcome.
Collapse
|
13
|
Son SH, Baek SI, Ju MS, Han SG, Jung ST, Yu YG. Development of Single-Chain Antibodies Specific to Lysophosphatidic Acid Receptor 2. B KOREAN CHEM SOC 2018. [DOI: 10.1002/bkcs.11414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Sang Hyeon Son
- Department of Applied Chemistry; Kookmin University; Seoul 02707 South Korea
| | - Seung-il Baek
- Department of Applied Chemistry; Kookmin University; Seoul 02707 South Korea
| | - Man-Seok Ju
- Department of Applied Chemistry; Kookmin University; Seoul 02707 South Korea
| | - Seong-Gu Han
- Department of Applied Chemistry; Kookmin University; Seoul 02707 South Korea
| | - Sang Taek Jung
- Department of Applied Chemistry; Kookmin University; Seoul 02707 South Korea
| | - Yeon Gyu Yu
- Department of Applied Chemistry; Kookmin University; Seoul 02707 South Korea
| |
Collapse
|
14
|
Afroz S, Yagi A, Fujikawa K, Rahman MM, Morito K, Fukuta T, Watanabe S, Kiyokage E, Toida K, Shimizu T, Ishida T, Kogure K, Tokumura A, Tanaka T. Lysophosphatidic acid in medicinal herbs enhances prostaglandin E 2 and protects against indomethacin-induced gastric cell damage in vivo and in vitro. Prostaglandins Other Lipid Mediat 2018; 135:36-44. [PMID: 29462674 DOI: 10.1016/j.prostaglandins.2018.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/13/2018] [Accepted: 01/24/2018] [Indexed: 02/08/2023]
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid that induces diverse biological responses. Recently, we found that LPA ameliorates NSAIDs-induced gastric ulcer in mice. Here, we quantified LPA in 21 medicinal herbs used for treatment of gastrointestinal (GI) disorders. We found that half of them contained LPA at relatively high levels (40-240 μg/g) compared to soybean seed powder (4.6 μg/g), which we previously identified as an LPA-rich food. The LPA in peony (Paeonia lactiflora) root powder is highly concentrated in the lipid fraction that ameliorates indomethacin-induced gastric ulcer in mice. Synthetic 18:1 LPA, peony root LPA and peony root lipid enhanced prostaglandin E2 production in a gastric cancer cell line, MKN74 cells that express LPA2 abundantly. These materials also prevented indomethacin-induced cell death and stimulated the proliferation of MKN74 cells. We found that LPA was present in stomach fluids at 2.4 μM, which is an effective LPA concentration for inducing a cellular response in vitro. These results indicated that LPA is one of the active components of medicinal herbs for the treatment of GI disorder and that orally administered LPA-rich herbs may augment the protective actions of endogenous LPA on gastric mucosa.
Collapse
Affiliation(s)
- Sheuli Afroz
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan
| | - Ayano Yagi
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan
| | - Kouki Fujikawa
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan
| | - M Motiur Rahman
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan
| | - Katsuya Morito
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan
| | - Tatsuya Fukuta
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan
| | - Shiro Watanabe
- Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Emi Kiyokage
- Department of Anatomy, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan; Department of Medical Technology, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki, Okayama 701-0193, Japan
| | - Kazunori Toida
- Department of Anatomy, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan; Ultra-High Voltage Electron Microscopy Research Center, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Taro Shimizu
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan
| | - Tatsuhiro Ishida
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan
| | - Kentaro Kogure
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan
| | - Akira Tokumura
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Yasuda Women's University, Hiroshima 731-0351, Japan
| | - Tamotsu Tanaka
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8505, Japan.
| |
Collapse
|
15
|
Carmona-Rosas G, Alfonzo-Méndez MA, Hernández-Espinosa DA, Romero-Ávila MT, García-Sáinz JA. A549 cells as a model to study endogenous LPA 1 receptor signaling and regulation. Eur J Pharmacol 2017; 815:258-265. [DOI: 10.1016/j.ejphar.2017.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/29/2017] [Accepted: 09/12/2017] [Indexed: 12/12/2022]
|
16
|
Han SG, Baek SI, Son TJ, Lee H, Kim NH, Yu YG. Preparation of functional human lysophosphatidic acid receptor 2 using a P9 ∗ expression system and an amphipathic polymer and investigation of its in vitro binding preference to G α proteins. Biochem Biophys Res Commun 2017; 487:103-108. [PMID: 28392399 DOI: 10.1016/j.bbrc.2017.04.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 04/06/2017] [Indexed: 12/12/2022]
Abstract
Human lysophosphatidic acid receptor 2 (LPA2), a member of the G-protein coupled receptor family, mediates lysophosphatidic acid (LPA)-dependent signaling by recruiting various G proteins. Particularly, it is directly implicated in the progression of colorectal and ovarian cancer through G protein signaling cascades. To investigate the biochemical binding properties of LPA2 against various alpha subunits of G protein (Gα), a functional recombinant LPA2 was overexpressed in E. coli membrane with a P9∗ expression system, and the purified protein was stabilized with an amphipathic polymer that had been synthesized by coupling octylamine, glucosamine, and diethyl aminoproylamine at the carboxylic groups of poly-γ-glutamic acid. The purified LPA2 stabilized with the amphipathic polymer showed selective binding activity to the various Gα proteins as well as agonist-dependent dissociation from Gαi3. Understanding the binding properties of LPA2 against various Gα proteins advances the understanding of downstream signaling cascades of LPA2. The functional LPA2 prepared using a P9∗ expression system and an amphipathic polymer could also facilitate the development of LPA2-targeting drugs.
Collapse
Affiliation(s)
- Seong-Gu Han
- Department of Chemistry, Kookmin University, 861-1 Jeongneung-dong, Seongbuk-gu, Seoul 136-702, South Korea
| | - Seung-Il Baek
- Department of Chemistry, Kookmin University, 861-1 Jeongneung-dong, Seongbuk-gu, Seoul 136-702, South Korea
| | - Tae Jin Son
- Department of Chemistry, Kookmin University, 861-1 Jeongneung-dong, Seongbuk-gu, Seoul 136-702, South Korea
| | - Hyeongjin Lee
- Department of Chemistry, Kookmin University, 861-1 Jeongneung-dong, Seongbuk-gu, Seoul 136-702, South Korea
| | - Nam Hyuk Kim
- Department of Chemistry, Kookmin University, 861-1 Jeongneung-dong, Seongbuk-gu, Seoul 136-702, South Korea
| | - Yeon Gyu Yu
- Department of Chemistry, Kookmin University, 861-1 Jeongneung-dong, Seongbuk-gu, Seoul 136-702, South Korea.
| |
Collapse
|
17
|
Phosphorylation and Internalization of Lysophosphatidic Acid Receptors LPA1, LPA2, and LPA3. PLoS One 2015; 10:e0140583. [PMID: 26473723 PMCID: PMC4608732 DOI: 10.1371/journal.pone.0140583] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/27/2015] [Indexed: 12/31/2022] Open
Abstract
Results The lysophosphatidic acid receptors LPA1, LPA2, and LPA3 were individually expressed in C9 cells and their signaling and regulation were studied. Agonist-activation increases intracellular calcium concentration in a concentration-dependent fashion. Phorbol myristate acetate markedly inhibited LPA1- and LPA3-mediated effect, whereas that mediated by LPA2 was only partially diminished; the actions of the phorbol ester were inhibited by bisindolylmaleimide I and by overnight incubation with the protein kinase C activator, which leads to down regulation of this protein kinase. Homologous desensitization was also observed for the three LPA receptors studied, with that of LPA2 receptors being consistently of lesser magnitude; neither inhibition nor down-regulation of protein kinase C exerted any effect on homologous desensitization. Activation of LPA1–3 receptors induced ERK 1/2 phosphorylation; this effect was markedly attenuated by inhibition of epidermal growth factor receptor tyrosine kinase activity, suggesting growth factor receptor transactivation in this effect. Lysophosphatidic acid and phorbol myristate acetate were able to induce LPA1–3 phosphorylation, in time- and concentration-dependent fashions. It was also clearly observed that agonists and protein kinase C activation induced internalization of these receptors. Phosphorylation of the LPA2 subtype required larger concentrations of these agents and its internalization was less intense than that of the other subtypes. Conclusion Our data show that these three LPA receptors are phosphoproteins whose phosphorylation state is modulated by agonist-stimulation and protein kinase C-activation and that differences in regulation and cellular localization exist, among the subtypes.
Collapse
|
18
|
Si J, Su Y, Wang Y, Yan YL, Tang YL. Expressions of lysophosphatidic acid receptors in the development of human ovarian carcinoma. Int J Clin Exp Med 2015; 8:17880-17890. [PMID: 26770382 PMCID: PMC4694282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/05/2015] [Indexed: 06/05/2023]
Abstract
AIM To investigate the associations between the expressions of three lysophosphatidic acid (LPA) receptors (LPA1-3) and the development of ovarian carcinoma (OC). METHOD Ovarian tissue specimens, including normal ovarian epithelium tissues, benign ovarian tumor tissues and OC tissues were collected from patients who underwent surgical resections between March 2012 and December 2014. Immunohistochemical staining was used to detect LPA receptor expressions in ovarian tissues. Reverse transcription-polymerase chain reaction and Western blotting were used to detect mRNA and protein expression of LPA receptors, respectively. Association analysis between LPA receptors protein expression and clinical pathological characteristics was conducted. The value of LPA2 and LPA3 in discriminating OC was confirmed by receiver-operator characteristic (ROC) curves analysis. RESULTS The positive expression rates of LPA2 and LPA3 in OC group was obviously higher than normal control and benign groups. The LPA2 and LPA3 mRNA and protein levels in OC group were higher than in normal control and benign groups. LPA2 and LPA3 mRNA expression levels were positively correlated with LPA2 and LPA3 protein expression in OC group. ROC curve analysis revealed that LPA2 yield a specificity of 96.3% and a sensitivity of 97.9%, and LPA3 yield a specificity of 98.5% and a sensitivity of 97.9% for the detection of OC. CONCLUSION LPA2 and LPA3 were highly expressed in OC tissues, which may be involved in the development of OC. Further, LPA2 and LPA3 had higher sensitivity and specificity in distinguishing the OC from benign ovarian tumors, which could be potential diagnostic indictors in OC.
Collapse
Affiliation(s)
- Jinge Si
- Department of Gynecology and Obstetrics, Zhujiang Hospital of Southern Medical UniversityGuangzhou 510515, P. R. China
| | - Yuanyuan Su
- Department of Gynecology and Obstetrics, Zhongshan Affiliated Hospital of Sun Yat-Sen UniversityZhongshan 528403, P. R. China
| | - Yifeng Wang
- Department of Gynecology and Obstetrics, Zhujiang Hospital of Southern Medical UniversityGuangzhou 510515, P. R. China
| | - You-Liang Yan
- Department of Gynecology, Boai Hospital of ZhongshanZhongshan 528403, P. R. China
| | - Ya-Ling Tang
- Department of Gynecology, The First Affiliated Hospital of Xiamen UniversityXiamen 361003, P. R. China
| |
Collapse
|
19
|
Jesionowska A, Cecerska-Heryc E, Matoszka N, Dolegowska B. Lysophosphatidic acid signaling in ovarian cancer. J Recept Signal Transduct Res 2015; 35:578-84. [PMID: 26393967 DOI: 10.3109/10799893.2015.1026444] [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] [Indexed: 11/13/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid that is involved in signal transduction between cells. Plasma and ascites levels of LPA are increased in ovarian cancer patients even in the early stages and thus LPA is considered as a potential diagnostic marker for this disease. This review presents the current knowledge regarding LPA signaling in epithelial ovarian cancer. LPA stimulates proliferation, migration and invasion of ovarian cancer cells through regulation of vascular endothelial growth factor, matrix metalloproteinases, urokinase plasminogen activator, interleukin-6, interleukin-8, CXC motif chemokine ligand 12/CXC receptor 4, COX2, cyclin D1, Hippo-Yap and growth-regulated oncogene α concentrations. In this article, all of these targets and signal pathways involved in LPA influence are described.
Collapse
Affiliation(s)
| | - Elzbieta Cecerska-Heryc
- b Department of Laboratory Diagnostics and Molecular Medicine , Pomeranian Medical University , Szczecin , Poland
| | - Natalia Matoszka
- b Department of Laboratory Diagnostics and Molecular Medicine , Pomeranian Medical University , Szczecin , Poland
| | - Barbara Dolegowska
- b Department of Laboratory Diagnostics and Molecular Medicine , Pomeranian Medical University , Szczecin , Poland
| |
Collapse
|
20
|
Chien HY, Lu CS, Chuang KH, Kao PH, Wu YL. Attenuation of LPS-induced cyclooxygenase-2 and inducible NO synthase expression by lysophosphatidic acid in macrophages. Innate Immun 2015; 21:635-46. [DOI: 10.1177/1753425915576345] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/12/2015] [Indexed: 01/09/2023] Open
Abstract
LPS can activate the inflammatory cascades by inducing various inflammatory mediators, such as prostaglandin E2 (PGE2) resulting from cyclooxygenase-2 (COX-2), and NO produced by inducible NO synthase (iNOS). Lysophosphatidic acid (LPA) has been demonstrated to participate in inflammation. This study aimed to clarify the impact and the involving mechanisms of LPA on LPS-incurred inflammation in macrophages. First, LPA appeared to attenuate LPS-induced protein and mRNA expression of COX-2 and iNOS genes, as well as production of PGE2 and NO. By using selective inhibitors targeting various signaling players, the inhibitory G protein alpha subunit (Gαi) seemed to be involved in the effect of LPA; p38, ERK and NF-κB were involved in the LPS-mediated COX-2/PGE2 pathway; and p38, JNK, phosphoinositide-3-kinase and NF-κB were involved in the LPS-mediated iNOS/NO pathway. LPA was able to diminish LPS-induced phosphorylation of p38 and Akt, as well as NF-κB p65 nuclear translocation. By utilization of inhibitors of COX-2 and iNOS, there appeared to be no modulation between the COX-2/PGE2 and the iNOS/NO signaling pathways. Our findings demonstrate a clear anti-inflammatory role of LPA acting via Gαi in LPS-mediated inflammatory response in macrophages, owing, at least in part, to its suppressive effect on LPS-induced activation of p38, Akt and NF-κB.
Collapse
Affiliation(s)
- Han-Yuan Chien
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Shen Lu
- Department of Neurosurgery, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Kun-Han Chuang
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Pu-Hong Kao
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yuh-Lin Wu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
21
|
Kihara Y, Maceyka M, Spiegel S, Chun J. Lysophospholipid receptor nomenclature review: IUPHAR Review 8. Br J Pharmacol 2014; 171:3575-94. [PMID: 24602016 PMCID: PMC4128058 DOI: 10.1111/bph.12678] [Citation(s) in RCA: 268] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/03/2014] [Accepted: 02/12/2014] [Indexed: 12/11/2022] Open
Abstract
Lysophospholipids encompass a diverse range of small, membrane-derived phospholipids that act as extracellular signals. The signalling properties are mediated by 7-transmembrane GPCRs, constituent members of which have continued to be identified after their initial discovery in the mid-1990s. Here we briefly review this class of receptors, with a particular emphasis on their protein and gene nomenclatures that reflect their cognate ligands. There are six lysophospholipid receptors that interact with lysophosphatidic acid (LPA): protein names LPA1 - LPA6 and italicized gene names LPAR1-LPAR6 (human) and Lpar1-Lpar6 (non-human). There are five sphingosine 1-phosphate (S1P) receptors: protein names S1P1 -S1P5 and italicized gene names S1PR1-S1PR5 (human) and S1pr1-S1pr5 (non-human). Recent additions to the lysophospholipid receptor family have resulted in the proposed names for a lysophosphatidyl inositol (LPI) receptor - protein name LPI1 and gene name LPIR1 (human) and Lpir1 (non-human) - and three lysophosphatidyl serine receptors - protein names LyPS1 , LyPS2 , LyPS3 and gene names LYPSR1-LYPSR3 (human) and Lypsr1-Lypsr3 (non-human) along with a variant form that does not appear to exist in humans that is provisionally named LyPS2L . This nomenclature incorporates previous recommendations from the International Union of Basic and Clinical Pharmacology, the Human Genome Organization, the Gene Nomenclature Committee, and the Mouse Genome Informatix.
Collapse
Affiliation(s)
- Yasuyuki Kihara
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research InstituteLa Jolla, CA, USA
| | - Michael Maceyka
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, School of Medicine, Virginia Commonwealth UniversityRichmond, VA, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, School of Medicine, Virginia Commonwealth UniversityRichmond, VA, USA
| | - Jerold Chun
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research InstituteLa Jolla, CA, USA
| |
Collapse
|
22
|
Knowlden SA, Capece T, Popovic M, Chapman TJ, Rezaee F, Kim M, Georas SN. Regulation of T cell motility in vitro and in vivo by LPA and LPA2. PLoS One 2014; 9:e101655. [PMID: 25003200 PMCID: PMC4086949 DOI: 10.1371/journal.pone.0101655] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 06/10/2014] [Indexed: 12/11/2022] Open
Abstract
Lysophosphatidic acid (LPA) and the LPA-generating enzyme autotaxin (ATX) have been implicated in lymphocyte trafficking and the regulation of lymphocyte entry into lymph nodes. High local concentrations of LPA are thought to be present in lymph node high endothelial venules, suggesting a direct influence of LPA on cell migration. However, little is known about the mechanism of action of LPA, and more work is needed to define the expression and function of the six known G protein-coupled receptors (LPA 1-6) in T cells. We studied the effects of 18∶1 and 16∶0 LPA on naïve CD4+ T cell migration and show that LPA induces CD4+ T cell chemorepulsion in a Transwell system, and also improves the quality of non-directed migration on ICAM-1 and CCL21 coated plates. Using intravital two-photon microscopy, lpa2-/- CD4+ T cells display a striking defect in early migratory behavior at HEVs and in lymph nodes. However, later homeostatic recirculation and LPA-directed migration in vitro were unaffected by loss of lpa2. Taken together, these data highlight a previously unsuspected and non-redundant role for LPA2 in intranodal T cell motility, and suggest that specific functions of LPA may be manipulated by targeting T cell LPA receptors.
Collapse
Affiliation(s)
- Sara A. Knowlden
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Tara Capece
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Milan Popovic
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Timothy J. Chapman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Fariba Rezaee
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Steve N. Georas
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
23
|
Tanaka T, Ohmoto M, Morito K, Kondo H, Urikura M, Satouchi K, Tokumura A. Type 2 lysophosphatidic acid receptor in gastric surface mucous cells: Possible implication of prostaglandin E2 production. Biofactors 2014; 40:355-61. [PMID: 24375908 DOI: 10.1002/biof.1147] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 08/24/2013] [Accepted: 09/06/2013] [Indexed: 02/03/2023]
Abstract
Lysophosphatidic acid (LPA) is a lipid mediator that induces various cell responses via its specific receptors. Recently, we found that orally administered LPA and phosphatidic acid (PA) ameliorate stress- or aspirin-induced stomach injury. However, the mechanisms underlying these effects have not been elucidated yet. In this study, we examined effect of LPA on prostaglandin (PG) E2 production in MKN74 cells, a gastric cell-line expressing type 2 LPA receptor (LPA2). When the cells were treated with LPA, the level of mRNA of COX-2 but not COX-1 was upregulated. The LPA effect was abolished when the cells were pretreated with pertussis toxin (PTX), suggesting the involvement of receptor(s) coupled with Gi. Pretreatment of MKN74 cells with LPA enhanced the PGE2 production triggered by calcium ionophore A23187. Again, PTX abolished the LPA effect. Fluorescent immunohistochemistry using an antibody against LPA2 showed that surface mucous cells (pit cells) in gastric mucosa of mice express LPA2 on the apical side of the plasma membrane. These results suggest that LPA in the diet or its digestion may contribute to the epithelial integrity of stomach mucosa by enhancement of PGE2 production via activation of LPA2.
Collapse
Affiliation(s)
- Tamotsu Tanaka
- Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | | | | | | | | | | | | |
Collapse
|
24
|
Yung YC, Stoddard NC, Chun J. LPA receptor signaling: pharmacology, physiology, and pathophysiology. J Lipid Res 2014; 55:1192-214. [PMID: 24643338 DOI: 10.1194/jlr.r046458] [Citation(s) in RCA: 556] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Indexed: 12/18/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a small ubiquitous lipid found in vertebrate and nonvertebrate organisms that mediates diverse biological actions and demonstrates medicinal relevance. LPA's functional roles are driven by extracellular signaling through at least six 7-transmembrane G protein-coupled receptors. These receptors are named LPA1-6 and signal through numerous effector pathways activated by heterotrimeric G proteins, including Gi/o, G12/13, Gq, and Gs LPA receptor-mediated effects have been described in numerous cell types and model systems, both in vitro and in vivo, through gain- and loss-of-function studies. These studies have revealed physiological and pathophysiological influences on virtually every organ system and developmental stage of an organism. These include the nervous, cardiovascular, reproductive, and pulmonary systems. Disturbances in normal LPA signaling may contribute to a range of diseases, including neurodevelopmental and neuropsychiatric disorders, pain, cardiovascular disease, bone disorders, fibrosis, cancer, infertility, and obesity. These studies underscore the potential of LPA receptor subtypes and related signaling mechanisms to provide novel therapeutic targets.
Collapse
Affiliation(s)
- Yun C Yung
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| | - Nicole C Stoddard
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037 Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine, La Jolla, CA 92037
| | - Jerold Chun
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
25
|
Jeong KJ, Cho KH, Panupinthu N, Kim H, Kang J, Park CG, Mills GB, Lee HY. EGFR mediates LPA-induced proteolytic enzyme expression and ovarian cancer invasion: inhibition by resveratrol. Mol Oncol 2012; 7:121-9. [PMID: 23127547 DOI: 10.1016/j.molonc.2012.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 10/08/2012] [Accepted: 10/08/2012] [Indexed: 01/09/2023] Open
Abstract
Lysophosphatidic acid (LPA) augments proliferation and metastasis of various cancer cells. We recently identified a critical role of the Rho/ROCK pathway for LPA-induced proteolytic enzyme expression and cancer cell progression. In the present study, we elucidate the underlying mechanisms by which LPA induces Rho activation and subsequent cellular invasion, and the reversal of these effects by resveratrol. We observed that both Gi and G13 contribute to LPA-induced EGFR activation. The activated EGFR in turn initiates a Ras/Rho/ROCK signaling cascade, leading to proteolytic enzyme secretion. Further we provide evidence that resveratrol inhibits EGFR phosphorylation and subsequent activation of a Ras/Rho/ROCK signaling. Therefore, we demonstrate a mechanistic cascade of LPA activating EGFR through Gi and G13 thus inducing a Ras/Rho/ROCK signaling for proteolytic enzyme expression and ovarian cancer cell invasion, as well as interference of the cascade by resveratrol through blocking EGFR phosphorylation.
Collapse
Affiliation(s)
- Kang Jin Jeong
- Department of Pharmacology, College of Medicine, Konyang University, Daejeon 302-718, Korea
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Gong YL, Tao CJ, Hu M, Chen JF, Cao XF, Lv GM, Li P. Expression of lysophosphatidic acid receptors and local invasiveness and metastasis in Chinese pancreatic cancers. ACTA ACUST UNITED AC 2012; 19:eS15-21. [PMID: 22876164 DOI: 10.3747/co.19.1138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVES The present study evaluated the potential role of lysophosphatidic acid receptors (lpars) in processes leading to local invasiveness and metastasis in Chinese pancreatic carcinoma. METHODS Real-time reverse-transcriptase polymerase chain reaction and Western blot analysis were used to detect expression of lpars in tumour and adjacent non-tumour tissues from patients with surgically resected pancreatic carcinoma. Surgical specimens from 50 patients were examined for relative expression of each receptor's messenger rna (mrna) and protein. Findings were analyzed for correlations with tumour size, pathologic classification, clinical stage, and infiltration of capsule and lymphonodi. RESULTS Increased levels of mrna of lpars (lpar1 ≈ lpar3 < lpar2) were found in the pancreatic cancer tissues examined. Low levels of transcripts for lpar1, lpar2, and lpar3 receptors were detectable in adjacent non-tumour tissues. The difference in lpar1 protein expression between tumour and adjacent non-tumour tissues does not seem significant, but the signals of lpar2 expression in pancreatic cancer tumour tissues were significantly amplified compared with those in adjacent non-tumour tissues. Tumour and adjacent non-tumour tissues both weakly expressed lpar3 protein with no statistical difference. However, expression of lpar1, lpar2, and lpar3 showed an obvious correlation with infiltration of capsule cells, surrounding lymphonodi, and specific histopathologic features. CONCLUSIONS Lysophosphatidic acid receptor is a promising indicator for pancreatic cancer, and our findings suggested that lpar2 might be a potential target for clinical treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Y L Gong
- Department of Oncology, Nanjing Hospital Affiliated to Nanjing Medical University, Nanjing, PR China
| | | | | | | | | | | | | |
Collapse
|
27
|
Mukherjee A, Wu J, Barbour S, Fang X. Lysophosphatidic acid activates lipogenic pathways and de novo lipid synthesis in ovarian cancer cells. J Biol Chem 2012; 287:24990-5000. [PMID: 22665482 DOI: 10.1074/jbc.m112.340083] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
One of the most common molecular changes in cancer is the increased endogenous lipid synthesis, mediated primarily by overexpression and/or hyperactivity of fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC). The changes in these key lipogenic enzymes are critical for the development and maintenance of the malignant phenotype. Previous efforts to control oncogenic lipogenesis have been focused on pharmacological inhibitors of FAS and ACC. Although they show anti-tumor effects in culture and in mouse models, these inhibitors are nonselective blockers of lipid synthesis in both normal and cancer cells. To target lipid anabolism in tumor cells specifically, it is important to identify the mechanism governing hyperactive lipogenesis in malignant cells. In this study, we demonstrate that lysophosphatidic acid (LPA), a growth factor-like mediator present at high levels in ascites of ovarian cancer patients, regulates the sterol regulatory element binding protein-FAS and AMP-activated protein kinase-ACC pathways in ovarian cancer cells but not in normal or immortalized ovarian epithelial cells. Activation of these lipogenic pathways is linked to increased de novo lipid synthesis. The pro-lipogenic action of LPA is mediated through LPA(2), an LPA receptor subtype overexpressed in ovarian cancer and other malignancies. Downstream of LPA(2), the G(12/13) and G(q) signaling cascades mediate LPA-dependent sterol regulatory element-binding protein activation and AMP-activated protein kinase inhibition, respectively. Moreover, inhibition of de novo lipid synthesis dramatically attenuated LPA-induced cell proliferation. These results demonstrate that LPA signaling is causally linked to the hyperactive lipogenesis in ovarian cancer cells, which can be exploited for development of new anti-cancer therapies.
Collapse
Affiliation(s)
- Abir Mukherjee
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia 23298, USA
| | | | | | | |
Collapse
|
28
|
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
|
29
|
Lin CC, Lin WN, Cheng SE, Tung WH, Wang HH, Yang CM. Transactivation of EGFR/PI3K/Akt involved in ATP-induced inflammatory protein expression and cell motility. J Cell Physiol 2012; 227:1628-38. [PMID: 21678415 DOI: 10.1002/jcp.22880] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Phenotype transition of vascular smooth muscle cells (VSMCs) is important in vascular diseases, such as atherosclerosis and restenosis. Once released, ATP may promote activation of VSMCs by stimulating cyclooxygenase-2 (COX-2), cytosolic phospholipase A(2) (cPLA(2)) expression and prostaglandin (PG)E(2) synthesis via activation of MAPKs and NF-κB. However, whether alternative signaling pathways participated in regulating COX-2 and cPLA(2) expression associated with cell migration were investigated in rat VSMCs. Western blot analysis, RT-PCR, promoter assay and PGE(2) ELISA were used to determine expression of COX-2, cPLA(2) and PGE(2). Specific inhibitors and siRNAs against various protein kinases or transcription factors were used to investigate the related signaling components in inflammatory protein induction by ATPγS. We found that ATPγS-induced COX-2 and cPLA(2) expression and PGE(2) release was attenuated by the pharmacological inhibitors or transfection with siRNA against PKCδ, c-Src, EGFR, PI3-K, Akt, p44/p42 MAPK or Elk-1. Moreover, ATPγS-stimulated phosphorylation of PKCδ, c-Src, EGFR, Akt, p42/p44 MAPK and Elk-1, suggesting the participation of PKCδ/c-Src/EGFR/PI3-K/Akt/p42/p44 MAPK cascade in mediating Elk-1 activities in VSMCs. In addition, migration assay revealed that ATPγS promoted cell mobility through up-regulation of COX-2 and cPLA(2) expression and PGE(2) release, which was attenuated by pretreatment with PGE(2) receptor antagonists. Taken together, these data showed that ATPγS up-regulated the expression of COX-2 and cPLA(2) through transactivation of PKCδ/c-Src/EGFR/PI3K/Akt/Elk-1 pathway. Newly synthesized PGE(2) acted on its receptors to promote cell motility of ATPγS-stimulated VSMCs.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital and Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Src-family Kinases (SFKs) participate in the regulation of proliferation, differentiation, apoptosis, autophagy, adhesion, migration, invasion and angiogenesis in normal and cancer cells. Abnormal expression of SFKs has been documented in cancers that arise in breast, colon, ovary, melanocyte, gastric mucosa, head and neck, pancreas, lung, and brain. Targeting SFKs in cancer cells has been shown to be a promising therapeutic strategy in solid tumors, particularly in ovarian, colon and breast cancers. Paclitaxel is one of most widely used chemotherapeutic agents for the management of ovarian, breast, lung and head/neck cancers. As a microtubule-stabilizing agent, paclitaxel possesses both mitosis-dependent and mitosis-independent activities against cancer cells. A variety of mechanisms such as deregulation of P-glycoprotein, alteration of tubulin isotypes, alteration of microtubule-regulatory proteins, deregulation of apoptotic signaling pathways, mutation of tubulins and overexpression of copper transporters have been implicated in the development of primary or secondary resistance to paclitaxel. By affecting cancer cell survival, proliferation, autophagy, microtubule stability, motility, and/or angiogenesis, SFKs interact with mechanisms that regulate paclitaxel sensitivity. Inhibition of SFKs can potentiate the anti-tumor activity of paclitaxel by enhancing apoptosis, autophagy and microtubule stability. Based on pre-clinical observations, administration of SFK inhibitors in combination with paclitaxel could improve treatment for ovarian, breast, lung and head/neck cancers. Identification and validation of predictive biomarkers could also permit personalization of the therapy.
Collapse
Affiliation(s)
- Xiao-Feng Le
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, USA.
| | | |
Collapse
|
31
|
Dutta S, Wang FQ, Wu HS, Mukherjee TJ, Fishman DA. The NF-κB pathway mediates lysophosphatidic acid (LPA)-induced VEGF signaling and cell invasion in epithelial ovarian cancer (EOC). Gynecol Oncol 2011; 123:129-37. [PMID: 21782227 DOI: 10.1016/j.ygyno.2011.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 06/07/2011] [Accepted: 06/08/2011] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Our previous report has implicated the involvement of VEGF-VEGFR-2 h signaling in LPA-induced EOC invasion. However, the mechanism by which LPA regulates VEGF and VEGFR-2 expression remains to be elucidated. In the present study, we systematically examined the signal transduction pathways activated by LPA and further evaluated whether LPA's effect on VEGF-VEGFR-2 signaling and EOC invasion was mediated by the activation of NF-κB pathway. METHODS Using a signal transduction PathwayFinder PCR array, we examined the expression change of 86 key genes representing 18 signal transduction pathways in DOV13 and SKOV3 cells upon LPA (20 μM) treatment. We also used quantitative PCR, Western blotting and ELISA to evaluate the effect of NF-κB pathway inhibition on VEGF(121), VEGF(165) and VEGFR-2 mRNA and protein expression/secretion with or without the presence of LPA (20 μM) in SKOV3. Cell invasion under various treatment conditions was assessed by Matrigel invasion assay and MMP-2 secretion was detected by gelatin zymography. RESULTS Our results showed that in both DOV13 and SKOV3, several of the NF-κB pathway components, such as TNF, are consistently activated by LPA stimulation. In addition, treatment with an NF-κB pathway activation inhibitor, at 10 μM, significantly decreased LPA-induced VEGF(121), VEGF(165) and VEGFR-2 mRNA expression and VEGF secretion, as well as LPA-induced SKOV3 invasion (p<0.05). When combined with an EGFR inhibitor, NF-κB pathway inhibition exhibited a significantly stronger effect than used alone (p<0.05) on reducing LPA-induced VEGF secretion and cell invasion. Additionally, NF-κB inhibition also decreased LPA-induced MMP-2 secretion and MMP-1 expression (p<0.05). CONCLUSIONS These results suggest that the NF-κB pathway plays an important role in LPA-induced VEGF signaling and EOC invasion and targeting this pathway may reveal potential therapeutic options for metastatic EOC.
Collapse
Affiliation(s)
- Sonia Dutta
- Department of Obstetrics, Gynecology and Reproductive Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | |
Collapse
|
32
|
Noske A, Schwabe M, Weichert W, Darb-Esfahani S, Buckendahl AC, Sehouli J, Braicu EI, Budczies J, Dietel M, Denkert C. An intracellular targeted antibody detects EGFR as an independent prognostic factor in ovarian carcinomas. BMC Cancer 2011; 11:294. [PMID: 21756326 PMCID: PMC3149030 DOI: 10.1186/1471-2407-11-294] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Accepted: 07/14/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In ovarian cancer, the reported rate of EGFR expression varies between 4-70% depending on assessment method and data on patient outcome are conflicting. METHODS In this study we investigated EGFR expression and its prognostic value in a cohort of 121 invasive ovarian carcinomas, using a novel antibody against the intracellular domain of the receptor. We further evaluated an association between EGFR, the nuclear transporter CRM1 as well as COX-2. Furthermore, we evaluated EGFR expression in ten ovarian cancer cell lines and incubated cancer cells with Leptomycin B, a CRM1 specific inhibitor. RESULTS We observed a membranous and cytoplasmic EGFR expression in 36.4% and 64% of ovarian carcinomas, respectively. Membranous EGFR was an independent prognostic factor for poor overall survival in ovarian cancer patients (HR 2.7, CI 1.1-6.4, p = 0.02) which was also found in the serous subtype (HR 4.6, CI 1.6-13.4, p = 0.004). We further observed a significant association of EGFR with COX-2 and nuclear CRM1 expression (chi-square test for trends, p = 0.006 and p = 0.013, respectively). In addition, combined membranous EGFR/COX-2 expression was significantly related to unfavorable overall survival (HR 7.2, CI 2.3-22.1, p = 0.001).In cell culture, we observed a suppression of EGFR protein levels after exposure to Leptomycin B in OVCAR-3 and SKOV-3 cells. CONCLUSIONS Our results suggest that the EGFR/COX-2/CRM1 interaction might be involved in progression of ovarian cancer and patient prognosis. Hence, it is an interesting anti-cancer target for a combination therapy. Further studies will also be needed to investigate whether EGFR is also predictive for benefit from EGFR targeted therapies.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Neoplasm/immunology
- Antibody Specificity
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/immunology
- Carcinoma/chemistry
- Carcinoma/mortality
- Carcinoma/pathology
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/metabolism
- Cohort Studies
- Cyclooxygenase 2/analysis
- ErbB Receptors/analysis
- ErbB Receptors/biosynthesis
- ErbB Receptors/genetics
- ErbB Receptors/immunology
- Fatty Acids, Unsaturated/pharmacology
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immunoenzyme Techniques
- Kaplan-Meier Estimate
- Karyopherins/analysis
- Karyopherins/antagonists & inhibitors
- Middle Aged
- Neoplasm Proteins/analysis
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Ovarian Neoplasms/chemistry
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/pathology
- Prognosis
- Protein Structure, Tertiary
- Rabbits
- Receptors, Cytoplasmic and Nuclear/analysis
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Single-Blind Method
- Exportin 1 Protein
Collapse
Affiliation(s)
- Aurelia Noske
- Institute of Pathology, University Hospital Charité Berlin, Germany
- Institute of Pathology, University Hospital Zurich, Switzerland
| | - Michael Schwabe
- Institute of Pathology, University Hospital Charité Berlin, Germany
| | - Wilko Weichert
- Institute of Pathology and National Center for Tumor Diseases, Ruprecht-Karls-Universität, Heidelberg, Germany
| | | | | | - Jalid Sehouli
- Department of Gynecology, University Hospital Charité Berlin, Germany
| | - Elena I Braicu
- Department of Gynecology, University Hospital Charité Berlin, Germany
| | - Jan Budczies
- Institute of Pathology, University Hospital Charité Berlin, Germany
| | - Manfred Dietel
- Institute of Pathology, University Hospital Charité Berlin, Germany
| | - Carsten Denkert
- Institute of Pathology, University Hospital Charité Berlin, Germany
| |
Collapse
|
33
|
LV G, Li P, Wang W, Wang S, Chen J, Gong Y. Lysophosphatidic acid (LPA) and endothelial differentiation gene (Edg) receptors in human pancreatic cancer. J Surg Oncol 2011; 104:685-91. [DOI: 10.1002/jso.22016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/09/2011] [Indexed: 11/06/2022]
|
34
|
Kitz K, Windischhofer W, Leis HJ, Huber E, Kollroser M, Malle E. 15-Deoxy-Δ12,14-prostaglandin J2 induces Cox-2 expression in human osteosarcoma cells through MAPK and EGFR activation involving reactive oxygen species. Free Radic Biol Med 2011; 50:854-65. [PMID: 21236332 DOI: 10.1016/j.freeradbiomed.2010.12.039] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 12/14/2010] [Accepted: 12/31/2010] [Indexed: 11/25/2022]
Abstract
Prostaglandins (PGs), important modulators in bone biology, may also contribute to tumor formation and progression in human osteosarcoma. 15-Deoxy-Δ(12,14)-PGJ(2) (15d-PGJ(2)), a metabolite of PGD(2) and PPARγ-ligand, exerts a panel of biological activities via receptor-dependent and -independent mechanisms. As inducible cyclooxygenase-2 (Cox-2) is a candidate inflammatory marker in human osteosarcoma and a rate-limiting enzyme in PG biosynthesis, this study aimed at investigating intracellular redox status and signaling cascades leading to Cox-2 induction in human MG-63 osteosarcoma cells. 15d-PGJ(2) induced the accumulation of reactive oxygen species (ROS) that in turn may lead to upregulation of Cox-2 via two different routes in a PPARγ-independent manner. First, phosphorylation of p38 MAPK directly enhances Cox-2 expression by promoting mRNA stability. Second, 15d-PGJ(2) induces activation of epidermal growth factor receptors and downstream activation of Cox-2 via phosphorylation of p42/44 MAPK. Glutathione precursor molecules reversed enhanced ROS levels and Cox-2 expression. Functional activity of Cox-2 expression was tested by measurement of PGE(2) and PGF(2α). The synthetic compound 9,10-dihydro-15d-PGJ(2) lacking the α,β-unsaturated carbonyl group in the cyclopentenone ring did not exhibit the cellular responses observed with 15d-PGJ(2). We conclude that the electrophilic carbon atom of 15d-PGJ(2) is responsible for alterations in intracellular redox status and Cox-2 expression.
Collapse
Affiliation(s)
- Kerstin Kitz
- Department of Pediatrics and Adolescence Medicine, Research Unit of Osteological Research and Analytical Mass Spectrometry, Medical University of Graz, A-8036 Graz, Austria
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
G protein-coupled receptors (GPCRs) belong to a superfamily of cell surface signalling proteins that have a pivotal role in many physiological functions and in multiple diseases, including the development of cancer and cancer metastasis. Current drugs that target GPCRs - many of which have excellent therapeutic benefits - are directed towards only a few GPCR members. Therefore, huge efforts are currently underway to develop new GPCR-based drugs, particularly for cancer. We review recent findings that present unexpected opportunities to interfere with major tumorigenic signals by manipulating GPCR-mediated pathways. We also discuss current data regarding novel GPCR targets that may provide promising opportunities for drug discovery in cancer prevention and treatment.
Collapse
|
36
|
Chen J, Ou-Yang X, Gao J, Zhu J, He X, Rong J. Knockdown of ribonuclease inhibitor expression with siRNA in non-invasive bladder cancer cell line BIU-87 promotes growth and metastasis potentials. Mol Cell Biochem 2010; 349:83-95. [PMID: 21125316 DOI: 10.1007/s11010-010-0663-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2010] [Accepted: 11/15/2010] [Indexed: 10/18/2022]
Abstract
Human ribonuclease inhibitor (RI) is a cytoplasmic acidic protein. RI is constructed almost entirely of leucine-rich repeats, which might be involved in some unknown biological functions like other structurally similar proteins besides inhibiting RNase A and angiogenin activities. Our previous experiments demonstrated that up-regulating RI might effectively inhibit some tumor growth and metastasis. However, the down-regulating RI influence on the tumor does not have any report until now, the mechanisms underlying antitumor of RI have not been fully understood. In this study, the efficient RNA interferences of RI were constructed using a plasmid vector and identified with RT-PCR, Western blot and Immunocytochemistry, then were transfected into non-invasive bladder cancer BIU-87 cells. We demonstrated that knockdown RI expression in BIU-87 cells could obviously change the cell morphology, rearrange the microfilaments and extend the lamellipodia, as well as enhance proliferation, increase migration, invasion and matrix metalloprotease level, and also reduce adhesion in vitro. BALB/C nude mice that were injected with the BIU-87 cells transfected RI siRNA showed a significant facilitation of the tumor with heavier tumor weight, higher density of microvessels, lower nm23-H1 and E-Cadherin expressions than those in the control group. Taken together, these experiments suggest that knockdown of RI could promote growth and metastasis potentials of BIU-87 cells. Our present findings reveal the novel mechanism that anti-tumor effect of RI is also involved in suppressing growth and metastasis, besides antiangiogenesis. The results show that RI may be a therapeutic target protein for bladder cancer and may be of biological importance.
Collapse
Affiliation(s)
- Junxia Chen
- Department of Cell Biology and Genetics, Chongqing Medical University, Yuzhong District, Chongqing, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
37
|
Holmström TE, Mattsson CL, Wang Y, Iakovleva I, Petrovic N, Nedergaard J. Non-transactivational, dual pathways for LPA-induced Erk1/2 activation in primary cultures of brown pre-adipocytes. Exp Cell Res 2010; 316:2664-75. [PMID: 20576526 DOI: 10.1016/j.yexcr.2010.05.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 05/21/2010] [Accepted: 05/24/2010] [Indexed: 12/17/2022]
Abstract
In many cell types, G-protein-coupled receptor (GPCR)-induced Erk1/2 MAP kinase activation is mediated via receptor tyrosine kinase (RTK) transactivation, in particular via the epidermal growth factor (EGF) receptor. Lysophosphatidic acid (LPA), acting via GPCRs, is a mitogen and MAP kinase activator in many systems, and LPA can regulate adipocyte proliferation. The mechanism by which LPA activates the Erk1/2 MAP kinase is generally accepted to be via EGF receptor transactivation. In primary cultures of brown pre-adipocytes, EGF can induce Erk1/2 activation, which is obligatory and determinant for EGF-induced proliferation of these cells. Therefore, we have here examined whether LPA, via EGF transactivation, can activate Erk1/2 in brown pre-adipocytes. We found that LPA could induce Erk1/2 activation. However, the LPA-induced Erk1/2 activation was independent of transactivation of EGF receptors (or PDGF receptors) in these cells (whereas in transformed HIB-1B brown adipocytes, the LPA-induced Erk1/2 activation indeed proceeded via EGF receptor transactivation). In the brown pre-adipocytes, LPA instead induced Erk1/2 activation via two distinct non-transactivational pathways, one G(i)-protein dependent, involving PKC and Src activation, the other, a PTX-insensitive pathway, involving PI3K (but not Akt) activation. Earlier studies showing LPA-induced Erk1/2 activation being fully dependent on RTK transactivation have all been performed in cell lines and transfected cells. The present study implies that in non-transformed systems, RTK transactivation may not be involved in the mediation of GPCR-induced Erk1/2 MAP kinase activation.
Collapse
Affiliation(s)
- Therese E Holmström
- Department of Physiology, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
38
|
Pua TL, Wang FQ, Fishman DA. Roles of LPA in ovarian cancer development and progression. Future Oncol 2010; 5:1659-73. [PMID: 20001802 DOI: 10.2217/fon.09.120] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Lysophosphatidic acid (LPA), a bioactive phospholipid, stimulates survival, proliferation, adhesion, migration and invasion of ovarian cancer cells through the activation of G-protein-coupled plasma membrane receptors. LPA and its receptors are aberrantly expressed in ovarian cancer, with high levels predominantly found in malignant ascites and in the plasma of ovarian cancer patients. LPA signals multiple intracellular pathways, such as Ras/MEKK1-MAPK and PI3K/Akt, to promote growth factors and protease expression, and induce angiogenesis and tumor cell invasion through the extracellular matrix and across the basement membrane. Only a small portion of this intricate lipid-signaling cascade has been characterized thus far. We believe that elucidation of this complex transduction network will provide further opportunities to understand the mechanism of ovarian carcinogenesis, invasion and metastasis.
Collapse
Affiliation(s)
- Tarah L Pua
- Gynecologic Oncology, New York University School of Medicine, New York, NY, USA
| | | | | |
Collapse
|
39
|
Target genes suitable for silencing approaches and protein product interference in ovarian epithelial cancer. Cancer Treat Rev 2010; 36:8-15. [PMID: 19945796 DOI: 10.1016/j.ctrv.2009.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Accepted: 10/27/2009] [Indexed: 12/25/2022]
|
40
|
Targeting the epidermal growth factor receptor in epithelial ovarian cancer: current knowledge and future challenges. JOURNAL OF ONCOLOGY 2010; 2010:568938. [PMID: 20037743 PMCID: PMC2796463 DOI: 10.1155/2010/568938] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Accepted: 08/31/2009] [Indexed: 02/03/2023]
Abstract
The epidermal growth factor receptor is overexpressed in up to 60% of ovarian epithelial malignancies. EGFR regulates complex cellular events due to the large number of ligands, dimerization partners, and diverse signaling pathways engaged. In ovarian cancer, EGFR activation is associated with increased malignant tumor phenotype and poorer patient outcome. However, unlike some other EGFR-positive solid tumors, treatment of ovarian tumors with anti-EGFR agents has induced minimal response. While the amount of information regarding EGFR-mediated signaling is considerable, current data provides little insight for the lack of efficacy of anti-EGFR agents in ovarian cancer. More comprehensive, systematic, and well-defined approaches are needed to dissect the roles that EGFR plays in the complex signaling processes in ovarian cancer as well as to identify biomarkers that can accurately predict sensitivity toward EGFR-targeted therapeutic agents. This new knowledge could facilitate the development of rational combinatorial therapies to sensitize tumor cells toward EGFR-targeted therapies.
Collapse
|
41
|
G protein-coupled receptors stimulation and the control of cell migration. Cell Signal 2009; 21:1045-53. [DOI: 10.1016/j.cellsig.2009.02.008] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Revised: 02/03/2009] [Accepted: 02/17/2009] [Indexed: 01/14/2023]
|