1
|
Rai P, Clark CJ, Kardam V, Womack CB, Thammathong J, Norman DD, Tigyi GJ, Bicker K, Weissmiller AM, Dubey KD, Banerjee S. Structure-Based Discovery of MolPort-137: A Novel Autotaxin Inhibitor That Improves Paclitaxel Efficacy. Int J Mol Sci 2025; 26:597. [PMID: 39859312 PMCID: PMC11765394 DOI: 10.3390/ijms26020597] [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: 12/19/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The autotaxin-lysophosphatidic acid receptor (ATX-LPAR) signaling axis is pivotal in various clinical conditions, including cancer and autoimmune disorders. This axis promotes tumorigenicity by interacting with the tumor microenvironment, facilitating metastasis, and conceding antitumor immunity, thereby fostering resistance to conventional cancer therapies. Recent studies highlight the promise of ATX/LPAR inhibitors in combination with conventional chemotherapeutic drugs to overcome some forms of this resistance, representing a novel therapeutic strategy. In the current study, we employed structure-based virtual screening, integrating pharmacophore modeling and molecular docking, to identify MolPort-137 as a novel ATX inhibitor with an IC50 value of 1.6 ± 0.2 μM in an autotaxin enzyme inhibition assay. Molecular dynamics simulations and binding free energy calculations elucidated the binding mode of MolPort-137 and its critical amino acid interactions. Remarkably, MolPort-137 exhibited no cytotoxicity as a single agent but enhanced the effectiveness of paclitaxel in 4T1 murine breast carcinoma cells and resensitized taxol-resistant cells to paclitaxel treatment, which highlights its potential in combination therapy.
Collapse
Affiliation(s)
- Prateek Rai
- Molecular Biosciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA; (P.R.); (C.J.C.); (K.B.); (A.M.W.)
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - Christopher J. Clark
- Molecular Biosciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA; (P.R.); (C.J.C.); (K.B.); (A.M.W.)
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - Vandana Kardam
- Department of Chemistry, Shiv Nadar Institution of Eminence, Delhi 201314, India;
| | - Carl B. Womack
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - Joshua Thammathong
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - Derek D. Norman
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 37132, USA; (D.D.N.); (G.J.T.)
| | - Gábor J. Tigyi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 37132, USA; (D.D.N.); (G.J.T.)
| | - Kevin Bicker
- Molecular Biosciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA; (P.R.); (C.J.C.); (K.B.); (A.M.W.)
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - April M. Weissmiller
- Molecular Biosciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA; (P.R.); (C.J.C.); (K.B.); (A.M.W.)
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | | | - Souvik Banerjee
- Molecular Biosciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA; (P.R.); (C.J.C.); (K.B.); (A.M.W.)
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| |
Collapse
|
2
|
Schirizzi A, Donghia R, De Nunzio V, Renna N, Centonze M, De Leonardis G, Lorusso V, Fantasia A, Coletta S, Stabile D, Ferro A, Notarnicola M, Ricci AD, Lotesoriere C, Lahn M, D'Alessandro R, Giannelli G. High levels of autotaxin and lysophosphatidic acid predict poor outcome in treatment of resectable gastric carcinoma. Eur J Cancer 2024; 213:115066. [PMID: 39426076 DOI: 10.1016/j.ejca.2024.115066] [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: 07/30/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Although early-stage gastric cancer is a candidate for curative surgical resection, the absence of specific early symptoms results in a late diagnosis and consequently most patients present advanced or metastatic disease. Identifying noveland tumor-specific biomarkers is needed to increase early detection and match patients to the appropriate treatment. The present study focused on the possible prognostic role of Ectonucleotide Pyrophosphatase/Phosphodiesterase 2 (ENPP2)/Autotaxin (ATX) and lysophosphatidic acid (LPA) in Gastro-Esophageal Adenocarcinoma (GEA). High levels of ATX/LPA are associated with several malignancies including gastrointestinal tumors. METHODS Using a bioinformatics analysis, the incidence of ENPP2 mutations together with its expression in the tumor tissues and the correlation between the presence of mutations and the survival rate were examined in databases of GEA patients. Furthermore, circulating levels of ATX and LPA were studied retrospectively and longitudinally both in patients receiving frontal surgery and in patients receiving preoperative chemotherapy. RESULTS Overall findings suggested that although ENPP2 mutations occur at low incidence, their presence was associated with a particular poor Overall Survival (OS). Furthermore, removal of the tumour by surgery resulted in a decrease in serum ATX and LPA levels within five days, regardless of any previous chemotherapy. Basal circulating ATX were associated with the aggressive diffuse GEA and could be considered of negative prognostic value, mainly in combination models with circulating Carcino-Embryonic Antigen (CEA). CONCLUSIONS Based on these observations, clinical trials with ATX-targeted drugs and standard chemotherapy regimens may benefit from selecting GEA patients based on their levels of ATX, LPA and CEA.
Collapse
Affiliation(s)
- Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Rossella Donghia
- Data Science Unit, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Valentina De Nunzio
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Natasha Renna
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Matteo Centonze
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Giampiero De Leonardis
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Vincenza Lorusso
- Clinical Trial Unit, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Alessia Fantasia
- Clinical Trial Unit, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Sergio Coletta
- Core Facility Biobank, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Dolores Stabile
- Core Facility Biobank, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Annalisa Ferro
- Clinical Pathology Unit, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Maria Notarnicola
- Clinical Pathology Unit, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Angela D Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Michael Lahn
- iOnctura Clinical Research, Avenue Secheron 15, 1202 Geneva, Switzerland.
| | - Rosalba D'Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, 70013 Castellana Grotte, BA, Italy.
| |
Collapse
|
3
|
Akasaka H, Sano FK, Shihoya W, Nureki O. Structural mechanisms of potent lysophosphatidic acid receptor 1 activation by nonlipid basic agonists. Commun Biol 2024; 7:1444. [PMID: 39506093 PMCID: PMC11541586 DOI: 10.1038/s42003-024-07152-y] [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: 12/12/2023] [Accepted: 10/25/2024] [Indexed: 11/08/2024] Open
Abstract
Lysophosphatidic acid receptor 1 (LPA1) is one of the G protein-coupled receptors activated by the lipid mediator, lysophosphatidic acid (LPA). LPA1 is associated with a variety of diseases, and LPA1 agonists have potential therapeutic value for treating obesity and depression. Although potent nonlipid LPA1 agonists have recently been identified, the mechanisms of nonlipid molecule-mediated LPA1 activation remain unclear. Here, we report a cryo-electron microscopy structure of the human LPA1-Gi complex bound to a nonlipid basic agonist, CpY, which has 30-fold higher agonistic activity as compared with LPA. Structural comparisons of LPA1 with other lipid GPCRs revealed that the negative charge in the characteristic binding pocket of LPA1 allows the selective recognition of CpY, which lacks a polar head. In addition, our structure show that the ethyl group of CpY directly pushes W2716.48 to fix the active conformation. Endogenous LPA lacks these chemical features, which thus represent the crucial elements of nonlipid agonists that potently activate LPA1. This study provides detailed mechanistic insights into the ligand recognition and activation of LPA1 by nonlipid agonists, expanding the scope for drug development targeting the LPA receptors.
Collapse
Affiliation(s)
- Hiroaki Akasaka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Fumiya K Sano
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Wataru Shihoya
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| | - Osamu Nureki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
4
|
Yun CC, Han Y, McConnell B. Lysophosphatidic Acid Signaling in the Gastrointestinal System. Cell Mol Gastroenterol Hepatol 2024; 18:101398. [PMID: 39233124 PMCID: PMC11532463 DOI: 10.1016/j.jcmgh.2024.101398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/06/2024]
Abstract
The intestinal epithelium undergoes continuous homeostatic renewal to conduct the digestion and absorption of nutrients. At the same time, the intestinal epithelial barrier separates the host from the intestinal lumen, preventing systemic infection from enteric pathogens. To maintain homeostasis and epithelial functionality, stem cells, which reside in the base of intestinal crypts, generate progenitor cells that ultimately differentiate to produce an array of secretory and absorptive cells. Intestinal regeneration is regulated by niche signaling pathways, specifically, Wnt, bone morphogenetic protein, Notch, and epidermal growth factor. In addition, growth factors and other peptides have emerged as potential modulators of intestinal repair and inflammation through their roles in cellular proliferation, differentiation, migration, and survival. Lysophosphatidic acid (LPA) is such a factor that modulates the proliferation, survival, and migration of epithelial cells while also regulating trafficking of immune cells, both of which are important for tissue homeostasis. Perturbation of LPA signaling, however, has been shown to promote cancer and inflammation. This review focuses on the recent advances in LPA-mediated signaling that contribute to physiological and pathophysiological regulation of the gastrointestinal system.
Collapse
Affiliation(s)
- C Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia.
| | - Yiran Han
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Beth McConnell
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
5
|
Brandt N, Köper F, Hausmann J, Bräuer AU. Spotlight on plasticity-related genes: Current insights in health and disease. Pharmacol Ther 2024; 260:108687. [PMID: 38969308 DOI: 10.1016/j.pharmthera.2024.108687] [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: 02/08/2024] [Revised: 06/07/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
The development of the central nervous system is highly complex, involving numerous developmental processes that must take place with high spatial and temporal precision. This requires a series of complex and well-coordinated molecular processes that are tighly controlled and regulated by, for example, a variety of proteins and lipids. Deregulations in these processes, including genetic mutations, can lead to the most severe maldevelopments. The present review provides an overview of the protein family Plasticity-related genes (PRG1-5), including their role during neuronal differentiation, their molecular interactions, and their participation in various diseases. As these proteins can modulate the function of bioactive lipids, they are able to influence various cellular processes. Furthermore, they are dynamically regulated during development, thus playing an important role in the development and function of synapses. First studies, conducted not only in mouse experiments but also in humans, revealed that mutations or dysregulations of these proteins lead to changes in lipid metabolism, resulting in severe neurological deficits. In recent years, as more and more studies have shown their involvement in a broad range of diseases, the complexity and broad spectrum of known and as yet unknown interactions between PRGs, lipids, and proteins make them a promising and interesting group of potential novel therapeutic targets.
Collapse
Affiliation(s)
- Nicola Brandt
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Franziska Köper
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Jens Hausmann
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Anja U Bräuer
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany; Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
| |
Collapse
|
6
|
Laface C, Ricci AD, Vallarelli S, Ostuni C, Rizzo A, Ambrogio F, Centonze M, Schirizzi A, De Leonardis G, D’Alessandro R, Lotesoriere C, Giannelli G. Autotaxin-Lysophosphatidate Axis: Promoter of Cancer Development and Possible Therapeutic Implications. Int J Mol Sci 2024; 25:7737. [PMID: 39062979 PMCID: PMC11277072 DOI: 10.3390/ijms25147737] [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: 05/30/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Autotaxin (ATX) is a member of the ectonucleotide pyrophosphate/phosphodiesterase (ENPP) family; it is encoded by the ENPP2 gene. ATX is a secreted glycoprotein and catalyzes the hydrolysis of lysophosphatidylcholine to lysophosphatidic acid (LPA). LPA is responsible for the transduction of various signal pathways through the interaction with at least six G protein-coupled receptors, LPA Receptors 1 to 6 (LPAR1-6). The ATX-LPA axis is involved in various physiological and pathological processes, such as angiogenesis, embryonic development, inflammation, fibrosis, and obesity. However, significant research also reported its connection to carcinogenesis, immune escape, metastasis, tumor microenvironment, cancer stem cells, and therapeutic resistance. Moreover, several studies suggested ATX and LPA as relevant biomarkers and/or therapeutic targets. In this review of the literature, we aimed to deepen knowledge about the role of the ATX-LPA axis as a promoter of cancer development, progression and invasion, and therapeutic resistance. Finally, we explored its potential application as a prognostic/predictive biomarker and therapeutic target for tumor treatment.
Collapse
Affiliation(s)
- Carmelo Laface
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Simona Vallarelli
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Carmela Ostuni
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Francesca Ambrogio
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Matteo Centonze
- Personalized Medicine Laboratory, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, “IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.)
| | - Giampiero De Leonardis
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, “IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.)
| | - Rosalba D’Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, “IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.)
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| |
Collapse
|
7
|
Ren Y, Wang M, Yuan H, Wang Z, Yu L. A novel insight into cancer therapy: Lipid metabolism in tumor-associated macrophages. Int Immunopharmacol 2024; 135:112319. [PMID: 38801810 DOI: 10.1016/j.intimp.2024.112319] [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: 03/19/2024] [Revised: 05/07/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
The tumor immune microenvironment (TIME) can limit the effectiveness and often leads to significant side effects of conventional cancer therapies. Consequently, there is a growing interest in identifying novel targets to enhance the efficacy of targeted cancer therapy. More research indicates that tumor-associated macrophages (TAMs), originating from peripheral blood monocytes generated from bone marrow myeloid progenitor cells, play a crucial role in the tumor microenvironment (TME) and are closely associated with resistance to traditional cancer therapies. Lipid metabolism alterations have been widely recognized as having a significant impact on tumors and their immune microenvironment. Lipids, lipid derivatives, and key substances in their metabolic pathways can influence the carcinogenesis and progression of cancer cells by modulating the phenotype, function, and activity of TAMs. Therefore, this review focuses on the reprogramming of lipid metabolism in cancer cells and their immune microenvironment, in which the TAMs are especially concentrated. Such changes impact TAMs activation and polarization, thereby affecting the tumor cell response to treatment. Furthermore, the article explores the potential of targeting the lipid metabolism of TAMs as a supplementary approach to conventional cancer therapies. It reviews and evaluates current strategies for enhancing efficacy through TAMs' lipid metabolism and proposes new lipid metabolism targets as potential synergistic options for chemo-radiotherapy and immunotherapy. These efforts aim to stimulate further research in this area.
Collapse
Affiliation(s)
- Yvxiao Ren
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Mingjie Wang
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People's Republic of China
| | - Hanghang Yuan
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People's Republic of China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People's Republic of China
| | - Lei Yu
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China.
| |
Collapse
|
8
|
Dietze R, Szymanski W, Ojasalu K, Finkernagel F, Nist A, Stiewe T, Graumann J, Müller R. Phosphoproteomics Reveals Selective Regulation of Signaling Pathways by Lysophosphatidic Acid Species in Macrophages. Cells 2024; 13:810. [PMID: 38786034 PMCID: PMC11119170 DOI: 10.3390/cells13100810] [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: 04/10/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Lysophosphatidic acid (LPA) species, prevalent in the tumor microenvironment (TME), adversely impact various cancers. In ovarian cancer, the 18:0 and 20:4 LPA species are selectively associated with shorter relapse-free survival, indicating distinct effects on cellular signaling networks. Macrophages represent a cell type of high relevance in the TME, but the impact of LPA on these cells remains obscure. Here, we uncovered distinct LPA-species-specific responses in human monocyte-derived macrophages through unbiased phosphoproteomics, with 87 and 161 phosphosites upregulated by 20:4 and 18:0 LPA, respectively, and only 24 shared sites. Specificity was even more pronounced for downregulated phosphosites (163 versus 5 sites). Considering the high levels 20:4 LPA in the TME and its selective association with poor survival, this finding may hold significant implications. Pathway analysis pinpointed RHO/RAC1 GTPase signaling as the predominantly impacted target, including AHRGEF and DOCK guanine exchange factors, ARHGAP GTPase activating proteins, and regulatory protein kinases. Consistent with these findings, exposure to 20:4 resulted in strong alterations to the actin filament network and a consequent enhancement of macrophage migration. Moreover, 20:4 LPA induced p38 phosphorylation, a response not mirrored by 18:0 LPA, whereas the pattern for AKT was reversed. Furthermore, RNA profiling identified genes involved in cholesterol/lipid metabolism as selective targets of 20:4 LPA. These findings imply that the two LPA species cooperatively regulate different pathways to support functions essential for pro-tumorigenic macrophages within the TME. These include cellular survival via AKT activation and migration through RHO/RAC1 and p38 signaling.
Collapse
Affiliation(s)
- Raimund Dietze
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, 35043 Marburg, Germany; (R.D.); (K.O.); (F.F.)
| | - Witold Szymanski
- Institute of Translational Proteomics, Biochemical Pharmacological Centre, Philipps University, 35043 Marburg, Germany
- Core Facility Translational Proteomics, Philipps University, 35043 Marburg, Germany
| | - Kaire Ojasalu
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, 35043 Marburg, Germany; (R.D.); (K.O.); (F.F.)
| | - Florian Finkernagel
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, 35043 Marburg, Germany; (R.D.); (K.O.); (F.F.)
- Bioinformatics Core Facility, Philipps University, 35043 Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, 35043 Marburg, Germany; (A.N.); (T.S.)
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, 35043 Marburg, Germany; (A.N.); (T.S.)
| | - Johannes Graumann
- Institute of Translational Proteomics, Biochemical Pharmacological Centre, Philipps University, 35043 Marburg, Germany
- Core Facility Translational Proteomics, Philipps University, 35043 Marburg, Germany
| | - Rolf Müller
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, 35043 Marburg, Germany; (R.D.); (K.O.); (F.F.)
| |
Collapse
|
9
|
Yoon D, Choi B, Kim JE, Kim EY, Chung SH, Min HJ, Sung Y, Chang EJ, Song JK. Autotaxin inhibition attenuates the aortic valve calcification by suppressing inflammation-driven fibro-calcific remodeling of valvular interstitial cells. BMC Med 2024; 22:122. [PMID: 38486246 PMCID: PMC10941471 DOI: 10.1186/s12916-024-03342-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Patients with fibro-calcific aortic valve disease (FCAVD) have lipid depositions in their aortic valve that engender a proinflammatory impetus toward fibrosis and calcification and ultimately valve leaflet stenosis. Although the lipoprotein(a)-autotaxin (ATX)-lysophosphatidic acid axis has been suggested as a potential therapeutic target to prevent the development of FCAVD, supportive evidence using ATX inhibitors is lacking. We here evaluated the therapeutic potency of an ATX inhibitor to attenuate valvular calcification in the FCAVD animal models. METHODS ATX level and activity in healthy participants and patients with FCAVD were analyzed using a bioinformatics approach using the Gene Expression Omnibus datasets, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and western blotting. To evaluate the efficacy of ATX inhibitor, interleukin-1 receptor antagonist-deficient (Il1rn-/-) mice and cholesterol-enriched diet-induced rabbits were used as the FCAVD models, and primary human valvular interstitial cells (VICs) from patients with calcification were employed. RESULTS The global gene expression profiles of the aortic valve tissue of patients with severe FCAVD demonstrated that ATX gene expression was significantly upregulated and correlated with lipid retention (r = 0.96) or fibro-calcific remodeling-related genes (r = 0.77) in comparison to age-matched non-FCAVD controls. Orally available ATX inhibitor, BBT-877, markedly ameliorated the osteogenic differentiation and further mineralization of primary human VICs in vitro. Additionally, ATX inhibition significantly attenuated fibrosis-related factors' production, with a detectable reduction of osteogenesis-related factors, in human VICs. Mechanistically, ATX inhibitor prohibited fibrotic changes in human VICs via both canonical and non-canonical TGF-β signaling, and subsequent induction of CTGF, a key factor in tissue fibrosis. In the in vivo FCAVD model system, ATX inhibitor exposure markedly reduced calcific lesion formation in interleukin-1 receptor antagonist-deficient mice (Il1rn-/-, P = 0.0210). This inhibition ameliorated the rate of change in the aortic valve area (P = 0.0287) and mean pressure gradient (P = 0.0249) in the FCAVD rabbit model. Moreover, transaortic maximal velocity (Vmax) was diminished with ATX inhibitor administration (mean Vmax = 1.082) compared to vehicle control (mean Vmax = 1.508, P = 0.0221). Importantly, ATX inhibitor administration suppressed the effects of a high-cholesterol diet and vitamin D2-driven fibrosis, in association with a reduction in macrophage infiltration and calcific deposition, in the aortic valves of this rabbit model. CONCLUSIONS ATX inhibition attenuates the development of FCAVD while protecting against fibrosis and calcification in VICs, suggesting the potential of using ATX inhibitors to treat FCAVD.
Collapse
Affiliation(s)
- Dohee Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Bongkun Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Ji-Eun Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Young Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Soo-Hyun Chung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyo-Jin Min
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yoolim Sung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Jae-Kwan Song
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
10
|
Simonetti J, Ficili M, Sgalla G, Richeldi L. Experimental autotaxin inhibitors for the treatment of idiopathic pulmonary fibrosis. Expert Opin Investig Drugs 2024; 33:133-143. [PMID: 38299617 DOI: 10.1080/13543784.2024.2305126] [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: 10/23/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
INTRODUCTION Idiopathic Pulmonary Fibrosis (IPF) is a progressive, irreversible, and fatal lung disease with unmet medical needs. Autotaxin (ATX) is an extracellular enzyme involved in the generation of lysophosphatidic acid (LPA). Preclinical and clinical data have suggested the ATX-LPAR signaling axis plays an important role in the pathogenesis and the progression of IPF. AREAS COVERED The aim of this review is to provide an update on the available evidence on autotaxin inhibitors in IPF and further details on the ongoing clinical studies involving these molecules. EXPERT OPINION The development of autotaxin inhibitors as a potential therapy for idiopathic pulmonary fibrosis has gained attention due to evidence of their involvement in the disease. Preclinical and early-phase clinical studies have explored these inhibitors' efficacy and safety, offering a novel approach in treating this disease. Combining autotaxin inhibitors with existing anti-fibrotic agents is considered for enhanced therapeutic effects. Large phase III trials assessed Ziritaxestat but yielded disappointing results, highlighting the importance of long-term observation and clinical outcomes in clinical research. Patient stratification and personalized medicine are crucial, as pulmonary fibrosis is a heterogeneous disease. Ongoing research and collaboration are essential for this advancement.
Collapse
Affiliation(s)
- Jacopo Simonetti
- Unita Operativa Complessa di Pneumologia, Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Unita Operativa Complessa di Pneumologia, Dipartimento di Neuroscienze, Organi di Senso e Torace, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Ficili
- Unita Operativa Complessa di Pneumologia, Dipartimento di Neuroscienze, Organi di Senso e Torace, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giacomo Sgalla
- Unita Operativa Complessa di Pneumologia, Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Unita Operativa Complessa di Pneumologia, Dipartimento di Neuroscienze, Organi di Senso e Torace, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Richeldi
- Unita Operativa Complessa di Pneumologia, Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Unita Operativa Complessa di Pneumologia, Dipartimento di Neuroscienze, Organi di Senso e Torace, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
11
|
Toyohara T, Yoshida M, Miyabe K, Hayashi K, Naitoh I, Kondo H, Hori Y, Kato A, Kachi K, Asano G, Sahashi H, Adachi A, Kuno K, Kito Y, Matsuo Y, Kataoka H. Dual role of autotaxin as novel biomarker and therapeutic target in pancreatic neuroendocrine neoplasms. Cancer Sci 2023; 114:4571-4582. [PMID: 37770812 PMCID: PMC10728022 DOI: 10.1111/cas.15980] [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: 06/15/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Pancreatic neuroendocrine neoplasms (panNENs) are rare pancreatic neoplasms, and descriptions of treatment remain limited. Autotaxin (ATX) is a secreted autocrine motility factor involved in the production of lysophosphatidic acid (LPA), a lipid mediator that promotes the progression of various cancers. The aim of this study was to clarify the importance of the ATX-LPA axis in panNENs and to confirm its contribution to panNEN progression using clinical data, cell lines, and a mouse model. Serum ATX level was higher in patients with panNEN than in patients with other pancreatic diseases (chronic pancreatitis, pancreatic ductal adenocarcinoma [PDAC], intraductal papillary mucinous neoplasm, autoimmune pancreatitis) and healthy controls, and 61% of clinical specimens stained strongly for ATX. In a case we encountered, serum ATX level fluctuated with disease progression. An in vitro study showed higher ATX mRNA expression in panNEN cell lines than in PDAC cell lines. Cell proliferation and migration in panNEN cell lines were stimulated via the ATX-LPA axis and suppressed by RNA interference or inhibitors. An in vivo study showed that intraperitoneal injection of GLPG1690, an ATX inhibitor, suppressed tumor progression in a xenograft model. These findings revealed that ATX expression is significantly elevated in panNEN and is related to the progression of panNEN. We showed the potential of ATX as a novel biomarker and therapeutic target.
Collapse
Affiliation(s)
- Tadashi Toyohara
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Michihiro Yoshida
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Katsuyuki Miyabe
- Department of GastroenterologyJapanese Red Cross Aichi Medical Center Nagoya Daini HospitalNagoyaJapan
| | - Kazuki Hayashi
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Itaru Naitoh
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Hiromu Kondo
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Yasuki Hori
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Akihisa Kato
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Kenta Kachi
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Go Asano
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Hidenori Sahashi
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Akihisa Adachi
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Kayoko Kuno
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Yusuke Kito
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Yoichi Matsuo
- Department of Gastroenterological SurgeryNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Hiromi Kataoka
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoyaJapan
| |
Collapse
|
12
|
Meng J, Ruan X, Wei F, Xue Q. High expression of ENPP2 is an independent predictor of poor prognosis in liver cancer. Medicine (Baltimore) 2023; 102:e34480. [PMID: 37543832 PMCID: PMC10402965 DOI: 10.1097/md.0000000000034480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/07/2023] Open
Abstract
Ectonucleotide pyrophosphatase/phosphodiesterase 2 (ENPP2) has been identified as a potential biomarker in lung and prostate cancers; however, its expression and clinical relevance in hepatocellular carcinoma (HCC) remain incompletely understood. This study comprehensively assessed ENPP2 expression in pan-cancer using bioinformatics. We analyzed the expression of ENPP2 mRNA in primary liver cancer and adjacent tissues of patients with HCC using data from the TCGA database. Cox regression and Kaplan-Meier methods were used to identify clinicopathological factors associated with survival, and the diagnostic value of ENPP2 expression was evaluated using receiver operating characteristic curve analysis. We also validated our findings by performing real-time PCR on clinical liver cancer samples. Furthermore, we conducted gene set enrichment analysis using the Cancer Genome Atlas dataset to gain additional insights into the biological significance of ENPP2 in HCC. High ENPP2 expression in HCC patients is associated with gender and clinical stage, and is a significant prognostic factor for worse outcomes. ENPP2 expression is an independent risk factor for progression-free and disease-specific survival in both cohorts, suggesting its potential as an HCC biomarker. ENPP2's diagnostic value in HCC patients was confirmed by the area under the receiver operating characteristic curve, which was 0.806. real-time PCR analysis validated the higher expression of ENPP2 in clinical liver cancer tissues. Gene set enrichment analysis identified pathways enriched in HCC patients with high ENPP2 expression, including those related to the cell cycle, MTOR and T cell receptor signaling, and phosphatidylinositol signaling systems. We have demonstrated that ENPP2 is highly expressed in HCC and is a potential independent molecular marker for the diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- Jiyu Meng
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | | | | | | |
Collapse
|
13
|
Qiu H, Ni C, Jia C, Rong X, Chu M, Wu R, Han B. CircRNA7632 down-regulation alleviates endothelial cell dysfunction in Kawasaki disease via regulating IL-33 expression. Cell Stress Chaperones 2023; 28:363-374. [PMID: 37166618 PMCID: PMC10352195 DOI: 10.1007/s12192-023-01333-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 05/12/2023] Open
Abstract
Kawasaki disease (KD) is a form of idiopathic vasculitis frequently accompanied by coronary artery lesions, which involves endothelial dysfunction. Recent studies have demonstrated that circular RNAs (circRNAs) are implicated in many cardiovascular diseases. However, few studies have examined the role of circRNAs on endothelial dysfunction in KD. In this study, we investigated the role of circ7632 on endothelial-mesenchymal transition (EndoMT) in KD and then explored the underlying mechanism. Children diagnosed with KD and age-matched healthy controls (HC) were included. Sera samples were collected. Primary human umbilical vein endothelial cells (HUVECs) were obtained and incubated with 15% HC and KD serum for 48 h. The mRNA and protein expression of mesenchymal markers vimentin and α-smooth muscle actin (α-SMA) and endothelial marker zonula occludens-1 (ZO-1) in HUVECs transfected with plasmid-circ7632 and si-circ7632 were detected by RT-qPCR and Western blot analysis. CCK8, scratch test, and migration test were performed to examine the effect of circ7632 on the cell proliferation and migration. The circ7632 level was higher in HUVECs treated by KD serum than in HUVECs treated with HC serum. Overexpression of circ7632 significantly increased vimentin and α-SMA expression, decreased ZO-1 expression, and also decreased cell proliferation. Down-regulation of circ7632 expression got the opposite results. RNA-seq analysis, and confirmatory experiment displayed that down-regulation of circ7632 decreased IL-33 expression, and IL-33 silencing mitigated KD serum-mediated EndoMT. Our study revealed that circ7632 level was elevated in KD serum-treated HUVECs. Circ7632 down-regulation could alleviate EndoMT likely through decreasing IL-33 expression. The circ7632 may become a potential therapeutic target for KD.
Collapse
Affiliation(s)
- Huixian Qiu
- Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chao Ni
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chang Jia
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xing Rong
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Maoping Chu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Rongzhou Wu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Bo Han
- Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China.
| |
Collapse
|
14
|
Gomez-Larrauri A, Gangoiti P, Camacho L, Presa N, Martin C, Gomez-Muñoz A. Phosphatidic Acid Stimulates Lung Cancer Cell Migration through Interaction with the LPA1 Receptor and Subsequent Activation of MAP Kinases and STAT3. Biomedicines 2023; 11:1804. [PMID: 37509443 PMCID: PMC10376810 DOI: 10.3390/biomedicines11071804] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Phosphatidic acid (PA) is a key bioactive glycerophospholipid that is implicated in the regulation of vital cell functions such as cell growth, differentiation, and migration, and is involved in a variety of pathologic processes. However, the molecular mechanisms by which PA exerts its pathophysiological actions are incompletely understood. In the present work, we demonstrate that PA stimulates the migration of the human non-small cell lung cancer (NSCLC) A549 adenocarcinoma cells, as determined by the transwell migration assay. PA induced the rapid phosphorylation of mitogen-activated protein kinases (MAPKs) ERK1-2, p38, and JNK, and the pretreatment of cells with selective inhibitors of these kinases blocked the PA-stimulated migration of cancer cells. In addition, the chemotactic effect of PA was inhibited by preincubating the cells with pertussis toxin (PTX), a Gi protein inhibitor, suggesting the implication of a Gi protein-coupled receptor in this action. Noteworthy, a blockade of LPA receptor 1 (LPA1) with the specific LPA1 antagonist AM966, or with the selective LPA1 inhibitors Ki1645 or VPC32193, abolished PA-stimulated cell migration. Moreover, PA stimulated the phosphorylation of the transcription factor STAT3 downstream of JAK2, and inhibitors of either JAK2 or STAT3 blocked PA-stimulated cell migration. It can be concluded that PA stimulates lung adenocarcinoma cell migration through an interaction with the LPA1 receptor and subsequent activation of the MAPKs ERK1-2, p38, and JNK, and that the JAK2/STAT3 pathway is also important in this process. These findings suggest that targeting PA formation and/or the LPA1 receptor may provide new strategies to reduce malignancy in lung cancer.
Collapse
Affiliation(s)
- Ana Gomez-Larrauri
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48980 Bilbao, Bizkaia, Spain
- Respiratory Department, Cruces University Hospital, 48903 Barakaldo, Bizkaia, Spain
| | - Patricia Gangoiti
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48980 Bilbao, Bizkaia, Spain
| | - Laura Camacho
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48980 Bilbao, Bizkaia, Spain
| | - Natalia Presa
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48980 Bilbao, Bizkaia, Spain
| | - Cesar Martin
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48980 Bilbao, Bizkaia, Spain
- Department of Molecular Biophysics, Biofisika Institute, University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC), 48940 Leioa, Bizkaia, Spain
| | - Antonio Gomez-Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48980 Bilbao, Bizkaia, Spain
| |
Collapse
|
15
|
Méaux MN, Regnier M, Portefaix A, Borel O, Alioli C, Peyruchaud O, Legrand M, Bacchetta J. Circulating autotaxin levels in healthy teenagers: Data from the Vitados cohort. Front Pediatr 2023; 11:1094705. [PMID: 36861069 PMCID: PMC9969100 DOI: 10.3389/fped.2023.1094705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/16/2023] [Indexed: 02/17/2023] Open
Abstract
Autotaxin (ATX) is a secreted enzyme with a lysophospholipase D activity, mainly secreted by adipocytes and widely expressed. Its major function is to convert lysophosphatidylcholine (LPC) into lysophosphatidic acid (LPA), an essential bioactive lipid involved in multiple cell processes. The ATX-LPA axis is increasingly studied because of its involvement in numerous pathological conditions, more specifically in inflammatory or neoplastic diseases, and in obesity. Circulating ATX levels gradually increase with the stage of some pathologies, such as liver fibrosis, thus making them a potentially interesting non-invasive marker for fibrosis estimation. Normal circulating levels of ATX have been established in healthy adults, but no data exist at the pediatric age. The aim of our study is to describe the physiological concentrations of circulating ATX levels in healthy teenagers through a secondary analysis of the VITADOS cohort. Our study included 38 teenagers of Caucasian origin (12 males, 26 females). Their median age was 13 years for males and 14 years for females, ranging from Tanner 1 to 5. BMI was at the 25th percentile for males and 54th percentile for females, and median blood pressure was normal. ATX median levels were 1,049 (450-2201) ng/ml. There was no difference in ATX levels between sexes in teenagers, which was in contrast to the male and female differences described in the adult population. ATX levels significantly decreased with age and pubertal status, reaching adult levels at the end of puberty. Our study also suggested positive correlations between ATX levels and blood pressure (BP), lipid metabolism, and bone biomarkers. However, except for LDL cholesterol, these factors were also significantly correlated with age, which might be a confounding factor. Still, a correlation between ATX and diastolic BP was described in obese adult patients. No correlation was found between ATX levels and inflammatory marker C-reactive protein (CRP), Body Mass Index (BMI), and biomarkers of phosphate/calcium metabolism. In conclusion, our study is the first to describe the decline in ATX levels with puberty and the physiological concentrations of ATX levels in healthy teenagers. It will be of utmost importance when performing clinical studies in children with chronic diseases to keep these kinetics in mind, as circulating ATX might become a non-invasive prognostic biomarker in pediatric chronic diseases.
Collapse
Affiliation(s)
- Marie-Noëlle Méaux
- INSERM, UMR 1033, Lyon, France.,Centre de Référence des Maladies Rares du Calcium et du Phosphate, filière OSCAR, Lyon, France.,Service de Néphrologie, Rhumatologie et Dermatologie Pédiatriques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Maitena Regnier
- INSERM, UMR 1033, Lyon, France.,Centre de Référence des Maladies Rares du Calcium et du Phosphate, filière OSCAR, Lyon, France.,Service de Néphrologie, Rhumatologie et Dermatologie Pédiatriques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Aurélie Portefaix
- Centre d'Investigation Clinique, CIC 1407, Hospices Civils de Lyon, Bron, France
| | | | | | | | - Mélanie Legrand
- INSERM, UMR 1033, Lyon, France.,Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Lyon, France.,Service de Rhumatologie, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Justine Bacchetta
- INSERM, UMR 1033, Lyon, France.,Centre de Référence des Maladies Rares du Calcium et du Phosphate, filière OSCAR, Lyon, France.,Service de Néphrologie, Rhumatologie et Dermatologie Pédiatriques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France.,Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
16
|
Pan-cancer functional analysis of somatic mutations in G protein-coupled receptors. Sci Rep 2022; 12:21534. [PMID: 36513718 PMCID: PMC9747925 DOI: 10.1038/s41598-022-25323-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
G Protein-coupled receptors (GPCRs) are the most frequently exploited drug target family, moreover they are often found mutated in cancer. Here we used a dataset of mutations found in patient samples derived from the Genomic Data Commons and compared it to the natural human variance as exemplified by data from the 1000 genomes project. We explored cancer-related mutation patterns in all GPCR classes combined and individually. While the location of the mutations across the protein domains did not differ significantly in the two datasets, a mutation enrichment in cancer patients was observed among class-specific conserved motifs in GPCRs such as the Class A "DRY" motif. A Two-Entropy Analysis confirmed the correlation between residue conservation and cancer-related mutation frequency. We subsequently created a ranking of high scoring GPCRs, using a multi-objective approach (Pareto Front Ranking). Our approach was confirmed by re-discovery of established cancer targets such as the LPA and mGlu receptor families, but also discovered novel GPCRs which had not been linked to cancer before such as the P2Y Receptor 10 (P2RY10). Overall, this study presents a list of GPCRs that are amenable to experimental follow up to elucidate their role in cancer.
Collapse
|
17
|
Drosouni A, Panagopoulou M, Aidinis V, Chatzaki E. Autotaxin in Breast Cancer: Role, Epigenetic Regulation and Clinical Implications. Cancers (Basel) 2022; 14:5437. [PMID: 36358855 PMCID: PMC9658281 DOI: 10.3390/cancers14215437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 08/02/2023] Open
Abstract
Autotaxin (ATX), the protein product of Ectonucleotide Pyrophosphatase Phosphodiesterase 2 (ENPP2), is a secreted lysophospholipase D (lysoPLD) responsible for the extracellular production of lysophosphatidic acid (LPA). ATX-LPA pathway signaling participates in several normal biological functions, but it has also been connected to cancer progression, metastasis and inflammatory processes. Significant research has established a role in breast cancer and it has been suggested as a therapeutic target and/or a clinically relevant biomarker. Recently, ENPP2 methylation was described, revealing a potential for clinical exploitation in liquid biopsy. The current review aims to gather the latest findings about aberrant signaling through ATX-LPA in breast cancer and discusses the role of ENPP2 expression and epigenetic modification, giving insights with translational value.
Collapse
Affiliation(s)
- Andrianna Drosouni
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Maria Panagopoulou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Institute of Agri-Food and Life Sciences, Hellenic Mediterranean University Research Centre, 71410 Heraklion, Greece
| | - Vassilis Aidinis
- Institute of BioInnovation, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece
| | - Ekaterini Chatzaki
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Institute of Agri-Food and Life Sciences, Hellenic Mediterranean University Research Centre, 71410 Heraklion, Greece
| |
Collapse
|
18
|
Akasaka H, Tanaka T, Sano FK, Matsuzaki Y, Shihoya W, Nureki O. Structure of the active Gi-coupled human lysophosphatidic acid receptor 1 complexed with a potent agonist. Nat Commun 2022; 13:5417. [PMID: 36109516 PMCID: PMC9477835 DOI: 10.1038/s41467-022-33121-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Lysophosphatidic acid receptor 1 (LPA1) is one of the six G protein-coupled receptors activated by the bioactive lipid, lysophosphatidic acid (LPA). LPA1 is a drug target for various diseases, including cancer, inflammation, and neuropathic pain. Notably, LPA1 agonists have potential therapeutic value for obesity and urinary incontinence. Here, we report a cryo-electron microscopy structure of the active human LPA1-Gi complex bound to ONO-0740556, an LPA analog with more potent activity against LPA1. Our structure elucidated the details of the agonist binding mode and receptor activation mechanism mediated by rearrangements of transmembrane segment 7 and the central hydrophobic core. A structural comparison of LPA1 and other phylogenetically-related lipid-sensing GPCRs identified the structural determinants for lipid preference of LPA1. Moreover, we characterized the structural polymorphisms at the receptor-G-protein interface, which potentially reflect the G-protein dissociation process. Our study provides insights into the detailed mechanism of LPA1 binding to agonists and paves the way toward the design of drug-like agonists targeting LPA1. LPA1 is one of the GPCRs that are drug targets for various diseases. Here the authors report a cryo-EM structure of the active human LPA1-Gi complex bound to an LPA analog with more potent activity against LPA1 and clarified the ligand recognition mechanism.
Collapse
|
19
|
Uranbileg B, Kurano M, Kano K, Sakai E, Arita J, Hasegawa K, Nishikawa T, Ishihara S, Yamashita H, Seto Y, Ikeda H, Aoki J, Yatomi Y. Sphingosine 1-phosphate lyase facilitates cancer progression through converting sphingolipids to glycerophospholipids. Clin Transl Med 2022; 12:e1056. [PMID: 36125914 PMCID: PMC9488530 DOI: 10.1002/ctm2.1056] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND In addition to potent agonist properties for sphingosine 1-phosphate (S1P) receptors, intracellularly, S1P is an intermediate in metabolic conversion pathway from sphingolipids to glycerolysophospholipids (glyceroLPLs). We hypothesized that this S1P metabolism and its products might possess some novel roles in the pathogenesis of cancer, where S1P lyase (SPL) is a key enzyme. METHODS The mRNA levels of sphingolipid-related and other cancer-related factors were measured in human hepatocellular carcinoma (HCC), colorectal cancer, and esophageal cancer patients' tumours and in their adjacent non-tumour tissues. Phospholipids (PL) and glyceroLPLs were measured by using liquid chromatography-tandem mass spectrometry (LC-MS/MS). In-vitro experiments were performed in Colon 26 cell line with modulation of the SPL and GPR55 expressions. Xenograft model was used for determination of the cancer progression and for pharmacological influence. RESULTS Besides high SPL levels in human HCC and colon cancer, SPL levels were specifically and positively linked with levels of glyceroLPLs, including lysophosphatidylinositol (LPI). Overexpression of SPL in Colon 26 cells resulted in elevated levels of LPI and lysophosphatidylglycerol (LPG), which are agonists of GPR55. SPL overexpression-enhanced cell proliferation was inhibited by GPR55 silencing. Conversely, inhibition of SPL led to the opposite outcome and reversed by adding LPI, LPG, and metabolites generated during S1P degradation, which is regulated by SPL. The xenograft model results suggested the contribution of SPL and glyceroLPLs to tumour progression depending on levels of SPL and GPR55. Moreover, the pharmacological inhibition of SPL prevented the progression of cancer. The underlying mechanisms for the SPL-mediated cancer progression are the activation of p38 and mitochondrial function through the LPI, LPG-GPR55 axis and the suppression of autophagy in a GPR55-independent manner. CONCLUSION A new metabolic pathway has been proposed here in HCC and colon cancer, SPL converts S1P to glyceroLPLs, mainly to LPI and LPG, and facilitates cancer development.
Collapse
Affiliation(s)
- Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Eri Sakai
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junichi Arita
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nishikawa
- Surgical Oncology and Vascular Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Soichiro Ishihara
- Surgical Oncology and Vascular Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Hiroharu Yamashita
- Gastrointestinal Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan.,Division of Digestive Surgery, Department of Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuyuki Seto
- Gastrointestinal Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Hitoshi Ikeda
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
20
|
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: 0] [Impact Index Per Article: 0] [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
|
21
|
Li T, Lei H, Yang J, Cao Z, Yang Y, Liu Z, Sun R, Yang X, Zhai X. Hybrid imidazo[1,2‐
a
]pyridine analogs as potent ATX inhibitors with concrete in vivo antifibrosis effect. Arch Pharm (Weinheim) 2022; 355:e2200171. [DOI: 10.1002/ardp.202200171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Tong Li
- Key Laboratory of Structure‐Based Drug Design and Discovery, Ministry of Education School of Pharmaceutical Engineering, Shenyang Pharmaceutical University Shenyang People's Republic of China
| | - Hongrui Lei
- Key Laboratory of Structure‐Based Drug Design and Discovery, Ministry of Education School of Pharmaceutical Engineering, Shenyang Pharmaceutical University Shenyang People's Republic of China
| | - Juanjuan Yang
- Key Laboratory of Structure‐Based Drug Design and Discovery, Ministry of Education School of Pharmaceutical Engineering, Shenyang Pharmaceutical University Shenyang People's Republic of China
| | - Zhi Cao
- Key Laboratory of Structure‐Based Drug Design and Discovery, Ministry of Education School of Pharmaceutical Engineering, Shenyang Pharmaceutical University Shenyang People's Republic of China
| | - Yu Yang
- Key Laboratory of Structure‐Based Drug Design and Discovery, Ministry of Education School of Pharmaceutical Engineering, Shenyang Pharmaceutical University Shenyang People's Republic of China
| | - Zimeng Liu
- Key Laboratory of Structure‐Based Drug Design and Discovery, Ministry of Education School of Pharmaceutical Engineering, Shenyang Pharmaceutical University Shenyang People's Republic of China
| | - Ruonan Sun
- Key Laboratory of Structure‐Based Drug Design and Discovery, Ministry of Education School of Pharmaceutical Engineering, Shenyang Pharmaceutical University Shenyang People's Republic of China
| | - Xinlian Yang
- Key Laboratory of Structure‐Based Drug Design and Discovery, Ministry of Education School of Pharmaceutical Engineering, Shenyang Pharmaceutical University Shenyang People's Republic of China
| | - Xin Zhai
- Key Laboratory of Structure‐Based Drug Design and Discovery, Ministry of Education School of Pharmaceutical Engineering, Shenyang Pharmaceutical University Shenyang People's Republic of China
| |
Collapse
|
22
|
Using GPCRs as Molecular Beacons to Target Ovarian Cancer with Nanomedicines. Cancers (Basel) 2022; 14:cancers14102362. [PMID: 35625966 PMCID: PMC9140059 DOI: 10.3390/cancers14102362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
The five-year survival rate for women with ovarian cancer is very poor despite radical cytoreductive surgery and chemotherapy. Although most patients initially respond to platinum-based chemotherapy, the majority experience recurrence and ultimately develop chemoresistance, resulting in fatal outcomes. The current administration of cytotoxic compounds is hampered by dose-limiting severe adverse effects. There is an unmet clinical need for targeted drug delivery systems that transport chemotherapeutics selectively to tumor cells while minimizing off-target toxicity. G protein-coupled receptors (GPCRs) are the largest family of membrane receptors, and many are overexpressed in solid tumors, including ovarian cancer. This review summarizes the progress in engineered nanoparticle research for drug delivery for ovarian cancer and discusses the potential use of GPCRs as molecular entry points to deliver anti-cancer compounds into ovarian cancer cells. A newly emerging treatment paradigm could be the personalized design of nanomedicines on a case-by-case basis.
Collapse
|
23
|
Han L, Seward C, Leone G, Ostrowski MC. Origin, activation and heterogeneity of fibroblasts associated with pancreas and breast cancers. Adv Cancer Res 2022; 154:169-201. [PMID: 35459469 DOI: 10.1016/bs.acr.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Pancreas and breast cancers both contain abundant stromal components within the tumor tissues. A prominent cell type within the stroma is cancer-associated fibroblasts (CAFs). CAFs play critical and complex roles establishing the tumor microenvironment to either promote or prevent tumor progression. Recently, complex genetic models and single cell-based techniques have provided emerging insights on the precise functions and cellular heterogeneity of CAFs. The transformation of normal fibroblasts into CAFs is a key event during tumor initiation and progression. Such coordination between tumor cells and fibroblasts plays an important role in cancer development. Reprograming fibroblasts is currently being explored for therapeutic benefits. In this review, we will discuss recent literature shedding light on the tissues of origin, activation mechanisms, and heterogeneity of CAFs comparing pancreas and breast cancers.
Collapse
Affiliation(s)
- Lu Han
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States.
| | - Cara Seward
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Gustavo Leone
- Department of Biochemistry, Medical College of Wisconsin Cancer Center, Medical college of Wisconsin, Milwaukee, WI, United States
| | - Michael C Ostrowski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
24
|
Liao Y, Liu L, Yang J, Shi Z. ATX/LPA axis regulates FAK activation, cell proliferation, apoptosis, and motility in human pancreatic cancer cells. In Vitro Cell Dev Biol Anim 2022; 58:307-315. [PMID: 35426066 DOI: 10.1007/s11626-022-00660-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 02/14/2022] [Indexed: 11/29/2022]
Abstract
Previous studies implicated ATX/LPA axis as a potential driver of tumorigenesis and progression in pancreatic cancer. This study aimed to determine the existence of the autocrine pathway of ATX/LPA action in pancreatic cancer cells, and to elucidate its influence on focal adhesion kinase (FAK) activation, cellular proliferation, apoptosis, and migration. Firstly, we identified the lysophosphatidic acid (LPA) concentrations in cultured cell supernatant by ELISA and observed the effect of the autotaxin (ATX)-specific inhibitor S32826 on LPA concentrations. We found the existence of a certain concentration of LPA in cellular supernatant, which was significantly decreased by S32826 in a dose- and time-dependent manner. A maximum response was observed at 50 μM for 72 h. Secondly, the effect of S32826 on the protein expression and intracellular sublocalization of total FAK and phosphorylated FAK (pY397 FAK) was determined by Western blot and immunofluorescence staining. It was found that the expression of total FAK and pY397 FAK and their distribution along the cell membrane where adhesion structures are located were significantly decreased by S32826. Finally, we observed the influence of S32826 on cell proliferation, apoptosis, and migration by CCK-8 assay, flow cytometric analysis, and transwell migration assay. Results showed that cell viability and migration were significantly declined, and the proportions of apoptotic cells were significantly increased by S32826. This study verified the existence of autocrine regulation of LPA secretion via producing ATX by pancreatic cancer cells in vitro and the important role of LPA/ATX axis on FAK activation, cell proliferation, apoptosis, and motility.
Collapse
Affiliation(s)
- Yan Liao
- Department of Gastroenterology, Wuhan No.1 Hospital (Wuhan Integrated TCM and Western Medicine Hospital), No. 215, Zhongshan Road, Wuhan, 430022, Hubei, China
| | - Lei Liu
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiayao Yang
- Department of Gastroenterology, Wuhan No.1 Hospital (Wuhan Integrated TCM and Western Medicine Hospital), No. 215, Zhongshan Road, Wuhan, 430022, Hubei, China
| | - Zhaohong Shi
- Department of Gastroenterology, Wuhan No.1 Hospital (Wuhan Integrated TCM and Western Medicine Hospital), No. 215, Zhongshan Road, Wuhan, 430022, Hubei, China.
| |
Collapse
|
25
|
Hoshino Y, Okuno T, Saigusa D, Kano K, Yamamoto S, Shindou H, Aoki J, Uchida K, Yokomizo T, Ito N. Lysophosphatidic acid receptor 1/3 antagonist inhibits the activation of satellite glial cells and reduces acute nociceptive responses. FASEB J 2022; 36:e22236. [PMID: 35218596 DOI: 10.1096/fj.202101678r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Lysophosphatidic acid (LPA) exerts various biological activities through six characterized G protein-coupled receptors (LPA1-6 ). While LPA-LPA1 signaling contributes toward the demyelination and retraction of C-fiber and induces neuropathic pain, the effects of LPA-LPA1 signaling on acute nociceptive pain is uncertain. This study investigated the role of LPA-LPA1 signaling in acute nociceptive pain using the formalin test. The pharmacological inhibition of the LPA-LPA1 axis significantly attenuated formalin-induced nociceptive behavior. The LPA1 mRNA was expressed in satellite glial cells (SGCs) in dorsal root ganglion (DRG) and was particularly abundant in SGCs surrounding large DRG neurons, which express neurofilament 200. Treatment with LPA1/3 receptor (LPA1/3 ) antagonist inhibited the upregulation of glial markers and inflammatory cytokines in DRG following formalin injection. The LPA1/3 antagonist also attenuated phosphorylation of extracellular signal-regulated kinase, especially in SGCs and cyclic AMP response element-binding protein in the dorsal horn following formalin injection. LPA amounts after formalin injection to the footpad were quantified by liquid chromatography/tandem mass spectrometry, and LPA levels were found to be increased in the innervated DRGs. Our results indicate that LPA produced in the innervated DRGs promotes the activation of SGCs through LPA1 , increases the sensitivity of primary neurons, and modulates pain behavior. These results facilitate our understanding of the pathology of acute nociceptive pain and demonstrate the possibility of the LPA1 on SGCs as a novel target for acute pain control.
Collapse
Affiliation(s)
- Yoko Hoshino
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Okuno
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan.,Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shota Yamamoto
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan.,Department of Lipid Medical Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kanji Uchida
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuko Ito
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
26
|
Wang W, Ma Y, He T, Mooney E, Guo C, Wang XY, Fang X. Histopathological Diagnosis of Nonalcoholic Steatohepatitis (NASH). Methods Mol Biol 2022; 2455:49-62. [PMID: 35212985 DOI: 10.1007/978-1-0716-2128-8_5] [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] [Indexed: 06/14/2023]
Abstract
Fatty acid beta oxidation (FAO) is a predominant bioenergetic pathway in mammals. Substantial investigations have demonstrated that FAO activity is dysregulated in many pathophysiological conditions including nonalcoholic steatohepatitis (NASH). Convenient and quantitative assays of FAO activities are important for studies of cell metabolism and the biological relevance of FAO to health and diseases. However, most current FAO assays are based on non-physiological culture conditions, measure FAO activity indirectly or lack adequate quantification. We herein describe details of practical protocols for measurement of basal and genetically or pharmacologically regulated FAO activities in the mammalian system. We also discuss the advantages and disadvantages of these assays in the context of experimental purposes.
Collapse
Affiliation(s)
- Wei Wang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Yibao Ma
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
- Alliance Pharma Inc, Malvern, PA, USA
| | - Tianhai He
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Erin Mooney
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Xianjun Fang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA.
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
27
|
Deng W, Chen F, Zhou Z, Huang Y, Lin J, Zhang F, Xiao G, Liu C, Liu C, Xu L. Hepatitis B Virus Promotes Hepatocellular Carcinoma Progression Synergistically With Hepatic Stellate Cells via Facilitating the Expression and Secretion of ENPP2. Front Mol Biosci 2021; 8:745990. [PMID: 34805271 PMCID: PMC8602366 DOI: 10.3389/fmolb.2021.745990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/13/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Hepatitis B virus (HBV) infection is a major risk factor causing hepatocellular carcinoma (HCC) development, but the molecular mechanisms are not fully elucidated. It has been reported that virus infection induces ectonucleotide pyrophosphatase-phosphodiesterase 2 (ENPP2) expression, the latter participates in tumor progression. Therefore, the aim of the present study was to investigate whether HBV induced HCC malignancy via ENPP2. Methods: HCC patient clinical data were collected and prognosis was analyzed. Transient transfection and stable ectopic expression of the HBV genome were established in hepatoma cell lines. Immunohistochemical staining, RT-qPCR, western blot, and ELISA assays were used to detect the expression and secretion of ENPP2. Finally, CCK-8, colony formation, and migration assays as well as a subcutaneous xenograft mouse model were used to investigate the influence of HBV infection, ENPP2 expression, and activated hepatic stellate cells (aHSCs) on HCC progression in vitro and in vivo. Results: The data from cancer databases indicated that the level of ENPP2 was significant higher in HCC compared within normal liver tissues. Clinical relevance analysis using 158 HCC patients displayed that ENPP2 expression was positively correlated with poor overall survival and disease-free survival. Statistical analysis revealed that compared to HBV-negative HCC tissues, HBV-positive tissues expressed a higher level of ENPP2. In vitro, HBV upregulated ENPP2 expression and secretion in hepatoma cells and promoted hepatoma cell proliferation, colony formation, and migration via enhancement of ENPP2; downregulation of ENPP2 expression or inhibition of its function suppressed HCC progression. In addition, aHSCs strengthened hepatoma cell proliferation, migration in vitro, and promoted tumorigenesis synergistically with HBV in vivo; a loss-function assay further verified that ENPP2 is essential for HBV/aHSC-induced HCC progression. Conclusion: HBV enhanced the expression and secretion of ENPP2 in hepatoma cells, combined with aHSCs to promote HCC progression via ENPP2.
Collapse
Affiliation(s)
- Wanyu Deng
- Department of Biliary Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,College of Life Science, Shangrao Normal University, Shangrao, China
| | - Fu Chen
- College of Life Science, Shangrao Normal University, Shangrao, China
| | - Ziyu Zhou
- Department of Biliary Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yipei Huang
- Department of Biliary Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junlong Lin
- Department of Biliary Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fapeng Zhang
- Department of Biliary Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Gang Xiao
- Department of Biliary Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chaoqun Liu
- Department of Biliary Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chao Liu
- Department of Biliary Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Leibo Xu
- Department of Biliary Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Gao P, Liu Q, Ai B, Fang Y, Wang Z, Wang J. Prognostic Value and Clinical Significance of LIPH in Breast Cancer. Int J Gen Med 2021; 14:7613-7623. [PMID: 34754232 PMCID: PMC8572048 DOI: 10.2147/ijgm.s332233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/26/2021] [Indexed: 01/23/2023] Open
Abstract
Background Lipase member H (LIPH), a novel member of the mammalian triglyceride lipase family, is localized on human chromosome 3q27-q28. Exploration of the importance of the new cancer-related gene LIPH in several carcinomas has been reported in previous studies. Our study aims to systematically assess the expression pattern of LIPH in breast cancer. Methods Our study explored 2994 breast cancer samples with transcriptome data from the Cancer Genome Atlas (TCGA) and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets. We systematically evaluated the mRNA expression of LIPH in breast cancer and the overall survival (OS) of patients. The protein expression of LIPH in breast cancer was evaluated with the Human Protein Atlas. We also explored the relationship between LIPH and the immune microenvironment in pan-cancer. Results Both mRNA and protein expression LIPH were found to be upregulated in breast cancer tumors. The overall survival rate of patients with high LIPH expression was lower than those of patients with low LIPH expression in both the TCGA dataset (p=0.0067) and METABRIC dataset (p<0.0001). Outcomes of the multivariate analysis found that the level of LIPH expression was an independent prognostic factor in both TCGA (p=0.001) and METABRIC (p=0.019) databases. The outcomes of the univariate analysis showed that LIPH was an important prognostic factor (p=0.01 in TCGA dataset, p=0.001 in METABRIC dataset). In the TCGA dataset, outcomes showed that LIPH expression was negatively correlated with the AJCC (American Joint Committee on Cancer) stage (p=2.3e-05) and triple-negative breast cancer (TNBC) tissues (p=3.1e-10). High LIPH expression showed lower OS in the TNBC subtype (P=0.011). Conclusion Compared to normal tissues, the expression of LIPH was higher in breast cancer tissues in both mRNA and protein levels. This study showed that the high level of LIPH expression might be related to the worse prognosis of breast cancer.
Collapse
Affiliation(s)
- Peng Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Qiang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Bolun Ai
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| |
Collapse
|
29
|
Autotaxin May Have Lysophosphatidic Acid-Unrelated Effects on Three-Dimension (3D) Cultured Human Trabecular Meshwork (HTM) Cells. Int J Mol Sci 2021; 22:ijms222112039. [PMID: 34769470 PMCID: PMC8584821 DOI: 10.3390/ijms222112039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 01/25/2023] Open
Abstract
PURPOSE The objective of the current study was to evaluate the effects of the autotaxin (ATX)-lysophosphatidic acid (LPA) signaling axis on the human trabecular meshwork (HTM) in two-dimensional (2D) and three-dimensional (3D) cultures of HTM cells. METHODS The effects were characterized by transendothelial electrical resistance (TEER) and FITC-dextran permeability (2D), measurements of size and stiffness (3D), and the expression of several genes, including extracellular matrix (ECM) molecules, their modulators, and endoplasmic reticulum (ER) stress-related factors. RESULTS A one-day exposure to 200 nM LPA induced significant down-sizing effects of the 3D HTM spheroids, and these effects were enhanced slightly on longer exposure. The TEER and FITC-dextran permeability data indicate that LPA induced an increase in the barrier function of the 2D HTM monolayers. A one-day exposure to a 2 mg/L solution of ATX also resulted in a significant decrease in the sizes of the 3D HTM spheroids, and an increase in stiffness was also observed. The gene expression of several ECMs, their regulators and ER-stress related factors by the 3D HTM spheroids were altered by both ATX and LPA, but in different manners. CONCLUSIONS The findings presented herein suggest that ATX may have additional roles in the human TM, in addition to the ATX-LPA signaling axis.
Collapse
|
30
|
Design, synthesis and anti-fibrosis evaluation of imidazo[1,2-a]pyridine derivatives as potent ATX inhibitors. Bioorg Med Chem 2021; 46:116362. [PMID: 34428714 DOI: 10.1016/j.bmc.2021.116362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 11/23/2022]
Abstract
A series of imidazo[1,2-a]pyridine compounds bearing urea moiety (8-27) were designed, synthesized and evaluated for their ATX inhibitory activities in vitro by FS-3 based enzymatic assay. Delightfully, benzylamine derivatives (14-27) exhibited higher ATX inhibitory potency with IC50 value ranging from 1.72 to 497 nM superior to benzamide analogues (8-13). Remarkably, benzylamine derivative 20 bearing 4-hydroxypiperidine exerted an amazing inhibitory activity (IC50 = 1.72 nM) which exceeded the positive control GLPG1690 (IC50 = 2.90 nM). Simultaneously, the binding model of 20 with ATX was established which rationalized the well performance of 20 in enzymatic assay. Accordingly, further in vivo studies were carried out to evaluate direct anti-fibrotic effects of 20 through Masson staining. Notably, 20 effectively alleviated lung structural damage with fewer fibrotic lesions at an oral dose of 60 mg/kg, qualifying 20 as a promising ATX inhibitor for IPF treatment.
Collapse
|
31
|
Tserendavga B, Ohshima N, Fujita C, Yuzawa K, Ohshima M, Yanaka N, Minamishima YA, Izumi T. Characterization of recombinant murine GDE4 and GDE7, enzymes producing lysophosphatidic acid and/or cyclic phosphatidic acid. J Biochem 2021; 170:713-727. [PMID: 34523685 DOI: 10.1093/jb/mvab091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
GDE4 and GDE7 are membrane-bound enzymes that exhibit lysophospholipase D (lysoPLD) activities. We found that GDE7 produced not only lysophosphatidic acid (LPA) but also cyclic phosphatidic acid (cPA) from lysophospholipids by a transphosphatidylation reaction. In contrast, GDE4 produced only LPA. The analysis of substrate specificity showed that 1-alkyl-lysophosphospholipids were preferred substrates for both enzymes rather than 1-alkyl-lysophospholipids and 1-alkenyl-lysophospholipids. Among the various lysophospholipids with different polar head groups that were tested, lysophosphatidylglycerol and lysophosphatidylserine were preferred substrates for GDE4 and GDE7, respectively. The detailed analysis of the dependency of the enzyme activities of GDE4 and GDE7 on divalent cations suggested multiple divalent cations were bound in the active sites of both enzymes. Taken together, these results suggest the possibility that GDE7 functions as a cPA-producing enzyme in the body.
Collapse
Affiliation(s)
- Binderiya Tserendavga
- Department of Biochemistry, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Noriyasu Ohshima
- Department of Biochemistry, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Chiaki Fujita
- Department of Biochemistry, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Koji Yuzawa
- Group of Pharmaceutical Analysis, ENVIRONMENTAL TECHNICAL CO., LTD, Takasaki, Gunma 370-3511, Japan
| | - Mari Ohshima
- Department of Biochemistry, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.,Group of Pharmaceutical Analysis, ENVIRONMENTAL TECHNICAL CO., LTD, Takasaki, Gunma 370-3511, Japan
| | - Noriyuki Yanaka
- Department of Molecular and Applied Bioscience, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Yoji Andrew Minamishima
- Department of Biochemistry, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Takashi Izumi
- Department of Biochemistry, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.,Faculty of Health Care, Teikyo Heisei University, Tokyo, 170-8445, Japan
| |
Collapse
|
32
|
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: 20] [Impact Index Per Article: 5.0] [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
|
33
|
Gremski LH, Matsubara FH, Polli NLC, Antunes BC, Schluga PHDC, da Justa HC, Minozzo JC, Wille ACM, Senff-Ribeiro A, Veiga SS. Prospective Use of Brown Spider Venom Toxins as Therapeutic and Biotechnological Inputs. Front Mol Biosci 2021; 8:706704. [PMID: 34222343 PMCID: PMC8247472 DOI: 10.3389/fmolb.2021.706704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/04/2021] [Indexed: 11/20/2022] Open
Abstract
Brown spider (genus Loxosceles) venoms are mainly composed of protein toxins used for predation and defense. Bites of these spiders most commonly produce a local dermonecrotic lesion with gravitational spread, edema and hemorrhage, which together are defined as cutaneous loxoscelism. Systemic loxoscelism, such as hematological abnormalities and renal injury, are less frequent but more lethal. Some Loxosceles venom toxins have already been isolated and extensively studied, such as phospholipases D (PLDs), which have been recombinantly expressed and were proven to reproduce toxic activities associated to the whole venom. PLDs have a notable potential to be engineered and converted in non-toxic antigens to produce a new generation of antivenoms or vaccines. PLDs also can serve as tools to discover inhibitors to be used as therapeutic agents. Other Loxosceles toxins have been identified and functionally characterized, such as hyaluronidases, allergen factor, serpin, TCTP and knottins (ICK peptides). All these toxins were produced as recombinant molecules and are biologically active molecules that can be used as tools for the potential development of chemical candidates to tackle many medical and biological threats, acting, for instance, as antitumoral, insecticides, analgesic, antigens for allergy tests and biochemical reagents for cell studies. In addition, these recombinant toxins may be useful to develop a rational therapy for loxoscelism. This review summarizes the main candidates for the development of drugs and biotechnological inputs that have been described in Brown spider venoms.
Collapse
Affiliation(s)
| | | | | | - Bruno Cesar Antunes
- Department of Cell Biology, Federal University of Paraná, Curitiba, Brazil.,Production and Research Center of Immunobiological Products, State Department of Health, Piraquara, Brazil
| | | | | | - João Carlos Minozzo
- Production and Research Center of Immunobiological Products, State Department of Health, Piraquara, Brazil
| | - Ana Carolina Martins Wille
- Department of Structural, Molecular Biology and Genetics, State University of Ponta Grossa, Ponta Grossa, Brazil
| | | | | |
Collapse
|
34
|
Arang N, Gutkind JS. G Protein-Coupled receptors and heterotrimeric G proteins as cancer drivers. FEBS Lett 2021; 594:4201-4232. [PMID: 33270228 DOI: 10.1002/1873-3468.14017] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/09/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptors (GPCRs) and heterotrimeric G proteins play central roles in a diverse array of cellular processes. As such, dysregulation of GPCRs and their coupled heterotrimeric G proteins can dramatically alter the signalling landscape and functional state of a cell. Consistent with their fundamental physiological functions, GPCRs and their effector heterotrimeric G proteins are implicated in some of the most prevalent human diseases, including a complex disease such as cancer that causes significant morbidity and mortality worldwide. GPCR/G protein-mediated signalling impacts oncogenesis at multiple levels by regulating tumour angiogenesis, immune evasion, metastasis, and drug resistance. Here, we summarize the growing body of research on GPCRs and their effector heterotrimeric G proteins as drivers of cancer initiation and progression, and as emerging antitumoural therapeutic targets.
Collapse
Affiliation(s)
- Nadia Arang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - J Silvio Gutkind
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
35
|
O'Regan A, O'Brien CJ, Eivers SB. The lysophosphatidic acid axis in fibrosis: Implications for glaucoma. Wound Repair Regen 2021; 29:613-626. [PMID: 34009724 DOI: 10.1111/wrr.12929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/13/2021] [Accepted: 04/28/2021] [Indexed: 12/27/2022]
Abstract
Glaucoma is a common progressive optic neuropathy that results in visual field defects and can lead to irreversible blindness. The pathophysiology of glaucoma involves dysregulated extracellular matrix remodelling in both the trabecular meshwork in the anterior chamber and in the lamina cribrosa of the optic nerve head. Fibrosis in these regions leads to raised intraocular pressure and retinal ganglion cell degeneration, respectively. Lysophosphatidic acid (LPA) is a bioactive lipid mediator which acts via six G-protein coupled receptors on the cell surface to activate intracellular pathways that promote cell proliferation, transcription and survival. LPA signalling has been implicated in both normal wound healing and pathological fibrosis. LPA enhances fibroblast proliferation, migration and contraction, and induces expression of pro-fibrotic mediators such as connective tissue growth factor. The LPA axis plays a major role in diseases such as idiopathic pulmonary fibrosis, where it has been identified as an important pharmacological target. In glaucoma, LPA is present in high levels in the aqueous humour, and its signalling has been found to increase resistance to aqueous humour outflow through altered trabecular meshwork cellular contraction and extracellular matrix deposition. LPA signalling may, therefore, also represent an attractive target for treatment of glaucoma. In this review we wish to describe the role of LPA and its related proteins in tissue fibrosis and glaucoma.
Collapse
Affiliation(s)
- Amy O'Regan
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Colm J O'Brien
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, Dublin, Ireland.,Department of Ophthalmology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Sarah B Eivers
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, Dublin, Ireland
| |
Collapse
|
36
|
Control of Intestinal Epithelial Permeability by Lysophosphatidic Acid Receptor 5. Cell Mol Gastroenterol Hepatol 2021; 12:1073-1092. [PMID: 33975030 PMCID: PMC8350072 DOI: 10.1016/j.jcmgh.2021.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/10/2022]
Abstract
BACKGROUND & AIMS Epithelial cells form a monolayer at mucosal surface that functions as a highly selective barrier. Lysophosphatidic acid (LPA) is a bioactive lipid that elicits a broad range of biological effects via cognate G protein-coupled receptors. LPA receptor 5 (LPA5) is highly expressed in intestinal epithelial cells, but its role in the intestine is not well-known. Here we determined the role of LPA5 in regulation of intestinal epithelial barrier. METHODS Epithelial barrier integrity was determined in mice with intestinal epithelial cell (IEC)-specific LPA5 deletion, Lpar5ΔIEC. LPA was orally administered to mice, and intestinal permeability was measured. Dextran sulfate sodium (DSS) was used to induce colitis. Human colonic epithelial cell lines were used to determine the LPA5-mediated signaling pathways that regulate epithelial barrier. RESULTS We observed increased epithelial permeability in Lpar5ΔIEC mice with reduced claudin-4 expression. Oral administration of LPA decreased intestinal permeability in wild-type mice, but the effect was greatly mitigated in Lpar5ΔIEC mice. Serum lipopolysaccharide level and bacterial loads in the intestine and liver were elevated in Lpar5ΔIEC mice. Lpar5ΔIEC mice developed more severe colitis induced with DSS. LPA5 transcriptionally regulated claudin-4, and this regulation was dependent on transactivation of the epidermal growth factor receptor, which induced localization of Rac1 at the cell membrane. LPA induced the translocation of Stat3 to the cell membrane and promoted the interaction between Rac1 and Stat3. Inhibition of Stat3 ablated LPA-mediated regulation of claudin-4. CONCLUSIONS This study identifies LPA5 as a regulator of the intestinal barrier. LPA5 promotes claudin-4 expression in IECs through activation of Rac1 and Stat3.
Collapse
|
37
|
Zhang X, Li M, Yin N, Zhang J. The Expression Regulation and Biological Function of Autotaxin. Cells 2021; 10:cells10040939. [PMID: 33921676 PMCID: PMC8073485 DOI: 10.3390/cells10040939] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
Autotaxin (ATX) is a secreted glycoprotein and functions as a key enzyme to produce extracellular lysophosphatidic acid (LPA). LPA interacts with at least six G protein-coupled receptors, LPAR1-6, on the cell membrane to activate various signal transduction pathways through distinct G proteins, such as Gi/0, G12/13, Gq/11, and Gs. The ATX-LPA axis plays an important role in physiological and pathological processes, including embryogenesis, obesity, and inflammation. ATX is one of the top 40 most unregulated genes in metastatic cancer, and the ATX-LPA axis is involved in the development of different types of cancers, such as colorectal cancer, ovarian cancer, breast cancer, and glioblastoma. ATX expression is under multifaceted controls at the transcription, post-transcription, and secretion levels. ATX and LPA in the tumor microenvironment not only promote cell proliferation, migration, and survival, but also increase the expression of inflammation-related circuits, which results in poor outcomes for patients with cancer. Currently, ATX is regarded as a potential cancer therapeutic target, and an increasing number of ATX inhibitors have been developed. In this review, we focus on the mechanism of ATX expression regulation and the functions of ATX in cancer development.
Collapse
Affiliation(s)
| | | | | | - Junjie Zhang
- Correspondence: ; Tel.: +86-10-58802137; Fax: +86-10-58807720
| |
Collapse
|
38
|
Lysophospholipids in Lung Inflammatory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:373-391. [PMID: 33788203 DOI: 10.1007/978-3-030-63046-1_20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The lysophospholipids (LPLs) belong to a group of bioactive lipids that play pivotal roles in several physiological and pathological processes. LPLs are derivatives of phospholipids and consist of a single hydrophobic fatty acid chain, a hydrophilic head, and a phosphate group with or without a large molecule attached. Among the LPLs, lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are the simplest, and have been shown to be involved in lung inflammatory symptoms and diseases such as acute lung injury, asthma, and chronic obstructive pulmonary diseases. G protein-coupled receptors (GPCRs) mediate LPA and S1P signaling. In this chapter, we will discuss on the role of LPA, S1P, their metabolizing enzymes, inhibitors or agonists of their receptors, and their GPCR-mediated signaling in lung inflammatory symptoms and diseases, focusing specially on acute respiratory distress syndrome, asthma, and chronic obstructive pulmonary disease.
Collapse
|
39
|
Li X, Nakayama K, Goto T, Akamatsu S, Kobayashi T, Shimizu K, Ogawa O, Inoue T. A narrative review of urinary phospholipids: from biochemical aspect towards clinical application. Transl Androl Urol 2021; 10:1829-1849. [PMID: 33968673 PMCID: PMC8100843 DOI: 10.21037/tau-20-1263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
As a newly emerged discipline, lipidomic studies have focused on the comprehensive characterization and quantification of lipids in a given biological system, which has remarkably advanced in recent years owing to the rapid development of analytical techniques, especially mass spectrometry. Among diverse lipid classes, phospholipids, which have fundamental roles in the formation of cellular membranes, signaling processes, and bioenergetics have gained momentum in several fields of research. The altered composition, concentration, spatial distribution, and metabolism of phospholipids in cells, tissues, and body fluids have been elucidated in various human diseases such as cancer, inflammation, as well as cardiovascular and metabolic disorders. Among the different kinds of phospholipid sources in the human body, urine has not been extensively investigated in recent years owing to the extremely low concentrations of phospholipids and high levels of salts and other contaminants, which can interfere with precise detection. However, with profound advances and rapid expansion in analytical methods, urinary phospholipids have attracted increasing attention in current biomedical research as urine is an easily available source for the discovery of noninvasive biomarkers. In this review, we provide an overview of urinary phospholipids, including their biochemical aspects and clinical applications, aimed at promoting this field of research.
Collapse
Affiliation(s)
- Xin Li
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Nakayama
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Goto
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shusuke Akamatsu
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Kobayashi
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Shimizu
- Clinical Research Center for Medical Equipment Development, Kyoto University Hospital, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Inoue
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
40
|
Yang G, Liu S, Maghsoudloo M, Shasaltaneh MD, Kaboli PJ, Zhang C, Deng Y, Heidari H, Entezari M, Fu S, Wen Q, Imani S. PLA1A expression as a diagnostic marker of BRAF-mutant metastasis in melanoma cancer. Sci Rep 2021; 11:6056. [PMID: 33723350 PMCID: PMC7961027 DOI: 10.1038/s41598-021-85595-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/02/2021] [Indexed: 01/31/2023] Open
Abstract
BRAF and NRAS are the most reported mutations associated to melanomagenesis. The lack of accurate diagnostic markers in response to therapeutic treatment in BRAF/NRAS-driven melanomagenesis is one of the main challenges in melanoma personalized therapy. In order to assess the diagnostic value of phosphatidylserine-specific phospholipase A1-alpha (PLA1A), a potent lysophospholipid mediating the production of lysophosphatidylserine, PLA1A mRNA and serum levels were compared in subjects with malignant melanoma (n = 18), primary melanoma (n = 13), and healthy subjects (n = 10). Additionally, the correlation between histopathological subtypes of BRAF/NRAS-mutated melanoma and PLA1A was analyzed. PLA1A expression was significantly increased during melanogenesis and positively correlated to disease severity and histopathological markers of metastatic melanoma. PLA1A mRNA and serum levels were significantly higher in patients with BRAF-mutated melanoma compared to the patients with NRAS-mutated melanoma. Notably, PLA1A can be used as a diagnostic marker for an efficient discrimination between naïve melanoma samples and advanced melanoma samples (sensitivity 91%, specificity 57%, and AUC 0.99), as well as BRAF-mutated melanoma samples (sensitivity 62%, specificity 61%, and AUC 0.75). Our findings suggest that PLA1A can be considered as a potential diagnostic marker for advanced and BRAF-mutated melanoma.
Collapse
Affiliation(s)
- Gang Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Oncology, Anyue Hospital of Traditional Chinese Medicine, Second Ziyang Hospital of Traditional Chinese Medicine, Ziyang, Sichuan, China
| | - Shuya Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Mazaher Maghsoudloo
- Laboratory of Systems Biology and Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Parham Jabbarzadeh Kaboli
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Cuiwei Zhang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Youcai Deng
- Institute of Materia Medical, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
| | - Hajar Heidari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - ShaoZhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - QingLian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Saber Imani
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
41
|
Hu HB, Song ZQ, Song GP, Li S, Tu HQ, Wu M, Zhang YC, Yuan JF, Li TT, Li PY, Xu YL, Shen XL, Han QY, Li AL, Zhou T, Chun J, Zhang XM, Li HY. LPA signaling acts as a cell-extrinsic mechanism to initiate cilia disassembly and promote neurogenesis. Nat Commun 2021; 12:662. [PMID: 33510165 PMCID: PMC7843646 DOI: 10.1038/s41467-021-20986-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 12/15/2020] [Indexed: 01/17/2023] Open
Abstract
Dynamic assembly and disassembly of primary cilia controls embryonic development and tissue homeostasis. Dysregulation of ciliogenesis causes human developmental diseases termed ciliopathies. Cell-intrinsic regulatory mechanisms of cilia disassembly have been well-studied. The extracellular cues controlling cilia disassembly remain elusive, however. Here, we show that lysophosphatidic acid (LPA), a multifunctional bioactive phospholipid, acts as a physiological extracellular factor to initiate cilia disassembly and promote neurogenesis. Through systematic analysis of serum components, we identify a small molecular-LPA as the major driver of cilia disassembly. Genetic inactivation and pharmacological inhibition of LPA receptor 1 (LPAR1) abrogate cilia disassembly triggered by serum. The LPA-LPAR-G-protein pathway promotes the transcription and phosphorylation of cilia disassembly factors-Aurora A, through activating the transcription coactivators YAP/TAZ and calcium/CaM pathway, respectively. Deletion of Lpar1 in mice causes abnormally elongated cilia and decreased proliferation in neural progenitor cells, thereby resulting in defective neurogenesis. Collectively, our findings establish LPA as a physiological initiator of cilia disassembly and suggest targeting the metabolism of LPA and the LPA pathway as potential therapies for diseases with dysfunctional ciliogenesis.
Collapse
Affiliation(s)
- Huai-Bin Hu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Zeng-Qing Song
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Guang-Ping Song
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Sen Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Hai-Qing Tu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Min Wu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Cheng Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Jin-Feng Yuan
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Ting-Ting Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Pei-Yao Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Ling Xu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Lin Shen
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Qiu-Ying Han
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Ai-Ling Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, USA
| | - Xue-Min Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China.
| | - Hui-Yan Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China.
- School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
42
|
Kim B, Hebert JM, Liu D, Auguste DT. A Lipid Targeting, pH‐Responsive Nanoemulsion Encapsulating a DNA Intercalating Agent and HDAC Inhibitor Reduces TNBC Tumor Burden. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Bumjun Kim
- Department of Chemical Engineering Northeastern University 360 Huntington Avenue Boston MA 02115 USA
- Department of Chemical and Biological Engineering Princeton University 50‐70 Olden St Princeton NJ 08540 USA
| | - Jacob M. Hebert
- Department of Chemical Engineering Northeastern University 360 Huntington Avenue Boston MA 02115 USA
| | - Daxing Liu
- Department of Chemical Engineering Northeastern University 360 Huntington Avenue Boston MA 02115 USA
- Department of Radiology Stony Brook University 100 Nicolls Rd, Stony Brook New York NY 11790 USA
| | - Debra T. Auguste
- Department of Chemical Engineering Northeastern University 360 Huntington Avenue Boston MA 02115 USA
| |
Collapse
|
43
|
Gui X, Li Y, Zhang X, Su K, Cao W. Circ_LDLR promoted the development of papillary thyroid carcinoma via regulating miR-195-5p/LIPH axis. Cancer Cell Int 2020; 20:241. [PMID: 32549788 PMCID: PMC7296738 DOI: 10.1186/s12935-020-01327-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/07/2020] [Indexed: 01/16/2023] Open
Abstract
Background Emerging studies have demonstrated that circular RNAs (circRNAs) are key regulators for tumorigenesis in cancers, including papillary thyroid carcinoma (PTC). In this study, we aimed to explore the effects of circ_LDLR on PTC. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to determine the levels of circ_LDLR, miR-195-5p and lipase H (LIPH). RNase R digestion assay and Actinomycin D assay were utilized to analyze the characteristics of circ_LDLR. Colony formation assay and 3-(4,5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay were conducted to evaluate cell proliferation. Western blot assay was used for the determination of protein levels. Flow cytometry analysis was applied to determine cell apoptosis. Transwell assay was performed to determine cell migration and invasion. Dual-luciferase reporter assay was used to verify the associations among circ_LDLR, miR-195-5p and LIPH. The murine xenograft model was constructed to explore the roles of circ_LDLR in vivo. Results Compared to normal tissues and cells, circ_LDLR was upregulated in PTC tissues and cells. Silencing of circ_LDLR suppressed PTC cell colony formation, proliferation, migration and invasion and promoted apoptosis in vitro and hampered tumor growth in vivo. For mechanism investigation, circ_LDLR could regulate LIPH expression via sponging miR-195-5p. Moreover, miR-195-5p inhibition restored the effects of circ_LDLR knockdown on the malignant behaviors of PTC cells. MiR-195-5p overexpression inhibited PTC cell colony formation, proliferation, migration and invasion and facilitated apoptosis by targeting LIPH. Conclusion Circ_LDLR knockdown decelerated PTC progression by regulating miR-195-5p/LIPH axis, which might provide a novel therapeutic target for PTC.
Collapse
Affiliation(s)
- Xiaolong Gui
- Department of Gastrointestinal & Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021 Guangxi China
| | - Yan Li
- Department of Pharmacy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Xiaobin Zhang
- Department of Gastrointestinal & Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021 Guangxi China
| | - Ka Su
- Department of Gastrointestinal & Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021 Guangxi China
| | - Wenlong Cao
- Department of Gastrointestinal & Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021 Guangxi China
| |
Collapse
|
44
|
Lei H, Guo M, Li X, Jia F, Li C, Yang Y, Cao M, Jiang N, Ma E, Zhai X. Discovery of Novel Indole-Based Allosteric Highly Potent ATX Inhibitors with Great In Vivo Efficacy in a Mouse Lung Fibrosis Model. J Med Chem 2020; 63:7326-7346. [DOI: 10.1021/acs.jmedchem.0c00506] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Hongrui Lei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ming Guo
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaopeng Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fang Jia
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Changtao Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Yang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Meng Cao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Nan Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Enlong Ma
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
45
|
Feng Y, Mischler WJ, Gurung AC, Kavanagh TR, Androsov G, Sadow PM, Herbert ZT, Priolo C. Therapeutic Targeting of the Secreted Lysophospholipase D Autotaxin Suppresses Tuberous Sclerosis Complex-Associated Tumorigenesis. Cancer Res 2020; 80:2751-2763. [PMID: 32393662 DOI: 10.1158/0008-5472.can-19-2884] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/25/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disease characterized by multiorgan hamartomas, including renal angiomyolipomas and pulmonary lymphangioleiomyomatosis (LAM). TSC2 deficiency leads to hyperactivation of mTOR Complex 1 (mTORC1), a master regulator of cell growth and metabolism. Phospholipid metabolism is dysregulated upon TSC2 loss, causing enhanced production of lysophosphatidylcholine (LPC) species by TSC2-deficient tumor cells. LPC is the major substrate of the secreted lysophospholipase D autotaxin (ATX), which generates two bioactive lipids, lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P). We report here that ATX expression is upregulated in human renal angiomyolipoma-derived TSC2-deficient cells compared with TSC2 add-back cells. Inhibition of ATX via the clinically developed compound GLPG1690 suppressed TSC2-loss associated oncogenicity in vitro and in vivo and induced apoptosis in TSC2-deficient cells. GLPG1690 suppressed AKT and ERK1/2 signaling and profoundly impacted the transcriptome of these cells while inducing minor gene expression changes in TSC2 add-back cells. RNA-sequencing studies revealed transcriptomic signatures of LPA and S1P, suggesting an LPA/S1P-mediated reprogramming of the TSC lipidome. In addition, supplementation of LPA or S1P rescued proliferation and viability, neutral lipid content, and AKT or ERK1/2 signaling in human TSC2-deficient cells treated with GLPG1690. Importantly, TSC-associated renal angiomyolipomas have higher expression of LPA receptor 1 and S1P receptor 3 compared with normal kidney. These studies increase our understanding of TSC2-deficient cell metabolism, leading to novel potential therapeutic opportunities for TSC and LAM. SIGNIFICANCE: This study identifies activation of the ATX-LPA/S1P pathway as a novel mode of metabolic dysregulation upon TSC2 loss, highlighting critical roles for ATX in TSC2-deficient cell fitness and in TSC tumorigenesis.
Collapse
Affiliation(s)
- You Feng
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - William J Mischler
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Ashish C Gurung
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Taylor R Kavanagh
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Grigoriy Androsov
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Peter M Sadow
- Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Zachary T Herbert
- Harvard Medical School, Boston, Massachusetts
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Carmen Priolo
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
46
|
Regulation of Tumor Immunity by Lysophosphatidic Acid. Cancers (Basel) 2020; 12:cancers12051202. [PMID: 32397679 PMCID: PMC7281403 DOI: 10.3390/cancers12051202] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/16/2022] Open
Abstract
The tumor microenvironment (TME) may be best conceptualized as an ecosystem comprised of cancer cells interacting with a multitude of stromal components such as the extracellular matrix (ECM), blood and lymphatic networks, fibroblasts, adipocytes, and cells of the immune system. At the center of this crosstalk between cancer cells and their TME is the bioactive lipid lysophosphatidic acid (LPA). High levels of LPA and the enzyme generating it, termed autotaxin (ATX), are present in many cancers. It is also well documented that LPA drives tumor progression by promoting angiogenesis, proliferation, survival, invasion and metastasis. One of the hallmarks of cancer is the ability to modulate and escape immune detection and eradication. Despite the profound role of LPA in regulating immune functions and inflammation, its role in the context of tumor immunity has not received much attention until recently where emerging studies highlight that this signaling axis may be a means that cancer cells adopt to evade immune detection and eradication. The present review aims to look at the immunomodulatory actions of LPA in baseline immunity to provide a broad understanding of the subject with a special emphasis on LPA and cancer immunity, highlighting the latest progress in this area of research.
Collapse
|
47
|
Esposito F, Boccarelli A, Del Buono N. An NMF-Based Methodology for Selecting Biomarkers in the Landscape of Genes of Heterogeneous Cancer-Associated Fibroblast Populations. Bioinform Biol Insights 2020; 14:1177932220906827. [PMID: 32425511 PMCID: PMC7218276 DOI: 10.1177/1177932220906827] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 01/22/2020] [Indexed: 01/27/2023] Open
Abstract
The rapid development of high-performance technologies has greatly promoted studies of molecular oncology producing large amounts of data. Even if these data are publicly available, they need to be processed and studied to extract information useful to better understand mechanisms of pathogenesis of complex diseases, such as tumors. In this article, we illustrated a procedure for mining biologically meaningful biomarkers from microarray datasets of different tumor histotypes. The proposed methodology allows to automatically identify a subset of potentially informative genes from microarray data matrices, which differs either in the number of rows (genes) and of columns (patients). The methodology integrates nonnegative matrix factorization method, a functional enrichment analysis web tool with a properly designed gene extraction procedure to allow the analysis of omics input data with different row size. The proposed methodology has been used to mine microarray of solid tumors of different embryonic origin to verify the presence of common genes characterizing the heterogeneity of cancer-associated fibroblasts. These automatically extracted biomarkers could be used to suggest appropriate therapies to inactivate the state of active fibroblasts, thus avoiding their action on tumor progression.
Collapse
Affiliation(s)
- Flavia Esposito
- Department of Electronic and Information Engineering, Politecnico di Bari, Bari, Italy
| | - Angelina Boccarelli
- Department of Biomedical Science and Human Oncology, University of Bari Medical School, Bari, Italy
| | | |
Collapse
|
48
|
Gremski LH, da Justa HC, da Silva TP, Polli NLC, Antunes BC, Minozzo JC, Wille ACM, Senff-Ribeiro A, Arni RK, Veiga SS. Forty Years of the Description of Brown Spider Venom Phospholipases-D. Toxins (Basel) 2020; 12:toxins12030164. [PMID: 32155765 PMCID: PMC7150852 DOI: 10.3390/toxins12030164] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 01/24/2023] Open
Abstract
Spiders of the genus Loxosceles, popularly known as Brown spiders, are considered a serious public health issue, especially in regions of hot or temperate climates, such as parts of North and South America. Although the venoms of these arachnids are complex in molecular composition, often containing proteins with distinct biochemical characteristics, the literature has primarily described a family of toxins, the Phospholipases-D (PLDs), which are highly conserved in all Loxosceles species. PLDs trigger most of the major clinical symptoms of loxoscelism i.e., dermonecrosis, thrombocytopenia, hemolysis, and acute renal failure. The key role played by PLDs in the symptomatology of loxoscelism was first described 40 years ago, when researches purified a hemolytic toxin that cleaved sphingomyelin and generated choline, and was referred to as a Sphingomyelinase-D, which was subsequently changed to Phospholipase-D when it was demonstrated that the enzyme also cleaved other cellular phospholipids. In this review, we present the information gleaned over the last 40 years about PLDs from Loxosceles venoms especially with regard to the production and characterization of recombinant isoforms. The history of obtaining these toxins is discussed, as well as their molecular organization and mechanisms of interaction with their substrates. We will address cellular biology aspects of these toxins and how they can be used in the development of drugs to address inflammatory processes and loxoscelism. Present and future aspects of loxoscelism diagnosis will be discussed, as well as their biotechnological applications and actions expected for the future in this field.
Collapse
Affiliation(s)
- Luiza Helena Gremski
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Hanna Câmara da Justa
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Thaís Pereira da Silva
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Nayanne Louise Costacurta Polli
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Bruno César Antunes
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
- Centro de Produção e Pesquisa de Imunobiológicos (CPPI), Piraquara 83302-200, PR, Brazil;
| | - João Carlos Minozzo
- Centro de Produção e Pesquisa de Imunobiológicos (CPPI), Piraquara 83302-200, PR, Brazil;
| | - Ana Carolina Martins Wille
- Departamento de Biologia Estrutural, Molecular e Genética, Universidade Estadual de Ponta Grossa, Ponta Grossa 84030-900, PR, Brazil;
| | - Andrea Senff-Ribeiro
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Raghuvir Krishnaswamy Arni
- Centro Multiusuário de Inovação Biomolecular, Departamento de Física, Universidade Estadual Paulista (UNESP), São José do Rio Preto 15054-000, SP, Brazil;
| | - Silvio Sanches Veiga
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
- Correspondence: ; Tel.: +55-(41)-3361-1776
| |
Collapse
|
49
|
Ma X, Feng J, Lu M, Tang W, Han J, Luo X, Zhao Q, Yang L. microRNA-501-5p promotes cell proliferation and migration in gastric cancer by downregulating LPAR1. J Cell Biochem 2019; 121:1911-1922. [PMID: 31746031 DOI: 10.1002/jcb.29426] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/08/2019] [Indexed: 12/24/2022]
Abstract
In spite of the achievement in treatment, the gastric cancer (GC) mortality still remains high. MicroRNAs (miRNAs) are a group of small noncoding RNAs that play a crucial part in tumor progression. In this study, we explored the expression and function of microRNA-501-5p (miR-501-5p) in GC cell lines. Quantitative real-time polymerase chain reaction assay results suggested that miR-501-5p was significantly upregulated in GC tissues and cell lines. And, the Cell Counting Kit-8 colony formation and cell migration assay results showed that the downregulation of miR-501-5p decreased GC cell proliferation and migration. Besides that, we found that GC cell cycle was arrested in G2 phase and cell apoptosis rate was increased by silencing the expression of miR-501-5p in GC cell lines using the flow cytometry. We also found that miR-501-5p could directly target lysophosphatidic acid receptor 1 (LPAR1) and negatively regulate LPAR1 expression in GC cell lines by performing dual-luciferase reporter gene assay and Western blot analysis. And, LPAR1 was significantly downregulated in GC tissues and inversely correlated with miR-501-5p expression. Furthermore, LPAR1 downregulation promoted cell proliferation and migration, which were attenuated by cotransfection of miR-501-5p inhibitor in GC cells. In conclusion, miR-501-5p can promote GC cell proliferation and migration by targeting and downregulating LPAR1. miR-501-5p/LPAR1 may become a potential therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Xiang Ma
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaxi Feng
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ming Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenjuan Tang
- Department of Newborn Infants, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianbo Han
- Department of General Surgery, Nanjing Red Cross Hospital, Nanjing, Jiangsu, China
| | - XiaGang Luo
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qinghong Zhao
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
50
|
Shin E, Koo JS. Expression of proteins related to autotaxin-lysophosphatidate signaling in thyroid tumors. J Transl Med 2019; 17:288. [PMID: 31455351 PMCID: PMC6712878 DOI: 10.1186/s12967-019-2028-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 08/18/2019] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND We aimed to investigate the expression of proteins related with autotaxin (ATX)-lysophosphatidate (LPA) signaling and the clinical implications in primary and metastatic thyroid tumors. METHODS We constructed tissue microarrays with 545 primary thyroid tumors [338 papillary thyroid carcinoma (PTC), 111 follicular carcinoma (FC), 69 medullary carcinoma (MC), 23 poorly differentiated carcinoma (PDC), and four anaplastic carcinoma (AC)]. Immunohistochemical stains for proteins related to ATX-LPA signaling (e.g., ATX, LPA1, LPA2, and LPA3) were performed. RESULTS The expression of ATX was highest in MC, while the LPA1 expression was higher in PDC and AC, and the expression of LPA2 and LPA3 was highest in PTC (p < 0.001). Additionally, the expression of ATX, LPA1, and LPA2 was higher in conventional-type PTC than in follicular-variant PTC (p < 0.05). PTC with BRAF V600E mutation showed higher expression of ATX, LPA1, LPA2, and LPA3 than PTC without BRAF V600E mutation (p < 0.001). In univariate analysis, ATX positivity (p = 0.005) and LPA1 positivity (p = 0.014) were correlated with shorter overall survival in PTC. CONCLUSION Proteins related to the ATX-LPA axis showed different levels of expression in primary thyroid tumors according to subtype.
Collapse
Affiliation(s)
- Eunah Shin
- Department of Pathology, CHA Gangnam Medical Center, CHA University School of Medicine, Seoul, South Korea.,Department of Pathology, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Ja Seung Koo
- Department of Pathology, CHA Gangnam Medical Center, CHA University School of Medicine, Seoul, South Korea. .,Department of Pathology, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea.
| |
Collapse
|