1
|
Belardo C, Jebali J, Boccella S, Infantino R, Fusco A, Perrone M, Bonsale R, Manzo I, Iannotta M, Scuteri D, Ferraraccio F, Panarese I, Ferrara G, Guida F, Luongo L, Palazzo E, Srairi-Abid N, Marrakchi N, Maione S. Biphasic Hormetic-like Effect of Lebecetin, a C-type Lectin of Snake Venom, on Formalin-induced Inflammation in Mice. Curr Neuropharmacol 2024; 22:1391-1405. [PMID: 38073106 PMCID: PMC11092918 DOI: 10.2174/1570159x22999231207105743] [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: 12/14/2022] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Integrins, important extracellular matrix (ECM) receptor proteins, are affected by inflammation and can participate in the maintenance of many painful conditions. Although they are ubiquitous and changeable across all cell types, the roles of these cell adhesion molecules in pathological pain have not been fully explored. OBJECTIVE We evaluated the effects of the subcutaneous injection of lebecetin, a C-type lectin isolated from Macrovipera lebetina snake venom, previously reported to inhibit α5β1 and αv integrin activity, on different components of inflammation induced by the formalin administration in the hind paw of mice. METHODS The formalin-induced nocifensive behavior, edema, and histopathological changes in the hind paw associated with cytokine, iNOS, and COX2 expression, nociceptive-specific neuron activity, and microglial activation analysis in the spinal cord were evaluated in mice receiving vehicle or lebecetin pretreatment. RESULTS Lebecetin inhibited the nocifensive responses in the formalin test, related edema, and cell infiltration in the injected paw in a biphasic, hormetic-like, and dose-dependent way. According to that hormetic trend, a reduction in pro-inflammatory cytokines IL-6, IL-8, and TNF-alpha and upregulation of the anti-inflammatory cytokine IL-10 in the spinal cord were found with the lowest doses of lebecetin. Moreover, COX2 and iNOS expression in serum and spinal cord followed the same biphasic pattern of cytokines. Finally, nociceptive neurons sensitization and activated microglia were normalized in the dorsal horn of the spinal cord by lebecetin. CONCLUSION These findings implicate specific roles of integrins in inflammation and tonic pain, as well as in the related central nervous system sequelae.
Collapse
Affiliation(s)
- Carmela Belardo
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Jed Jebali
- Laboratory of Biomolecules, Venoms and Theranostic Applications, LR20IPT01, Institut Pasteur of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Serena Boccella
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Antimo Fusco
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Michela Perrone
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Roozbe Bonsale
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Iolanda Manzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Damiana Scuteri
- Pharmacotechnology Documentation and Transfer Unit, Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Franca Ferraraccio
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Iacopo Panarese
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Giovanna Ferrara
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Enza Palazzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Najet Srairi-Abid
- Laboratory of Biomolecules, Venoms and Theranostic Applications, LR20IPT01, Institut Pasteur of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Naziha Marrakchi
- Laboratory of Biomolecules, Venoms and Theranostic Applications, LR20IPT01, Institut Pasteur of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Sabatino Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, Naples, Italy
| |
Collapse
|
2
|
Giedt MS, Thomalla JM, White RP, Johnson MR, Lai ZW, Tootle TL, Welte MA. Adipose triglyceride lipase promotes prostaglandin-dependent actin remodeling by regulating substrate release from lipid droplets. Development 2023; 150:dev201516. [PMID: 37306387 PMCID: PMC10281261 DOI: 10.1242/dev.201516] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
Lipid droplets (LDs), crucial regulators of lipid metabolism, accumulate during oocyte development. However, their roles in fertility remain largely unknown. During Drosophila oogenesis, LD accumulation coincides with the actin remodeling necessary for follicle development. Loss of the LD-associated Adipose Triglyceride Lipase (ATGL) disrupts both actin bundle formation and cortical actin integrity, an unusual phenotype also seen when the prostaglandin (PG) synthase Pxt is missing. Dominant genetic interactions and PG treatment of follicles indicate that ATGL acts upstream of Pxt to regulate actin remodeling. Our data suggest that ATGL releases arachidonic acid (AA) from LDs to serve as the substrate for PG synthesis. Lipidomic analysis detects AA-containing triglycerides in ovaries, and these are increased when ATGL is lost. High levels of exogenous AA block follicle development; this is enhanced by impairing LD formation and suppressed by reducing ATGL. Together, these data support the model that AA stored in LD triglycerides is released by ATGL to drive the production of PGs, which promote the actin remodeling necessary for follicle development. We speculate that this pathway is conserved across organisms to regulate oocyte development and promote fertility.
Collapse
Affiliation(s)
- Michelle S. Giedt
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - Roger P. White
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Matthew R. Johnson
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Zon Weng Lai
- Harvard T.H. Chan Advanced Multi-omics Platform, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tina L. Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Michael A. Welte
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
3
|
Talbot DE, Vormezeele BJ, Kimble GC, Wineland DM, Kelpsch DJ, Giedt MS, Tootle TL. Prostaglandins limit nuclear actin to control nucleolar function during oogenesis. Front Cell Dev Biol 2023; 11:1072456. [PMID: 36875757 PMCID: PMC9981675 DOI: 10.3389/fcell.2023.1072456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
Prostaglandins (PGs), locally acting lipid signals, regulate female reproduction, including oocyte development. However, the cellular mechanisms of PG action remain largely unknown. One cellular target of PG signaling is the nucleolus. Indeed, across organisms, loss of PGs results in misshapen nucleoli, and changes in nucleolar morphology are indicative of altered nucleolar function. A key role of the nucleolus is to transcribe ribosomal RNA (rRNA) to drive ribosomal biogenesis. Here we take advantage of the robust, in vivo system of Drosophila oogenesis to define the roles and downstream mechanisms whereby PGs regulate the nucleolus. We find that the altered nucleolar morphology due to PG loss is not due to reduced rRNA transcription. Instead, loss of PGs results in increased rRNA transcription and overall protein translation. PGs modulate these nucleolar functions by tightly regulating nuclear actin, which is enriched in the nucleolus. Specifically, we find that loss of PGs results in both increased nucleolar actin and changes in its form. Increasing nuclear actin, by either genetic loss of PG signaling or overexpression of nuclear targeted actin (NLS-actin), results in a round nucleolar morphology. Further, loss of PGs, overexpression of NLS-actin or loss of Exportin 6, all manipulations that increase nuclear actin levels, results in increased RNAPI-dependent transcription. Together these data reveal PGs carefully balance the level and forms of nuclear actin to control the level of nucleolar activity required for producing fertilization competent oocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tina L. Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
4
|
Significance of Pulmonary Endothelial Injury and the Role of Cyclooxygenase-2 and Prostanoid Signaling. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010117. [PMID: 36671689 PMCID: PMC9855370 DOI: 10.3390/bioengineering10010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
The endothelium plays a key role in the dynamic balance of hemodynamic, humoral and inflammatory processes in the human body. Its central importance and the resulting therapeutic concepts are the subject of ongoing research efforts and form the basis for the treatment of numerous diseases. The pulmonary endothelium is an essential component for the gas exchange in humans. Pulmonary endothelial dysfunction has serious consequences for the oxygenation and the gas exchange in humans with the potential of consecutive multiple organ failure. Therefore, in this review, the dysfunction of the pulmonary endothel due to viral, bacterial, and fungal infections, ventilator-related injury, and aspiration is presented in a medical context. Selected aspects of the interaction of endothelial cells with primarily alveolar macrophages are reviewed in more detail. Elucidation of underlying causes and mechanisms of damage and repair may lead to new therapeutic approaches. Specific emphasis is placed on the processes leading to the induction of cyclooxygenase-2 and downstream prostanoid-based signaling pathways associated with this enzyme.
Collapse
|
5
|
Matsumoto N, Singh N, Lee KS, Barnych B, Morisseau C, Hammock BD. The epoxy fatty acid pathway enhances cAMP in mammalian cells through multiple mechanisms. Prostaglandins Other Lipid Mediat 2022; 162:106662. [PMID: 35779854 PMCID: PMC9530012 DOI: 10.1016/j.prostaglandins.2022.106662] [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: 03/17/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 10/17/2022]
Abstract
The cellular mechanism by which epoxy fatty acids (EpFA) improves disease status is not well characterized. Previous studies suggest the involvement of cellular receptors and cyclic AMP (cAMP). Herein, the action of EpFAs derived from linoleic acid (LA), arachidonic acid (ARA), and docosahexaenoic acid on cAMP levels was studied in multiple cell types to elucidate relationships between EpFAs, receptors and cells' origin. cAMP levels were enhanced in HEK293 and LLC-PK1 cells by EpFAs from LA and ARA. Using selective antagonists, the EpFA effects on cAMP levels appear dependent on the prostaglandin E2 receptor 2 (EP2) but not 4 (EP4). Human coronary artery smooth muscle cells responded similarly to the EpFAs. However, we were not able to show the involvement of any of the receptors tested in this cell type. The results pinpointed distinct cell lines and receptor subtypes that natively respond to EpFA.
Collapse
Affiliation(s)
- Naoki Matsumoto
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis CA, USA
| | - Nalin Singh
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis CA, USA
| | - Kin Sing Lee
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing MI, USA
| | - Bogdan Barnych
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis CA, USA
| | - Christophe Morisseau
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis CA, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis CA, USA.
| |
Collapse
|
6
|
Svec KV, Howe AK. Protein Kinase A in cellular migration—Niche signaling of a ubiquitous kinase. Front Mol Biosci 2022; 9:953093. [PMID: 35959460 PMCID: PMC9361040 DOI: 10.3389/fmolb.2022.953093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/30/2022] [Indexed: 12/28/2022] Open
Abstract
Cell migration requires establishment and maintenance of directional polarity, which in turn requires spatial heterogeneity in the regulation of protrusion, retraction, and adhesion. Thus, the signaling proteins that regulate these various structural processes must also be distinctly regulated in subcellular space. Protein Kinase A (PKA) is a ubiquitous serine/threonine kinase involved in innumerable cellular processes. In the context of cell migration, it has a paradoxical role in that global inhibition or activation of PKA inhibits migration. It follows, then, that the subcellular regulation of PKA is key to bringing its proper permissive and restrictive functions to the correct parts of the cell. Proper subcellular regulation of PKA controls not only when and where it is active but also specifies the targets for that activity, allowing the cell to use a single, promiscuous kinase to exert distinct functions within different subcellular niches to facilitate cell movement. In this way, understanding PKA signaling in migration is a study in context and in the elegant coordination of distinct functions of a single protein in a complex cellular process.
Collapse
Affiliation(s)
- Kathryn V. Svec
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
| | - Alan K. Howe
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, V T, United States
- University of Vermont Cancer Center, University of Vermont, Burlington, VT, United States
- *Correspondence: Alan K. Howe,
| |
Collapse
|
7
|
Tao X, Zhang R, Du R, Yu T, Yang H, Li J, Wang Y, Liu Q, Zuo S, Wang X, Lazarus M, Zhou L, Wang B, Yu Y, Shen Y. EP3 enhances adhesion and cytotoxicity of NK cells toward hepatic stellate cells in a murine liver fibrosis model. J Exp Med 2022; 219:213141. [PMID: 35420633 DOI: 10.1084/jem.20212414] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/07/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells exhibit antifibrotic properties in liver fibrosis (LF) by suppressing activated hepatic stellate cell (HSC) populations. Prostaglandin E2 (PGE2) plays a dual role in innate and adaptive immunity. Here, we found that E-prostanoid 3 receptor (EP3) was markedly downregulated in NK cells from liver fibrosis mice and patients with liver cirrhosis. NK cell-specific deletion of EP3 aggravated hepatic fibrogenesis in mouse models of LF. Loss of EP3 selectively reduced the cytotoxicity of the CD27+CD11b+ double positive (DP) NK subset against activated HSCs. Mechanistically, deletion of EP3 impaired the adhesion and cytotoxicity of DP NK cells toward HSCs through modulation of Itga4-VCAM1 binding. EP3 upregulated Itga4 expression in NK cells through promoting Spic nuclear translocation via PKC-mediated phosphorylation of Spic at T191. Activation of EP3 by sulprostone alleviated CCL4-induced liver fibrosis in mice. Thus, EP3 is required for adhesion and cytotoxicity of NK cells toward HSCs and may serve as a therapeutic target for the management of LF.
Collapse
Affiliation(s)
- Xixi Tao
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Rui Zhang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ronglu Du
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Tingting Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hui Yang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jiwen Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yuhong Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qian Liu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shengkai Zuo
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xi Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Ying Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yujun Shen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
8
|
Yasmin IA, Mohana Sundaram S, Banerjee A, Varier L, Dharmarajan A, Warrier S. Netrin-like domain of sFRP4, a Wnt antagonist inhibits stemness, metastatic and invasive properties by specifically blocking MMP-2 in cancer stem cells from human glioma cell line U87MG. Exp Cell Res 2021; 409:112912. [PMID: 34762897 DOI: 10.1016/j.yexcr.2021.112912] [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: 09/06/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 11/26/2022]
Abstract
Rapid proliferation, high stemness potential, high invasiveness and apoptotic evasion are the distinctive hallmarks of glioma malignancy. The dysregulation of the Wnt/β-catenin pathway is the key factor regulating glioma malignancy. Wnt antagonist, secreted frizzled-related protein 4 (sFRP4), which has a prominent pro-apoptotic role in glioma stem cells, has two functional domains, the netrin-like domain (NLD), and cysteine-rich domain (CRD) both of which contribute to apoptotic properties of the whole protein. However, there are no reports elucidating the specific effects of individual domains of sFRP4 in inhibiting the invasive properties of glioma. This study explores the efficacy of the domains of sFRP4 in inhibiting the key hallmarks of glioblastoma such as invasion, metastasis, and stemness. We overexpressed sFRP4 and its domains in the glioblastoma cell line, U87MG cells and observed that both CRD and NLD domains played prominent roles in attenuating cancer stem cell properties. Significantly, we could demonstrate for the first time that both NLD and CRD domains negatively impacted the key driver of metastasis and migration, the matrix metalloproteinase-2 (MMP-2). Mechanistically, compared to CRD, NLD domain suppressed MMP-2 mediated invasion more effectively in glioma cells as observed in matrigel invasion assay and a function-blocking antibody assay. Fluorescent matrix degradation assay further revealed that NLD reduces matrix degradation. NLD also significantly disrupted fibronectin assembly and decreased cell adhesion in another glioma cell line LN229. In conclusion, the NLD peptide of sFRP4 could be a potent short peptide therapeutic candidate for targeting MMP-2-mediated invasion in the highly malignant glioblastoma multiforme.
Collapse
Affiliation(s)
- Ishmat Ara Yasmin
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560 065, India
| | - S Mohana Sundaram
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560 065, India
| | - Anasuya Banerjee
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560 065, India
| | | | - Arun Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, 600 116, India
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560 065, India; Cuor Stem Cellutions Pvt Ltd, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560 065, India.
| |
Collapse
|
9
|
Dhavalikar P, Robinson A, Lan Z, Jenkins D, Chwatko M, Salhadar K, Jose A, Kar R, Shoga E, Kannapiran A, Cosgriff-Hernandez E. Review of Integrin-Targeting Biomaterials in Tissue Engineering. Adv Healthc Mater 2020; 9:e2000795. [PMID: 32940020 PMCID: PMC7960574 DOI: 10.1002/adhm.202000795] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/27/2020] [Indexed: 12/12/2022]
Abstract
The ability to direct cell behavior has been central to the success of numerous therapeutics to regenerate tissue or facilitate device integration. Biomaterial scientists are challenged to understand and modulate the interactions of biomaterials with biological systems in order to achieve effective tissue repair. One key area of research investigates the use of extracellular matrix-derived ligands to target specific integrin interactions and induce cellular responses, such as increased cell migration, proliferation, and differentiation of mesenchymal stem cells. These integrin-targeting proteins and peptides have been implemented in a variety of different polymeric scaffolds and devices to enhance tissue regeneration and integration. This review first presents an overview of integrin-mediated cellular processes that have been identified in angiogenesis, wound healing, and bone regeneration. Then, research utilizing biomaterials are highlighted with integrin-targeting motifs as a means to direct these cellular processes to enhance tissue regeneration. In addition to providing improved materials for tissue repair and device integration, these innovative biomaterials provide new tools to probe the complex processes of tissue remodeling in order to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Prachi Dhavalikar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Robinson
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Dana Jenkins
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Malgorzata Chwatko
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Karim Salhadar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Anupriya Jose
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ronit Kar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Erik Shoga
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Aparajith Kannapiran
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | | |
Collapse
|
10
|
Lamb MC, Tootle TL. Fascin in Cell Migration: More Than an Actin Bundling Protein. BIOLOGY 2020; 9:biology9110403. [PMID: 33212856 PMCID: PMC7698196 DOI: 10.3390/biology9110403] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022]
Abstract
Simple Summary Cell migration is an essential biological process that regulates both development and diseases, such as cancer metastasis. Therefore, understanding the factors that promote cell migration is crucial. One of the factors known to regulate cell migration is the actin-binding protein, Fascin. Fascin is typically thought to promote cell migration through bundling actin to form migratory structures such as filopodia and invadapodia. However, Fascin has many other functions in the cell that may contribute to cell migration. How these novel functions promote cell migration and are regulated is still not well understood. Here, we review the structure of Fascin, the many functions of Fascin and how they may promote cell migration, how Fascin is regulated, and Fascin’s role in diseases such as cancer metastasis. Abstract Fascin, an actin-binding protein, regulates many developmental migrations and contributes to cancer metastasis. Specifically, Fascin promotes cell motility, invasion, and adhesion by forming filopodia and invadopodia through its canonical actin bundling function. In addition to bundling actin, Fascin has non-canonical roles in the cell that are thought to promote cell migration. These non-canonical functions include regulating the activity of other actin-binding proteins, binding to and regulating microtubules, mediating mechanotransduction to the nucleus via interaction with the Linker of the Nucleoskeleton and Cytoskeleton (LINC) Complex, and localizing to the nucleus to regulate nuclear actin, the nucleolus, and chromatin modifications. The many functions of Fascin must be coordinately regulated to control cell migration. While much remains to be learned about such mechanisms, Fascin is regulated by post-translational modifications, prostaglandin signaling, protein–protein interactions, and transcriptional means. Here, we review the structure of Fascin, the various functions of Fascin and how they contribute to cell migration, the mechanisms regulating Fascin, and how Fascin contributes to diseases, specifically cancer metastasis.
Collapse
|
11
|
Du Y, Taylor CG, Aukema HM, Zahradka P. Role of oxylipins generated from dietary PUFAs in the modulation of endothelial cell function. Prostaglandins Leukot Essent Fatty Acids 2020; 160:102160. [PMID: 32717531 DOI: 10.1016/j.plefa.2020.102160] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/17/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022]
Abstract
Oxylipins, which are circulating bioactive lipids generated from polyunsaturated fatty acids (PUFAs) by cyclooxygenase, lipooxygenase and cytochrome P450 enzymes, have diverse effects on endothelial cells. Although studies of the effects of oxylipins on endothelial cell function are accumulating, a review that provides a comprehensive compilation of current knowledge and recent advances in the context of vascular homeostasis is lacking. This is the first compilation of the various in vitro, ex vivo and in vivo reports to examine the effects and potential mechanisms of action of oxylipins on endothelial cells. The aggregate data indicate docosahexaenoic acid-derived oxylipins consistently show beneficial effects related to key endothelial cell functions, whereas oxylipins derived from other PUFAs exhibit both positive and negative effects. Furthermore, information is lacking for certain oxylipin classes, such as those derived from α-linolenic acid, which suggests additional studies are required to achieve a full understanding of how oxylipins affect endothelial cells.
Collapse
Affiliation(s)
- Youjia Du
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Physiology and Pathophysiology, University of Manitoba, MB R3E 0J9, Canada
| | - Carla G Taylor
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Physiology and Pathophysiology, University of Manitoba, MB R3E 0J9, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada
| | - Harold M Aukema
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada
| | - Peter Zahradka
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Physiology and Pathophysiology, University of Manitoba, MB R3E 0J9, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada.
| |
Collapse
|
12
|
CP-25 inhibits PGE2-induced angiogenesis by down-regulating EP4/AC/cAMP/PKA-mediated GRK2 translocation. Clin Sci (Lond) 2020; 134:331-347. [PMID: 31967309 DOI: 10.1042/cs20191032] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/19/2022]
Abstract
G protein-coupled receptor kinase 2 (GRK2), a type of cytosolic enzyme, transiently translocates to the plasma membrane upon G protein-coupled receptors (GPCRs) activation, and it also binds to extracellular signal-regulated kinase (ERK) to inhibit the activation of ERK. GRK2 deficiency in endothelial cells (ECs) leads to increased pro-inflammatory signaling and promotes recruitment of leukocytes to activated ECs. However, the role of GRK2 in regulating angiogenesis remains unclear. Here, we show that GRK2 is a novel regulatory molecule on migration and tube formation of ECs, vessel sprouting ex vivo and angiogenesis in vivo. We identify that EP4/AC/cAMP/protein kinase A (PKA)-mediated GRK2 translocation to cells membrane decreases the binding of GRK2 and ERK1/2 to inhibit ERK1/2 activation, which promotes prostaglandin E2 (PGE2)-induced angiogenesis. GRK2 small interfering RNA (siRNA) inhibits the increase in PGE2-induced HUVECs migration and tube formation. In vivo, PGE2 increases ECs sprouting from normal murine aortic segments and angiogenesis in mice, but not from GRK2-deficient ones, on Matrigel. Further research found that Lys220 and Ser685 of GRK2 play an important role in angiogenesis by regulating GRK2 translocation. Paeoniflorin-6'-O-benzene sulfonate (CP-25), as a novel ester derivative of paeoniflorin (pae), has therapeutic potential for the treatment of adjuvant arthritis (AA) and collagen-induced arthritis (CIA), but the underlying mechanism of CP-25 on angiogenesis has not been elucidated. In our study, CP-25 inhibits the migration and tube formation of HUVECs, and angiogenesis in mice by down-regulating GRK2 translocation activation without affecting GRK2 total expression. Taken together, the present results revealed that CP-25 down-regulates EP4/AC/cAMP/PKA-mediated GRK2 translocation, restoring the inhibition of GRK2 for ERK1/2, thereby inhibiting PGE2-stimulated angiogenesis.
Collapse
|
13
|
Pharmaco-Genetic Screen To Uncover Actin Regulators Targeted by Prostaglandins During Drosophila Oogenesis. G3-GENES GENOMES GENETICS 2019; 9:3555-3565. [PMID: 31506320 PMCID: PMC6829128 DOI: 10.1534/g3.119.400704] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Prostaglandins (PGs) are lipid signaling molecules with numerous physiologic functions, including pain/inflammation, fertility, and cancer. PGs are produced downstream of cyclooxygenase (COX) enzymes, the targets of non-steroidal anti-inflammatory drugs (NSAIDs). In numerous systems, PGs regulate actin cytoskeletal remodeling, however, their mechanisms of action remain largely unknown. To address this deficiency, we undertook a pharmaco-genetic interaction screen during late-stage Drosophila oogenesis. Drosophila oogenesis is as an established model for studying both actin dynamics and PGs. Indeed, during Stage 10B, cage-like arrays of actin bundles surround each nurse cell nucleus, and during Stage 11, the cortical actin contracts, squeezing the cytoplasmic contents into the oocyte. Both of these cytoskeletal properties are required for follicle development and fertility, and are regulated by PGs. Here we describe a pharmaco-genetic interaction screen that takes advantage of the fact that Stage 10B follicles will mature in culture and COX inhibitors, such as aspirin, block this in vitro follicle maturation. In the screen, aspirin was used at a concentration that blocks 50% of the wild-type follicles from maturing in culture. By combining this aspirin treatment with heterozygosity for mutations in actin regulators, we quantitatively identified enhancers and suppressors of COX inhibition. Here we present the screen results and initial follow-up studies on three strong enhancers – Enabled, Capping protein, and non-muscle Myosin II Regulatory Light Chain. Overall, these studies provide new insight into how PGs regulate both actin bundle formation and cellular contraction, properties that are not only essential for development, but are misregulated in disease.
Collapse
|
14
|
Abstract
Cancer development and metastasis are associated to perturbation in metabolic functions of tumor cells and surrounding inflammatory and stromal cell responses. Eicosanoids and lipid mediators, in this regard, attract potential attention during cancer development. Eicosanoids, which include prostaglandin, prostacyclin, thromboxane, and leukotriene, are synthesized from arachidonic acid when cells are stimulated by stress, cytokines, or other growth factors. However, the underlying mechanism of eicosanoids in cancer development, specially their interactions with proto-oncogene factors in tumor microenvironment, remain unexplored. On the other hand, matrix metalloproteinases (MMPs) are a group of zinc-dependent endopeptidases which are involved in degradation of different extracellular matrix (ECM) proteins. MMPs are associated with different physiological responses, including embryogenesis, vasculogenesis, and cellular remodeling, as well as different disease pathogenesis. Induced MMP responses are especially associated with cancer metastasis and secondary tumor development through proteolytic cleavage of several ECM and non-ECM proteins. Although both eicosanoids and MMPs are involved with cancer progression and metastasis, the interrelation between these two molecules are less explored. The present review discusses relevant studies that connect eicosanoids and MMPs and highlight the crosstalk between them offering novel therapeutic approach in cancer treatment.
Collapse
|
15
|
Khan GA, Bhagat S, Alam MI. PGE 2 -induced migration of human brain endothelial cell is mediated though protein kinase A in cooperation of EP receptors. J Leukoc Biol 2019; 105:705-717. [PMID: 30835912 DOI: 10.1002/jlb.2a0918-361r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/21/2019] [Accepted: 02/10/2019] [Indexed: 12/14/2022] Open
Abstract
PGE2 plays a critical role in angiogenesis, ischemic, and neuro-inflammatory disorders of the brain, which breakdown the blood-brain barrier (BBB). However, the effects of PGE2 on human brain endothelial cell (HBECs) migration, a key process in the angiogenic response and BBB stability, are not well defined. In this study, we investigated the mechanism of PGE2 in HBECs migration in vitro. Here we showed that PGE2 stimulated migration of HBECs in a dose-time and matrix-dependent manner, evaluated by the Boyden chamber assay, but other prostanoids failed to do so. PGE2 receptor (EP2; butaprost), EP3 (sulprostone), and EP4 (PGE1 -OH) receptor agonists stimulated HBECs migration, but the silencing of EP significantly attenuated this effect. EP1 agonist (11-trinor PGE1 ) had no effect on HBECs migration on silencing of the EP1 receptor. We further showed that PGE2 stimulated cAMP production and activated protein kinase A (PKA), whereas pretreatment with the adenyl cyclase inhibitor (dideoxyadenosine; 1 μM) or PKA inhibitors, H89 (0.5 μM)/PKAI (1 μM), completely abrogated PGE2 -induced migration. Furthermore, silencing of the EP2/EP4 receptors significantly inhibited PGE2 -induced cAMP and PKA activation, whereas EP3 receptor silencing failed to do so. These results suggest that PGE2 regulates HBEC migration via cooperation of EP2, EP3, and EP4 receptors. Coupling of PGE2 to these receptors resulted in increased production of cAMP, which regulates HBEC migration via PKA pathway. The elucidation of molecular events involved is critical for the development of targeted strategies to treat cerebrovascular diseases associated with dysregulated angiogenesis.
Collapse
Affiliation(s)
- Gausal A Khan
- Department of Physiology, Defence Institute of Physiology and Allied Sciences, Timarpur, New Delhi, India
| | - Saumya Bhagat
- Department of Physiology, Defence Institute of Physiology and Allied Sciences, Timarpur, New Delhi, India
| | - Md Iqbal Alam
- Department of Physiology, HIMSR, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| |
Collapse
|
16
|
Park A, Lee Y, Kim MS, Kang YJ, Park YJ, Jung H, Kim TD, Lee HG, Choi I, Yoon SR. Prostaglandin E2 Secreted by Thyroid Cancer Cells Contributes to Immune Escape Through the Suppression of Natural Killer (NK) Cell Cytotoxicity and NK Cell Differentiation. Front Immunol 2018; 9:1859. [PMID: 30140269 PMCID: PMC6094168 DOI: 10.3389/fimmu.2018.01859] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/27/2018] [Indexed: 12/23/2022] Open
Abstract
Natural killer (NK) cells play important roles in immune surveillance. However, the tumor microenvironment suppresses NK cell function and allows cancer cells to evade immune detection. In this study, we investigated whether the thyroid cancer cell microenvironment has this effect on NK cells. We found that prostaglandin (PG) E2 produced by thyroid cancer cells suppressed the cytolytic activity of NK cells by inhibiting the expression of the natural cytotoxicity receptors NKp44 and NKp30 and the death receptor tumor necrosis factor-related apoptosis-inducing ligand. PGE2 and cyclooxygenase-2 were highly expressed in thyroid cancer cells; moreover, anaplastic thyroid cancer cells released higher amounts of PGE2 than the papillary subtype, which was associated with suppression of NK cell-inducing nuclear factor-κB and mitogen-activated protein kinase/extracellular signal-regulated kinase pathways via PGE2 receptor (EP) 2 and EP4 expressed on the NK cell surface. In addition, PGE2 inhibited the functional maturation of NK cells and reduced their cytotoxicity against target cells. These results indicate that PGE2 promotes thyroid cancer progression by inhibiting NK cell maturation and cytotoxicity. Thus, therapeutic strategies that target PGE2 in thyroid cancer could potentiate the immune response and improve patient prognosis.
Collapse
Affiliation(s)
- Arum Park
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science & Technology, Daejeon, South Korea
| | - Yunhee Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biochemistry, College of Pharmacy, Chungnam National University, Daejeon, South Korea
| | - Mi Sun Kim
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Young Ju Kang
- New Drug Development Center, OSONG Medical Innovation Foundation, Cheongju-si, South Korea
| | - Young-Jun Park
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science & Technology, Daejeon, South Korea
| | - Haiyoung Jung
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science & Technology, Daejeon, South Korea
| | - Tae-Don Kim
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science & Technology, Daejeon, South Korea
| | - Hee Gu Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science & Technology, Daejeon, South Korea
| | - Inpyo Choi
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science & Technology, Daejeon, South Korea
| | - Suk Ran Yoon
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science & Technology, Daejeon, South Korea
| |
Collapse
|
17
|
Nedvetsky PI, Zhao X, Mathivet T, Aspalter IM, Stanchi F, Metzger RJ, Mostov KE, Gerhardt H. cAMP-dependent protein kinase A (PKA) regulates angiogenesis by modulating tip cell behavior in a Notch-independent manner. Development 2017; 143:3582-3590. [PMID: 27702786 DOI: 10.1242/dev.134767] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/08/2016] [Indexed: 01/06/2023]
Abstract
cAMP-dependent protein kinase A (PKA) is a ubiquitously expressed serine/threonine kinase that regulates a variety of cellular functions. Here, we demonstrate that endothelial PKA activity is essential for vascular development, specifically regulating the transition from sprouting to stabilization of nascent vessels. Inhibition of endothelial PKA by endothelial cell-specific expression of dominant-negative PKA in mice led to perturbed vascular development, hemorrhage and embryonic lethality at mid-gestation. During perinatal retinal angiogenesis, inhibition of PKA resulted in hypersprouting as a result of increased numbers of tip cells. In zebrafish, cell autonomous PKA inhibition also increased and sustained endothelial cell motility, driving cells to become tip cells. Although these effects of PKA inhibition were highly reminiscent of Notch inhibition effects, our data demonstrate that PKA and Notch independently regulate tip and stalk cell formation and behavior.
Collapse
Affiliation(s)
- Pavel I Nedvetsky
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Xiaocheng Zhao
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Thomas Mathivet
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Irene M Aspalter
- Vascular Biology Laboratory, London Research Institute - Cancer Research UK, Lincoln's Inn Fields Laboratories, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Fabio Stanchi
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ross J Metzger
- Department of Anatomy, University of California San Francisco, Genentech Hall, 600 16th Street, San Francisco, CA 94143-2140, USA
| | - Keith E Mostov
- Department of Anatomy, University of California San Francisco, Genentech Hall, 600 16th Street, San Francisco, CA 94143-2140, USA
| | - Holger Gerhardt
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium Vascular Biology Laboratory, London Research Institute - Cancer Research UK, Lincoln's Inn Fields Laboratories, 44 Lincoln's Inn Fields, London WC2A 3LY, UK Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, Berlin 13125, Germany DZHK (German Center for Cardiovascular Research), partner site Berlin Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
18
|
Sun YY, Zhang WJ, Dong CL, Zhang XF, Ji J, Wang X, Wang L, Hu WL, Du WJ, Cui CL, Zhang CF, Li F, Wang CZ, Yuan CS. Baicalin Alleviates Nitroglycerin-induced Migraine in Rats via the Trigeminovascular System. Phytother Res 2017; 31:899-905. [DOI: 10.1002/ptr.5811] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 03/07/2017] [Accepted: 03/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Yu-Yao Sun
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Wen-Jun Zhang
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Cui-Lan Dong
- The People's Hospital of Zhangqiu; Zhangqiu 250200 China
| | - Xiao-Fan Zhang
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Jun Ji
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Xue Wang
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Ling Wang
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Wan-Li Hu
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Wen-Juan Du
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Cheng-Long Cui
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Chun-Feng Zhang
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care; The University of Chicago; Chicago IL 60637 USA
| | - Fei Li
- State Key Laboratory of Natural Medicines; China Pharmaceutical University; Nanjing JS 210009 China
| | - Chong-Zhi Wang
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care; The University of Chicago; Chicago IL 60637 USA
| | - Chun-Su Yuan
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care; The University of Chicago; Chicago IL 60637 USA
| |
Collapse
|
19
|
Lomba LA, Vogt PH, Souza VEP, Leite-Avalca MCG, Verdan MH, Stefanello MEA, Zampronio AR. A Naphthoquinone from Sinningia canescens Inhibits Inflammation and Fever in Mice. Inflammation 2017; 40:1051-1061. [DOI: 10.1007/s10753-017-0548-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
20
|
The Andes Virus Nucleocapsid Protein Directs Basal Endothelial Cell Permeability by Activating RhoA. mBio 2016; 7:mBio.01747-16. [PMID: 27795403 PMCID: PMC5080385 DOI: 10.1128/mbio.01747-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Andes virus (ANDV) predominantly infects microvascular endothelial cells (MECs) and nonlytically causes an acute pulmonary edema termed hantavirus pulmonary syndrome (HPS). In HPS patients, virtually every pulmonary MEC is infected, MECs are enlarged, and infection results in vascular leakage and highly lethal pulmonary edema. We observed that MECs infected with the ANDV hantavirus or expressing the ANDV nucleocapsid (N) protein showed increased size and permeability by activating the Rheb and RhoA GTPases. Expression of ANDV N in MECs increased cell size by preventing tuberous sclerosis complex (TSC) repression of Rheb-mTOR-pS6K. N selectively bound the TSC2 N terminus (1 to 1403) within a complex containing TSC2/TSC1/TBC1D7, and endogenous TSC2 reciprocally coprecipitated N protein from ANDV-infected MECs. TSCs normally restrict RhoA-induced MEC permeability, and we found that ANDV infection or N protein expression constitutively activated RhoA. This suggests that the ANDV N protein alone is sufficient to activate signaling pathways that control MEC size and permeability. Further, RhoA small interfering RNA, dominant-negative RhoA(N19), and the RhoA/Rho kinase inhibitors fasudil and Y27632 dramatically reduced the permeability of ANDV-infected MECs by 80 to 90%. Fasudil also reduced the bradykinin-directed permeability of ANDV and Hantaan virus-infected MECs to control levels. These findings demonstrate that ANDV activation of RhoA causes MEC permeability and reveal a potential edemagenic mechanism for ANDV to constitutively inhibit the basal barrier integrity of infected MECs. The central importance of RhoA activation in MEC permeability further suggests therapeutically targeting RhoA, TSCs, and Rac1 as potential means of resolving capillary leakage during hantavirus infections. HPS is hallmarked by acute pulmonary edema, hypoxia, respiratory distress, and the ubiquitous infection of pulmonary MECs that occurs without disrupting the endothelium. Mechanisms of MEC permeability and targets for resolving lethal pulmonary edema during HPS remain enigmatic. Our findings suggest a novel underlying mechanism of MEC dysfunction resulting from ANDV activation of the Rheb and RhoA GTPases that, respectively, control MEC size and permeability. Our studies show that inhibition of RhoA blocks ANDV-directed permeability and implicate RhoA as a potential therapeutic target for restoring capillary barrier function to the ANDV-infected endothelium. Since RhoA activation forms a downstream nexus for factors that cause capillary leakage, blocking RhoA activation is liable to restore basal capillary integrity and prevent edema amplified by tissue hypoxia and respiratory distress. Targeting the endothelium has the potential to resolve disease during symptomatic stages, when replication inhibitors lack efficacy, and to be broadly applicable to other hemorrhagic and edematous viral diseases.
Collapse
|
21
|
Lian S, Xia Y, Ung TT, Khoi PN, Yoon HJ, Lee SG, Kim KK, Jung YD. Prostaglandin E 2 stimulates urokinase-type plasminogen activator receptor via EP2 receptor-dependent signaling pathways in human AGS gastric cancer cells. Mol Carcinog 2016; 56:664-680. [PMID: 27377703 DOI: 10.1002/mc.22524] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 06/27/2016] [Accepted: 07/01/2016] [Indexed: 12/29/2022]
Abstract
Aberrant expression of urokinase-type plasminogen activator receptor (uPAR) has been observed in human gastric cancers. Prostaglandin E2 (PGE2 ), whose biosynthesis is catalyzed by cyclooxygenase-2 (COX-2), is implicated in cancer metastasis; however, the cellular and molecular mechanisms of PGE2 -driven uPAR expression are yet to be elucidated in human gastric cancer AGS cells. In this study, we showed that PGE2 induces uPAR expression in concentration- and time-dependent manners. Furthermore, using antagonists and siRNA, we found that among the four subtypes of PGE2 receptors, EP2 receptors are involved in PGE2 -induced uPAR expression. PGE2 induced the activation of Src, epidermal growth factor receptor (EGFR), c-Jun NH2 -terminal kinase (JNK), extracellular signal-regulated kinase (Erk), and p38 mitogen activated protein kinase (p38 MAPK). Specific inhibitor and mutagenesis studies showed that Src, EGFR, JNK1/2, and Erk1/2 are involved in PGE2 -induced uPAR expression. PGE2 induces EP2-dependent phosphorylation of Src, while the activation of Src-dependent EGFR leads to the phosphorylation of JNK1/2 and Erk1/2. Deletion and site-directed mutagenesis studies demonstrated the involvement of transcription factor activator protein (AP)-1 and nuclear factor-kappa B (NF-κB) in PGE2 -induced uPAR expression. EGFR-dependent MAPKs (JNK1/2 and Erk1/2) function as the upstream signaling molecules in the activation of AP-1 and NF-κB, respectively. AGS cells pre-treated with PGE2 showed remarkably enhanced invasiveness, which was partially abrogated by uPAR-neutralizing antibodies. To the best of our knowledge, this is the first report that PGE2 -induced uPAR expression, which stimulates invasiveness of human gastric cancer AGS cells, is mediated by the EP2 receptor-dependent Src/EGFR/JNK1/2, Erk1/2/AP-1, and Src/EGFR/JNK1/2, Erk1/2/NF-κB cascades. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sen Lian
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yong Xia
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Trong Thuan Ung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Pham Ngoc Khoi
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hyun Joong Yoon
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Sam Gyu Lee
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Kyung Keun Kim
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Young Do Jung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
22
|
Broekgaarden M, Weijer R, van Gulik TM, Hamblin MR, Heger M. Tumor cell survival pathways activated by photodynamic therapy: a molecular basis for pharmacological inhibition strategies. Cancer Metastasis Rev 2015; 34:643-90. [PMID: 26516076 PMCID: PMC4661210 DOI: 10.1007/s10555-015-9588-7] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Photodynamic therapy (PDT) has emerged as a promising alternative to conventional cancer therapies such as surgery, chemotherapy, and radiotherapy. PDT comprises the administration of a photosensitizer, its accumulation in tumor tissue, and subsequent irradiation of the photosensitizer-loaded tumor, leading to the localized photoproduction of reactive oxygen species (ROS). The resulting oxidative damage ultimately culminates in tumor cell death, vascular shutdown, induction of an antitumor immune response, and the consequent destruction of the tumor. However, the ROS produced by PDT also triggers a stress response that, as part of a cell survival mechanism, helps cancer cells to cope with the PDT-induced oxidative stress and cell damage. These survival pathways are mediated by the transcription factors activator protein 1 (AP-1), nuclear factor E2-related factor 2 (NRF2), hypoxia-inducible factor 1 (HIF-1), nuclear factor κB (NF-κB), and those that mediate the proteotoxic stress response. The survival pathways are believed to render some types of cancer recalcitrant to PDT and alter the tumor microenvironment in favor of tumor survival. In this review, the molecular mechanisms are elucidated that occur post-PDT to mediate cancer cell survival, on the basis of which pharmacological interventions are proposed. Specifically, pharmaceutical inhibitors of the molecular regulators of each survival pathway are addressed. The ultimate aim is to facilitate the development of adjuvant intervention strategies to improve PDT efficacy in recalcitrant solid tumors.
Collapse
Affiliation(s)
- Mans Broekgaarden
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ruud Weijer
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Thomas M van Gulik
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA, USA
| | - Michal Heger
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
23
|
Vitali E, Cambiaghi V, Spada A, Tresoldi A, Zerbi A, Peverelli E, Carnaghi C, Mantovani G, Lania AG. cAMP effects in neuroendocrine tumors: The role of Epac and PKA in cell proliferation and adhesion. Exp Cell Res 2015; 339:241-51. [PMID: 26589262 DOI: 10.1016/j.yexcr.2015.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 10/18/2015] [Accepted: 11/12/2015] [Indexed: 01/08/2023]
Abstract
cAMP effects have been initially attributed to protein kinase A (PKA) activation. Subsequently, two exchange proteins directly activated by cAMP (Epac1/2) have been identified as cAMP targets. Aim of this study was to investigate cAMP effects in pancreatic-NET (P-NET) and bronchial carcinoids and in corresponding cell lines (QGP-1 and H727) on cell proliferation and adhesion and to determine PKA and Epac role in mediating these effects. We found that cAMP increased cyclin D1 expression in P-NET and QGP-1 cells, whereas it had opposite effects on bronchial carcinoids and H727 cells and it promoted cell adhesion in QGP-1 and H727 cells. These effects are mimicked by Epac and PKA specific analogs, activating the small GTPase Rap1. In conclusion, we demonstrated that cAMP exerted divergent effects on proliferation and promoted cell adhesion of different neuroendocrine cell types, these effects being mediated by both Epac and PKA and involving the same effector GTPase Rap1.
Collapse
Affiliation(s)
- E Vitali
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Clinical and Research Institute Humanitas, Rozzano, Italy
| | - V Cambiaghi
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Clinical and Research Institute Humanitas, Rozzano, Italy
| | - A Spada
- Fondazione IRCCS Ospedale Maggiore Policlinico, Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, University of Milan, Italy
| | - A Tresoldi
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Clinical and Research Institute Humanitas, Rozzano, Italy
| | - A Zerbi
- Pancreas Surgery Unit, IRCCS Humanitas Clinical Institute, Rozzano, Italy
| | - E Peverelli
- Fondazione IRCCS Ospedale Maggiore Policlinico, Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, University of Milan, Italy
| | - C Carnaghi
- Medical Oncology and Hematology Unit, Cancer Center, Humanitas Clinical and Research Center, Milan, Rozzano, Italy
| | - G Mantovani
- Fondazione IRCCS Ospedale Maggiore Policlinico, Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, University of Milan, Italy
| | - A G Lania
- Department of Biomedical Sciences, Humanitas University, Milan, Rozzano, Italy; Endocrinology Unit, Humanitas Research Hospital, Milan, Rozzano, Italy.
| |
Collapse
|
24
|
Tuncer S, Banerjee S. Eicosanoid pathway in colorectal cancer: Recent updates. World J Gastroenterol 2015; 21:11748-11766. [PMID: 26557000 PMCID: PMC4631974 DOI: 10.3748/wjg.v21.i41.11748] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/25/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
Enzymatic metabolism of the 20C polyunsaturated fatty acid (PUFA) arachidonic acid (AA) occurs via the cyclooxygenase (COX) and lipoxygenase (LOX) pathways, and leads to the production of various bioactive lipids termed eicosanoids. These eicosanoids have a variety of functions, including stimulation of homeostatic responses in the cardiovascular system, induction and resolution of inflammation, and modulation of immune responses against diseases associated with chronic inflammation, such as cancer. Because chronic inflammation is essential for the development of colorectal cancer (CRC), it is not surprising that many eicosanoids are implicated in CRC. Oftentimes, these autacoids work in an antagonistic and highly temporal manner in inflammation; therefore, inhibition of the pro-inflammatory COX-2 or 5-LOX enzymes may subsequently inhibit the formation of their essential products, or shunt substrates from one pathway to another, leading to undesirable side-effects. A better understanding of these different enzymes and their products is essential not only for understanding the importance of eicosanoids, but also for designing more effective drugs that solely target the inflammatory molecules found in both chronic inflammation and cancer. In this review, we have evaluated the cancer promoting and anti-cancer roles of different eicosanoids in CRC, and highlighted the most recent literature which describes how those molecules affect not only tumor tissue, but also the tumor microenvironment. Additionally, we have attempted to delineate the roles that eicosanoids with opposing functions play in neoplastic transformation in CRC through their effects on proliferation, apoptosis, motility, metastasis, and angiogenesis.
Collapse
|
25
|
Groen CM, Jayo A, Parsons M, Tootle TL. Prostaglandins regulate nuclear localization of Fascin and its function in nucleolar architecture. Mol Biol Cell 2015; 26:1901-17. [PMID: 25808493 PMCID: PMC4436834 DOI: 10.1091/mbc.e14-09-1384] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 03/18/2015] [Indexed: 01/14/2023] Open
Abstract
Fascin, a conserved actin-bundling protein, is not only cytoplasmic but also localizes to the nucleus and nuclear periphery in both Drosophila and mammalian cell contexts. In Drosophila, prostaglandin signaling regulates this localization. In addition, Fascin plays a critical role in nucleolar architecture in both Drosophila and mammalian cells. Fascin, a highly conserved actin-bundling protein, localizes and functions at new cellular sites in both Drosophila and multiple mammalian cell types. During Drosophila follicle development, in addition to being cytoplasmic, Fascin is in the nuclei of the germline-derived nurse cells during stages 10B–12 (S10B–12) and at the nuclear periphery during stage 13 (S13). This localization is specific to Fascin, as other actin-binding proteins, Villin and Profilin, do not exhibit the same subcellular distribution. In addition, localization of fascin1 to the nucleus and nuclear periphery is observed in multiple mammalian cell types. Thus the regulation and function of Fascin at these new cellular locations is likely to be highly conserved. In Drosophila, loss of prostaglandin signaling causes a global reduction in nuclear Fascin and a failure to relocalize to the nuclear periphery. Alterations in nuclear Fascin levels result in defects in nucleolar morphology in both Drosophila follicles and cultured mammalian cells, suggesting that nuclear Fascin plays an important role in nucleolar architecture. Given the numerous roles of Fascin in development and disease, including cancer, our novel finding that Fascin has functions within the nucleus sheds new light on the potential roles of Fascin in these contexts.
Collapse
Affiliation(s)
- Christopher M Groen
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Asier Jayo
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, United Kingdom
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, United Kingdom
| | - Tina L Tootle
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
26
|
Biocompatibility and inflammatory response in vitro and in vivo to gelatin-based biomaterials with tailorable elastic properties. Biomaterials 2014; 35:9755-9766. [DOI: 10.1016/j.biomaterials.2014.08.023] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/12/2014] [Indexed: 11/17/2022]
|
27
|
Dufour M, Faes S, Dormond-Meuwly A, Demartines N, Dormond O. PGE2-induced colon cancer growth is mediated by mTORC1. Biochem Biophys Res Commun 2014; 451:587-91. [PMID: 25128827 DOI: 10.1016/j.bbrc.2014.08.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/06/2014] [Indexed: 02/04/2023]
Abstract
The inflammatory prostaglandin E2 (PGE2) cytokine plays a key role in the development of colon cancer. Several studies have shown that PGE2 directly induces the growth of colon cancer cells and furthermore promotes tumor angiogenesis by increasing the production of the vascular endothelial growth factor (VEGF). The signaling intermediaries implicated in these processes have however not been fully characterized. In this report, we show that the mechanistic target of rapamycin complex 1 (mTORC1) plays an important role in PGE2-induced colon cancer cell responses. Indeed, stimulation of LS174T cells with PGE2 increased mTORC1 activity as observed by the augmentation of S6 ribosomal protein phosphorylation, a downstream effector of mTORC1. The PGE2 EP4 receptor was responsible for transducing the signal to mTORC1. Moreover, PGE2 increased colon cancer cell proliferation as well as the growth of colon cancer cell colonies grown in matrigel and blocking mTORC1 by rapamycin or ATP-competitive inhibitors of mTOR abrogated these effects. Similarly, the inhibition of mTORC1 by downregulation of its component raptor using RNA interference blocked PGE2-induced LS174T cell growth. Finally, stimulation of LS174T cells with PGE2 increased VEGF production which was also prevented by mTORC1 inhibition. Taken together, these results show that mTORC1 is an important signaling intermediary in PGE2 mediated colon cancer cell growth and VEGF production. They further support a role for mTORC1 in inflammation induced tumor growth.
Collapse
Affiliation(s)
- Marc Dufour
- Department of Visceral Surgery, University Hospital of Lausanne, Pavillon 4, Av. de Beaumont, 1011 Lausanne, Switzerland.
| | - Seraina Faes
- Department of Visceral Surgery, University Hospital of Lausanne, Pavillon 4, Av. de Beaumont, 1011 Lausanne, Switzerland.
| | - Anne Dormond-Meuwly
- Department of Visceral Surgery, University Hospital of Lausanne, Pavillon 4, Av. de Beaumont, 1011 Lausanne, Switzerland.
| | - Nicolas Demartines
- Department of Visceral Surgery, University Hospital of Lausanne, Pavillon 4, Av. de Beaumont, 1011 Lausanne, Switzerland.
| | - Olivier Dormond
- Department of Visceral Surgery, University Hospital of Lausanne, Pavillon 4, Av. de Beaumont, 1011 Lausanne, Switzerland.
| |
Collapse
|
28
|
Mena MP, Papiewska-Pajak I, Przygodzka P, Kozaczuk A, Boncela J, Cierniewski CS. NFAT2 regulates COX-2 expression and modulates the integrin repertoire in endothelial cells at the crossroads of angiogenesis and inflammation. Exp Cell Res 2014; 324:124-36. [PMID: 24657343 DOI: 10.1016/j.yexcr.2014.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 02/17/2014] [Accepted: 03/10/2014] [Indexed: 12/19/2022]
Abstract
The mechanisms controlling the switch between the pro-angiogenic and pro-inflammatory states of endothelial cells are still poorly understood. In this paper, we show that: (a) COX-2 expression induced by VEGF-A is NFAT2-dependent; and (b) the integrin profile in endothelial cells induced by the pro-angiogenic VEGF-A is distinct from that brought on by the inflammatory cytokine TNF-α. Two groups of integrin subunits specifically upregulated over time by both cytokines were identified using RT-PCR and Western Immunoblotting. The first group included α4, α5, α6, and β5 subunits that were upregulated by VEGF-A; the second group consisted of αV and β3 induced by TNF-α. Both cytokines significantly enhanced the expression of β1 and modulated α2 mRNA. In contrast to TNF-α, VEGF-A induction of integrin subunits depended on the activation of the calcineurin/NFAT pathway. Both calcineurin inhibitors (cyclosporineA and 11R-VIVIT) and downregulation of NFAT2 with specific siRNA decreased induction of integrin subunits. This process of induction could be increased by upregulation of NFAT2 by pBJ5-NFAT2 transfection. This suggests that NFAT2 mediates VEGF-induced upregulation of integrin subunit synthesis by providing a constant supply of newly synthesized "refreshed" mature integrin receptors, particularly α2β1, α5β1, α4β1, α6β1 and αVβ5, which are involved at different stages of angiogenesis.
Collapse
Affiliation(s)
- Mari-Pau Mena
- Institute of Medical Biology, Polish Academy of Science, Lodz 93-232, Poland
| | - Izabela Papiewska-Pajak
- Institute of Medical Biology, Polish Academy of Science, Lodz 93-232, Poland; Department of Molecular and Medical Biophysics, Medical University of Lodz, Lodz, Poland.
| | - Patrycja Przygodzka
- Institute of Medical Biology, Polish Academy of Science, Lodz 93-232, Poland
| | - Anna Kozaczuk
- Institute of Medical Biology, Polish Academy of Science, Lodz 93-232, Poland
| | - Joanna Boncela
- Institute of Medical Biology, Polish Academy of Science, Lodz 93-232, Poland
| | - Czeslaw S Cierniewski
- Institute of Medical Biology, Polish Academy of Science, Lodz 93-232, Poland; Department of Molecular and Medical Biophysics, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
29
|
Spracklen AJ, Kelpsch DJ, Chen X, Spracklen CN, Tootle TL. Prostaglandins temporally regulate cytoplasmic actin bundle formation during Drosophila oogenesis. Mol Biol Cell 2013; 25:397-411. [PMID: 24284900 PMCID: PMC3907279 DOI: 10.1091/mbc.e13-07-0366] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Tight regulation of actin remodeling is essential for development, and misregulation results in disease. Cytoskeletal dynamics are regulated by prostaglandins (PGs)—lipid signals. PGs temporally regulate actin remodeling during Drosophila oogenesis, at least in part, by modulating the activity of the actin elongation factor Enabled. Prostaglandins (PGs)—lipid signals produced downstream of cyclooxygenase (COX) enzymes—regulate actin dynamics in cell culture and platelets, but their roles during development are largely unknown. Here we define a new role for Pxt, the Drosophila COX-like enzyme, in regulating the actin cytoskeleton—temporal restriction of actin remodeling during oogenesis. PGs are required for actin filament bundle formation during stage 10B (S10B). In addition, loss of Pxt results in extensive early actin remodeling, including actin filaments and aggregates, within the posterior nurse cells of S9 follicles; wild-type follicles exhibit similar structures at a low frequency. Hu li tai shao (Hts-RC) and Villin (Quail), an actin bundler, localize to all early actin structures, whereas Enabled (Ena), an actin elongation factor, preferentially localizes to those in pxt mutants. Reduced Ena levels strongly suppress early actin remodeling in pxt mutants. Furthermore, loss of Pxt results in reduced Ena localization to the sites of bundle formation during S10B. Together these data lead to a model in which PGs temporally regulate actin remodeling during Drosophila oogenesis by controlling Ena localization/activity, such that in S9, PG signaling inhibits, whereas at S10B, it promotes Ena-dependent actin remodeling.
Collapse
Affiliation(s)
- Andrew J Spracklen
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242
| | | | | | | | | |
Collapse
|
30
|
Waclawik A, Kaczynski P, Jabbour HN. Autocrine and paracrine mechanisms of prostaglandin E₂ action on trophoblast/conceptus cells through the prostaglandin E₂ receptor (PTGER2) during implantation. Endocrinology 2013; 154:3864-76. [PMID: 23861370 DOI: 10.1210/en.2012-2271] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The conceptus and endometrium secrete large amounts of prostaglandin E₂ (PGE₂) into the porcine uterine lumen during the periimplantation period. We hypothesized that PGE₂ acts on conceptus/trophoblast cells through auto- and paracrine mechanisms. Real-time RT-PCR analysis revealed that PGE₂ receptor (PTGER)2 mRNA was 14-fold greater in conceptuses/trophoblasts on days 14-25 (implantation and early placentation period) vs preimplantation day 10-13 conceptuses (P < .05). Similarly, expression of PTGER2 protein increased during implantation. Conceptus expression of PTGER4 mRNA and protein did not differ on days 10-19. PGE₂ stimulated PTGER2 mRNA expression in day 15 trophoblast cells through PTGER2 receptor signaling. PGE₂ elevated aromatase expression and estradiol-17β secretion by trophoblast cells. Moreover, PGE₂ and the PTGER2 agonist, butaprost, increased the adhesive capacity of both human HTR-8/SVneo trophoblast and primary porcine trophoblast cells to extracellular matrix. This PGE₂-induced alteration in trophoblast cell adhesion to extracellular matrix was abolished by incubation of these cells with AH6809 (PTGER2 antagonist), ITGAVB3-directed tetrapeptide arg-gly-asp-ser or integrin ITGAVB3 antibody. PGE₂ stimulated adhesion of porcine trophoblast cells via the estrogen receptor and MEK/MAPK signaling pathway. PGE₂ induced phosphorylation of MAPK1/MAPK3 through PTGER2 and up-regulated expression of cell adhesion proteins such as focal adhesion kinase and intercellular adhesion molecule-1. Our study indicates that elevated PGE₂ in the periimplantation uterine lumen stimulates conceptus PTGER2 expression, which in turn promotes trophoblast adhesion via integrins, and synthesis and secretion of the porcine embryonic signal estradiol-17β. Moreover, the mechanism through which PGE₂ increases trophoblast adhesion is not species specific because it is PTGER2- and integrin-dependent in both porcine and human trophoblast cells.
Collapse
MESH Headings
- Animals
- Autocrine Communication/drug effects
- Cell Adhesion/drug effects
- Cell Line
- Cells, Cultured
- Crosses, Genetic
- Dinoprostone/agonists
- Dinoprostone/antagonists & inhibitors
- Dinoprostone/metabolism
- Embryo, Mammalian/drug effects
- Embryo, Mammalian/metabolism
- Embryonic Development/drug effects
- Estradiol/metabolism
- Extracellular Matrix/metabolism
- Female
- Gene Expression Regulation, Developmental/drug effects
- Humans
- Integrins/antagonists & inhibitors
- Integrins/metabolism
- MAP Kinase Signaling System/drug effects
- Paracrine Communication/drug effects
- Prostaglandin Antagonists/pharmacology
- Receptors, Prostaglandin E, EP2 Subtype/agonists
- Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Sus scrofa
- Trophoblasts/cytology
- Trophoblasts/drug effects
- Trophoblasts/metabolism
Collapse
Affiliation(s)
- Agnieszka Waclawik
- Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland.
| | | | | |
Collapse
|
31
|
Lee J, Banu SK, Burghardt RC, Starzinski-Powitz A, Arosh JA. Selective inhibition of prostaglandin E2 receptors EP2 and EP4 inhibits adhesion of human endometriotic epithelial and stromal cells through suppression of integrin-mediated mechanisms. Biol Reprod 2013; 88:77. [PMID: 23242524 DOI: 10.1095/biolreprod.112.100883] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Endometriosis is a chronic gynecological disease of reproductive age women characterized by the presence of functional endometrial tissues outside the uterine cavity. Interactions between the endometriotic cells and the peritoneal extracellular matrix proteins (ECM) are crucial mechanisms that allow adhesion of the endometriotic cells into peritoneal mesothelia. Prostaglandin E2 (PGE2) plays an important role in the pathogenesis of endometriosis. In previous studies, we have reported that selective inhibition of PGE2 receptors PTGER2 and PTGER4 decreases survival and invasion of human endometriotic epithelial and stromal cells through multiple mechanisms. Results of the present study indicates that selective inhibition of PTGER2- and PTGER4-mediated PGE2 signaling 1) decreases the expression and/or activity of specific integrin receptor subunits Itgb1 (beta1) and Itgb3 (beta3) but not Itgb5 (beta5), Itga1 (alpha1), Itga2 (alpha2), Itga5 (alpha5), and Itgav (alphav); 2) decreases integrin-signaling components focal adhesion kinase or protein kinase 2 (PTK2) and talin proteins; 3) inhibits interactions between Itgb1/Itgb3 subunits, PTK2, and talin and PTGER2/PTGER4 proteins through beta-arrestin-1 and Src kinase protein complex in human endometriotic epithelial cells 12Z and stromal cells 22B; and 4) decreases adhesion of 12Z and 22B cells to ECM collagen I, collagen IV, fibronectin, and vitronectin in a substrate-specific manner. These novel findings provide an important molecular framework for further evaluation of selective inhibition of PTGER2 and PTGER4 as potential nonsteroidal therapy to expand the spectrum of currently available treatment options for endometriosis in child-bearing age women.
Collapse
Affiliation(s)
- JeHoon Lee
- Reproductive Endocrinology and Cell Signaling Laboratory, Department of Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | | | | | | | | |
Collapse
|
32
|
Corti F, Finetti F, Ziche M, Simons M. The syndecan-4/protein kinase Cα pathway mediates prostaglandin E2-induced extracellular regulated kinase (ERK) activation in endothelial cells and angiogenesis in vivo. J Biol Chem 2013; 288:12712-21. [PMID: 23525101 DOI: 10.1074/jbc.m113.452383] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Prostaglandin E2 (PGE2) is regarded as the main mediator of inflammatory symptoms. In addition, it also plays an important role in tumor growth and angiogenesis. In this study, we examined the mechanism of PGE2-induced angiogenic response. We show that in the absence of proteoglycan syndecan-4 (Sdc4), PGE2-induced ERK activation is decreased significantly, as is endothelial cell migration and cord formation in a two-dimensional Matrigel assay. In vivo, PGE2-induced angiogenesis is reduced dramatically in Sdc4(-/-) mice. The mechanism was traced to Sdc4-dependent activation of protein kinase Cα (PKCα). Transduction of an Sdc4 S183E mutant (a cytoplasmic domain mutation that blocks Sdc4-dependent PKCα activation) into Sdc4(-/-) endothelial cells was not able to rescue the loss of PGE2-induced ERK activation, whereas a transduction with full-length Sdc4 resulted in full rescue. Furthermore, PGE2-induced angiogenesis was also reduced in PKCα(-/-) mice. Taken together, these results demonstrate that PGE2-induced activation of angiogenesis is mediated via syndecan-4-dependent activation of PKCα.
Collapse
Affiliation(s)
- Federico Corti
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | |
Collapse
|
33
|
Menon J, Doebele RC, Gomes S, Bevilacqua E, Reindl KM, Rosner MR. A novel interplay between Rap1 and PKA regulates induction of angiogenesis in prostate cancer. PLoS One 2012; 7:e49893. [PMID: 23166790 PMCID: PMC3499522 DOI: 10.1371/journal.pone.0049893] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 10/15/2012] [Indexed: 11/18/2022] Open
Abstract
Angiogenesis inhibition is an important therapeutic strategy for advanced stage prostate cancer. Previous work from our laboratory showed that sustained stimulation of Rap1 by 8-pCPT-2'-O-Me-cAMP (8CPT) via activation of Epac, a Rap1 GEF, or by expression of a constitutively active Rap1 mutant (cRap1) suppresses endothelial cell chemotaxis and subsequent angiogenesis. When we tested this model in the context of a prostate tumor xenograft, we found that 8CPT had no significant effect on prostate tumor growth alone. However, in cells harboring cRap1, 8CPT dramatically inhibited not only prostate tumor growth but also VEGF expression and angiogenesis within the tumor microenvironment. Subsequent analysis of the mechanism revealed that, in prostate tumor epithelial cells, 8CPT acted via stimulation of PKA rather than Epac/Rap1. PKA antagonizes Rap1 and hypoxic induction of 1α protein expression, VEGF production and, ultimately, angiogenesis. Together these findings provide evidence for a novel interplay between Rap1, Epac, and PKA that regulates tumor-stromal induction of angiogenesis.
Collapse
Affiliation(s)
- Jyotsana Menon
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, United States of America
| | - Robert C. Doebele
- School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Suzana Gomes
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, United States of America
| | - Elena Bevilacqua
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, United States of America
| | - Katie M. Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota, United States of America
| | - Marsha Rich Rosner
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
34
|
Eligini S, Songia P, Cavalca V, Crisci M, Tremoli E, Colli S. Cytoskeletal architecture regulates cyclooxygenase-2 in human endothelial cells: autocrine modulation by prostacyclin. J Cell Physiol 2012; 227:3847-56. [PMID: 22495438 DOI: 10.1002/jcp.24097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelium is a highly dynamic tissue that controls vascular homeostasis. This requires constant rearrangements of the shape or function of endothelial cells that cannot set aside the role of the cytoskeleton. The aim of this study was to determine the mechanisms by means of which cytoskeletal alterations induce cyclooxygenase-2 (Cox-2) expression in human endothelial cells using compounds that interfere with microtubule or actin architecture. Microtubule disruption by nocodazole markedly increased Cox-2 expression and activity, and provoked paracellular gap formation, a cardinal feature of endothelial barrier dysfunction. The Cox-2 metabolite prostacyclin down-regulated Cox-2 through an autocrine receptor-mediated mechanism, and partially prevented the disassembly of endothelial monolayers. There was also an interaction between microtubules and actin filaments in nocodazole-induced Cox-2 expression. Nocodazole provoked the dissolution of the F-actin cortical ring and stress fiber formation, increased actin glutathionylation, and concomitantly lowered intracellular levels of reduced glutathione. The restoration of glutathione levels by N-acetylcysteine opposed Cox-2 expression and preserved the integrity of endothelial monolayers. Among the signaling pathways connecting microtubule disruption with Cox-2 up-regulation, crucial roles are played by Src family kinase activation, serine/threonine phosphatase 2A inhibition, and the phosphorylation of mitogen activated protein kinase p38. Our findings provide a mechanistic insight into the observation that Cox-2 is induced in endothelial cells under cytoskeleton-perturbing conditions such as those occurring in the presence of atherogenic/inflammatory stimuli and oxidative stress. In this scenario, Cox-2 up-regulation by endothelia exposed to noxious conditions can be considered protective of the vasodilatory and anti-thrombotic properties of the vessel wall.
Collapse
|
35
|
Groen CM, Spracklen AJ, Fagan TN, Tootle TL. Drosophila Fascin is a novel downstream target of prostaglandin signaling during actin remodeling. Mol Biol Cell 2012; 23:4567-78. [PMID: 23051736 PMCID: PMC3510018 DOI: 10.1091/mbc.e12-05-0417] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Prostaglandins (PGs) regulate the actin cytoskeleton. However, their mechanisms of action are unknown. Use of Drosophila oogenesis—specifically nurse cell dumping—as a model shows that PGs regulate the actin bundler Fascin to control parallel actin filament bundle formation and cortical actin integrity. Although prostaglandins (PGs)—lipid signals produced downstream of cyclooxygenase (COX) enzymes—regulate actin cytoskeletal dynamics, their mechanisms of action are unknown. We previously established Drosophila oogenesis, in particular nurse cell dumping, as a new model to determine how PGs regulate actin remodeling. PGs, and thus the Drosophila COX-like enzyme Pxt, are required for both the parallel actin filament bundle formation and the cortical actin strengthening required for dumping. Here we provide the first link between Fascin (Drosophila Singed, Sn), an actin-bundling protein, and PGs. Loss of either pxt or fascin results in similar actin defects. Fascin interacts, both pharmacologically and genetically, with PGs, as reduced Fascin levels enhance the effects of COX inhibition and synergize with reduced Pxt levels to cause both parallel bundle and cortical actin defects. Conversely, overexpression of Fascin in the germline suppresses the effects of COX inhibition and genetic loss of Pxt. These data lead to the conclusion that PGs regulate Fascin to control actin remodeling. This novel interaction has implications beyond Drosophila, as both PGs and Fascin-1, in mammalian systems, contribute to cancer cell migration and invasion.
Collapse
Affiliation(s)
- Christopher M Groen
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
36
|
Shah T, Stasinopoulos I, Wildes F, Kakkad S, Artemov D, Bhujwalla ZM. Noninvasive imaging identifies new roles for cyclooxygenase-2 in choline and lipid metabolism of human breast cancer cells. NMR IN BIOMEDICINE 2012; 25:746-54. [PMID: 21953546 PMCID: PMC4337877 DOI: 10.1002/nbm.1789] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 07/27/2011] [Accepted: 08/12/2011] [Indexed: 05/19/2023]
Abstract
The expression of cyclooxygenase-2 (COX-2) is observed in approximately 40% of breast cancers. A major product of the COX-2-catalyzed reaction, prostaglandin E(2), is an inflammatory mediator that participates in several biological processes, and influences invasion, vascularization and metastasis. Using noninvasive MRI and MRS, we determined the effect of COX-2 downregulation on the metabolism and invasion of intact poorly differentiated MDA-MB-231 human breast cancer cells stably expressing COX-2 short hairpin RNA. Dynamic tracking of invasion, extracellular matrix degradation and metabolism was performed with an MRI- and MRS-compatible cell perfusion assay under controlled conditions of pH, temperature and oxygenation over the course of 48 h. COX-2-silenced cells exhibited a significant decrease in invasion relative to parental cells that was consistent with the reduced expression of invasion-associated matrix metalloproteinase genes and an increased level of the tissue inhibitor of metalloproteinase-1. We identified, for the first time, a role for COX-2 in mediating changes in choline phospholipid metabolism, and established that choline kinase expression is partly dependent on COX-2 function. COX-2 silencing resulted in a significant decrease in phosphocholine and total choline that was detected by MRS. In addition, a significant increase in lipids, as well as lipid droplet formation, was observed. COX-2 silencing transformed parental cell metabolite patterns to those characteristic of less aggressive cancer cells. These new functional roles of COX-2 may identify new biomarkers and new targets for use in combination with COX-2 targeting to prevent invasion and metastasis.
Collapse
Affiliation(s)
| | | | | | | | | | - Zaver M. Bhujwalla
- Correspondence to: Z. M. Bhujwalla, Department of Radiology, Johns Hopkins University School of Medicine, 208C Traylor Bldg., 720 Rutland Ave., Baltimore, MD 21205, USA.
| |
Collapse
|
37
|
Prostaglandins in cancer cell adhesion, migration, and invasion. Int J Cell Biol 2012; 2012:723419. [PMID: 22505934 PMCID: PMC3299390 DOI: 10.1155/2012/723419] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 10/08/2011] [Indexed: 12/21/2022] Open
Abstract
Prostaglandins exert a profound influence over the adhesive, migratory, and invasive behavior of cells during the development and progression of cancer. Cyclooxygenase-2 (COX-2) and microsomal prostaglandin E2 synthase-1 (mPGES-1) are upregulated in inflammation and cancer. This results in the production of prostaglandin E2 (PGE2), which binds to and activates G-protein-coupled prostaglandin E1–4 receptors (EP1–4). Selectively targeting the COX-2/mPGES-1/PGE2/EP1–4 axis of the prostaglandin pathway can reduce the adhesion, migration, invasion, and angiogenesis. Once stimulated by prostaglandins, cadherin adhesive connections between epithelial or endothelial cells are lost. This enables cells to invade through the underlying basement membrane and extracellular matrix (ECM). Interactions with the ECM are mediated by cell surface integrins by “outside-in signaling” through Src and focal adhesion kinase (FAK) and/or “inside-out signaling” through talins and kindlins. Combining the use of COX-2/mPGES-1/PGE2/EP1–4 axis-targeted molecules with those targeting cell surface adhesion receptors or their downstream signaling molecules may enhance cancer therapy.
Collapse
|
38
|
Liu L, Cao Z, Chen J, Li R, Cao Y, Zhu C, Wu K, Wu J, Liu F, Zhu Y. Influenza A virus induces interleukin-27 through cyclooxygenase-2 and protein kinase A signaling. J Biol Chem 2012; 287:11899-910. [PMID: 22343630 DOI: 10.1074/jbc.m111.308064] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We previously reported that IL-27, which belongs to the IL-12 family of cytokines, is elevated in the serum of patients infected with influenza A virus (IAV). Here, we show that the expression of IL-27 was significantly up-regulated in A549 human lung epithelial cells and human peripheral blood mononuclear cells infected with IAV. Additionally, IAV triggered IL-27 expression through protein kinase A and cAMP-response element-binding protein signaling, which was mediated by cyclooxygenase-2-derived prostaglandin E(2). IL-27 inhibited IAV replication by STAT1/2/3 phosphorylation and activated antiviral factor protein kinase R phosphorylation. Clinical analysis showed that IL-27 levels were significantly elevated in a cohort of patients infected with IAV compared with healthy individuals and that circulating IL-27 levels were tightly and positively correlated with prostaglandin E(2) levels. These results indicate that IL-27 expression is one host immune factor produced in response to IAV infection and that elevated IL-27 levels inhibit viral replication.
Collapse
Affiliation(s)
- Li Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sancho P, Martín-Sanz P, Fabregat I. Reciprocal regulation of NADPH oxidases and the cyclooxygenase-2 pathway. Free Radic Biol Med 2011; 51:1789-98. [PMID: 21907277 DOI: 10.1016/j.freeradbiomed.2011.08.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 08/03/2011] [Accepted: 08/11/2011] [Indexed: 01/05/2023]
Abstract
The objective of this work was to analyze the possible association between cyclooxygenase-2 (COX-2) and NADPH oxidases (NOX) in liver cells, in response to various proinflammatory and toxic insults. First, we observed that treatment of Chang liver (CHL) cells with various COX-2 inducers increased reactive oxygen species (ROS) production concomitant with GSH depletion, phorbol 12-myristate 13-acetate (PMA) being the most effective treatment. Moreover, early changes in the oxidative status induced by PMA were inhibited by glutathione ethyl ester, which also impeded COX-2 induction. In fact, CHL cells expressed NOX1 and NOX4, although only NOX4 expression was up-regulated in the presence of PMA. Knock-down experiments suggested that PMA initiated a pathway in which NOX1 activation controlled COX-2 expression and activity, which, in turn, induced NOX4 expression by activation of the prostaglandin receptor EP4. In addition, CHL cells overexpressing COX-2 showed higher NOX4 expression and ROS content, which were decreased in the presence of the COX-2 inhibitor DFU. Interestingly, we found that addition of prostaglandin E(2) (PGE(2)) also induced NOX4 expression and ROS production, which might promote cell adhesion. Finally, we determined that NOX4 induction by PGE(2) was dependent on ERK1/2 signaling. Taken together, these results indicate that NOX proteins and COX-2 are reciprocally regulated in liver cells.
Collapse
Affiliation(s)
- Patricia Sancho
- Biological Clues of the Invasive and Metastatic Phenotype Group, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | | | | |
Collapse
|
40
|
Integrin-mediated cell-matrix interaction in physiological and pathological blood vessel formation. JOURNAL OF ONCOLOGY 2011; 2012:125278. [PMID: 21941547 PMCID: PMC3175391 DOI: 10.1155/2012/125278] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 07/15/2011] [Indexed: 02/07/2023]
Abstract
Physiological as well as pathological blood vessel formation are fundamentally dependent on cell-matrix interaction. Integrins, a family of major cell adhesion receptors, play a pivotal role in development, maintenance, and remodeling of the vasculature. Cell migration, invasion, and remodeling of the extracellular matrix (ECM) are integrin-regulated processes, and the expression of certain integrins also correlates with tumor progression. Recent advances in the understanding of how integrins are involved in the regulation of blood vessel formation and remodeling during tumor progression are highlighted. The increasing knowledge of integrin function at the molecular level, together with the growing repertoire of integrin inhibitors which allow their selective pharmacological manipulation, makes integrins suited as potential diagnostic markers and therapeutic targets.
Collapse
|
41
|
Harmine inhibits tumour specific neo-vessel formation by regulating VEGF, MMP, TIMP and pro-inflammatory mediators both in vivo and in vitro. Eur J Pharmacol 2010; 649:64-73. [DOI: 10.1016/j.ejphar.2010.09.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 08/21/2010] [Accepted: 09/09/2010] [Indexed: 11/22/2022]
|
42
|
Grape seed extract ameliorates tumor necrosis factor-α-induced inflammatory status of human umbilical vein endothelial cells. Eur J Nutr 2010; 50:401-9. [PMID: 21113812 DOI: 10.1007/s00394-010-0151-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 11/15/2010] [Indexed: 12/17/2022]
Abstract
BACKGROUND Inflammation has played a key role in the causation of atherosclerosis. However, the effects of grape seed extract (GSE) on the pro-inflammatory intracellular signaling, enzyme activity, and inflammatory mediators of endothelial cells have not been sufficiently studied, and less information exists on the comparison between GSE and vitamin C, a well-known antioxidant compound, on their anti-inflammatory properties. PURPOSE We investigated the effects of GSE and vitamin C on the cell viability, oxidative stress, monocyte adhesion, the expression of nuclear factor-κB inhibitor (IκB), intercellular adhesion molecule-1 (ICAM-1) and cyclooxygenase-2 (COX-2), and the production of prostaglandin E(2) (PG E(2)) in TNF-α-treated human umbilical vein endothelial cells (HUVECs). METHODS Cell viability was measured by MTT assay. The adhesion of THP-1 to HUVECs was evaluated by cell adhesion assay. The oxidized nucleoside 8-hydroxydeoxyguanosine (8-OHdG) (an indicator of oxidative damage to DNA), ICAM-1, and PG E(2) were measured by ELISA. IκB and COX-2 expression were evaluated by western blot analysis. RESULTS TNF-α (10, 20, and 50 ng/mL), GSE (50 and 200 μg/mL), or vitamin C (100 μM) did not affect cell viability. GSE (50-100 μg/mL) attenuated TNF-α (20 ng/mL)-induced 8-OHdG production, THP-1 adhesion, the expression of IκB degradation, ICAM-1 and COX-2, and the production of PGE(2) in a dose-dependent manner. Vitamin C (100 μM) also showed significant antioxidative and anti-inflammatory effects. CONCLUSIONS GSE effectively ameliorates TNF-α-induced inflammatory status of HUVECs. The findings of the present study suggest that consumption of GSE may be beneficial to inflammatory atherosclerosis.
Collapse
|
43
|
Malik G, Knowles LM, Dhir R, Xu S, Yang S, Ruoslahti E, Pilch J. Plasma fibronectin promotes lung metastasis by contributions to fibrin clots and tumor cell invasion. Cancer Res 2010; 70:4327-34. [PMID: 20501851 DOI: 10.1158/0008-5472.can-09-3312] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The attachment of circulating tumor cells to the blood vessels of distant organs is an important step in metastasis. We show here that experimental lung metastasis by two cell lines, B16F1 melanoma and 3LL lung carcinoma, is greatly reduced in transgenic mice that lack plasma fibronectin. This multifunctional adhesive glycoprotein becomes cross-linked to fibrin during clotting. Here, we report that eliminating plasma fibronectin from the blood circulation reverses the prometastatic effects of blood clotting and tumor cell integrin alphavbeta3. In vitro studies showed that fibrin-fibronectin complexes, but not purified fibrin, supported tumor cell attachment and invasion. These functions correlate with the ability of fibrin-fibronectin complexes to induce the activation of integrin alphavbeta3. Our findings reveal an important contribution of plasma fibronectin in lung metastasis. Furthermore, they suggest that the previously noted effects of blood clotting on lung metastasis might be mediated in part by a fibronectin-alphavbeta3 integrin axis, in which plasma fibronectin has to be incorporated into the blood clot.
Collapse
Affiliation(s)
- Gunjan Malik
- Departments of Urology and Pathology, University of Pittsburgh School of Medicine, Shadyside Medical Center, Prostate Cancer Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Antiangiogenic Effect of a Selective 5-HT4 Receptor Agonist. J Surg Res 2010; 159:696-704. [DOI: 10.1016/j.jss.2008.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 10/27/2008] [Accepted: 11/03/2008] [Indexed: 01/24/2023]
|
45
|
Abstract
Eicosanoids, including prostaglandins and leukotrienes, are biologically active lipids that have been implicated in various pathological processes, such as inflammation and cancer. This Review highlights our understanding of the intricate roles of eicosanoids in epithelial-derived tumours and their microenvironment. The knowledge of how these lipids orchestrate the complex interactions between transformed epithelial cells and the surrounding stromal cells is crucial for understanding tumour evolution, progression and metastasis. Understanding the molecular mechanisms underlying the role of prostaglandins and other eicosanoids in cancer progression will help to develop more effective cancer chemopreventive and/or therapeutic agents.
Collapse
Affiliation(s)
- Dingzhi Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | | |
Collapse
|
46
|
Sharma-Walia N, Paul AG, Bottero V, Sadagopan S, Veettil MV, Kerur N, Chandran B. Kaposi's sarcoma associated herpes virus (KSHV) induced COX-2: a key factor in latency, inflammation, angiogenesis, cell survival and invasion. PLoS Pathog 2010; 6:e1000777. [PMID: 20169190 PMCID: PMC2820536 DOI: 10.1371/journal.ppat.1000777] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Accepted: 01/19/2010] [Indexed: 12/22/2022] Open
Abstract
Kaposi's sarcoma (KS), an enigmatic endothelial cell vascular neoplasm, is characterized by the proliferation of spindle shaped endothelial cells, inflammatory cytokines (ICs), growth factors (GFs) and angiogenic factors. KSHV is etiologically linked to KS and expresses its latent genes in KS lesion endothelial cells. Primary infection of human micro vascular endothelial cells (HMVEC-d) results in the establishment of latent infection and reprogramming of host genes, and cyclooxygenase-2 (COX-2) is one of the highly up-regulated genes. Our previous study suggested a role for COX-2 in the establishment and maintenance of KSHV latency. Here, we examined the role of COX-2 in the induction of ICs, GFs, angiogenesis and invasive events occurring during KSHV de novo infection of endothelial cells. A significant amount of COX-2 was detected in KS tissue sections. Telomerase-immortalized human umbilical vein endothelial cells supporting KSHV stable latency (TIVE-LTC) expressed elevated levels of functional COX-2 and microsomal PGE2 synthase (m-PGES), and secreted the predominant eicosanoid inflammatory metabolite PGE2. Infected HMVEC-d and TIVE-LTC cells secreted a variety of ICs, GFs, angiogenic factors and matrix metalloproteinases (MMPs), which were significantly abrogated by COX-2 inhibition either by chemical inhibitors or by siRNA. The ability of these factors to induce tube formation of uninfected endothelial cells was also inhibited. PGE2, secreted early during KSHV infection, profoundly increased the adhesion of uninfected endothelial cells to fibronectin by activating the small G protein Rac1. COX-2 inhibition considerably reduced KSHV latent ORF73 gene expression and survival of TIVE-LTC cells. Collectively, these studies underscore the pivotal role of KSHV induced COX-2/PGE2 in creating KS lesion like microenvironment during de novo infection. Since COX-2 plays multiple roles in KSHV latent gene expression, which themselves are powerful mediators of cytokine induction, anti-apoptosis, cell survival and viral genome maintainence, effective inhibition of COX-2 via well-characterized clinically approved COX-2 inhibitors could potentially be used in treatment to control latent KSHV infection and ameliorate KS. Kaposi's sarcoma associated herpes virus (KSHV), with a 160 kb DNA genome, has evolved with two distinct life cycle phases, namely latency and lytic replication. KS, a complex angioproliferative disease, is regulated by a balance between pro-angiogenic and anti-angiogenic factors. In our previous study, we showed that KSHV modulates host factors COX-2/PGE2 for its own advantage to promote its latent (persistent) infection. The premise that COX-2 is involved in growth and progression of several types of solid cancers and inflammation associated diseases has been well documented but has never been studied in KS. Here, utilizing COX-2 inhibition strategies, including chemical inhibition and a gene silencing approach, we systematically identified the potential role of KSHV induced COX-2/PGE2 in viral pathogenesis related events such as secretion of inflammatory and angiogenic cytokines, MMPs and cell adhesion in de novo infected or latently infected endothelial cells. We report that COX-2/PGE2 inhibition down-regulates viral latent gene expression and survival of latently infected endothelial cells. The data emanating from our in vitro studies is valuable, informative and requires further examination using an in vitro angiogenic model and in vivo nude mice model to further validate COX-2 as a novel therapeutic to target latent infection and the associated diseases like KS.
Collapse
Affiliation(s)
- Neelam Sharma-Walia
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America.
| | | | | | | | | | | | | |
Collapse
|
47
|
Choi SH, Aid S, Choi U, Bosetti F. Cyclooxygenases-1 and -2 differentially modulate leukocyte recruitment into the inflamed brain. THE PHARMACOGENOMICS JOURNAL 2009; 10:448-57. [PMID: 20038958 DOI: 10.1038/tpj.2009.68] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Peripheral leukocyte recruitment in neuroinflammatory conditions can exacerbate brain tissue damage by releasing cytotoxic mediators and by increasing vascular permeability. Cyclooxygenase (COX)-derived prostaglandins promote the migration of several immune cells in vitro, however, the specific roles of COX-1 and -2 on leukocyte recruitment in vivo have not been investigated. To examine the specific effects of COX-1 or COX-2 deficiency on neuroinflammation-induced leukocyte infiltration, we used a model of intracerebroventricular lipopolysaccharide (LPS)-induced neuroinflammation in COX-1(-/-), COX-2(-/-), and their respective wild-type (WT) ((+/+)) mice. After LPS, leukocyte infiltration and inflammatory response were attenuated in COX-1(-/-) and increased in COX-2(-/-) mice, compared with their respective WT controls. This influx of leukocytes was accompanied by a marked disruption of blood-brain barrier and differential expression of chemokines. These results indicate that COX-1 and COX-2 deletion differentially modulate leukocyte recruitment during neuroinflammation, and suggest that inhibition of COX-1 activity is beneficial, whereas COX-2 inhibition is detrimental, during a primary neuroinflammatory response.
Collapse
Affiliation(s)
- S-H Choi
- Molecular Neuroscience Unit, Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
48
|
Kawabe JI, Yuhki KI, Okada M, Kanno T, Yamauchi A, Tashiro N, Sasaki T, Okumura S, Nakagawa N, Aburakawa Y, Takehara N, Fujino T, Hasebe N, Narumiya S, Ushikubi F. Prostaglandin I2 promotes recruitment of endothelial progenitor cells and limits vascular remodeling. Arterioscler Thromb Vasc Biol 2009; 30:464-70. [PMID: 20007911 DOI: 10.1161/atvbaha.109.193730] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Endothelial progenitor cells (EPCs) play an important role in the self-healing of a vascular injury by participating in the reendothelialization that limits vascular remodeling. We evaluated whether prostaglandin I(2) plays a role in the regulation of the function of EPCs to limit vascular remodeling. METHODS AND RESULTS EPCs (Lin(-)cKit(+)Flk-1(+) cells) were isolated from the bone marrow (BM) of wild-type (WT) mice or mice lacking the prostaglandin I(2) receptor IP (IP(-/-) mice). Reverse transcription-polymerase chain reaction analysis showed that EPCs among BM cells specifically express IP. The cellular properties of EPCs, adhesion, migration, and proliferation on fibronectin were significantly attenuated in IP-deficient EPCs compared with WT EPCs. In contrast, IP agonists facilitated these functions in WT EPCs, but not in IP-deficient EPCs. The specific deletion of IP in BM cells, which was performed by transplanting BM cells of IP(-/-) mice to WT mice, accelerated wire injury-mediated neointimal hyperplasia in the femoral artery. Notably, transfused WT EPCs, but not IP-deficient EPCs, were recruited to the injured vessels, participated in reendothelialization, and efficiently rescued the accelerated vascular remodeling. CONCLUSIONS These findings clearly indicate that the prostaglandin I(2)-IP system is essential for EPCs to accomplish their function and plays a critical role in the regulation of vascular remodeling.
Collapse
Affiliation(s)
- Jun-Ichi Kawabe
- Department of Pharmacology, Asahikawa Medical College, Midorigaoka Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Amano H, Ito Y, Suzuki T, Kato S, Matsui Y, Ogawa F, Murata T, Sugimoto Y, Senior R, Kitasato H, Hayashi I, Satoh Y, Narumiya S, Majima M. Roles of a prostaglandin E-type receptor, EP3, in upregulation of matrix metalloproteinase-9 and vascular endothelial growth factor during enhancement of tumor metastasis. Cancer Sci 2009; 100:2318-24. [PMID: 19799610 PMCID: PMC11159505 DOI: 10.1111/j.1349-7006.2009.01322.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Cyclooxygenase (COX)-2 is known to correlate with poor cancer prognosis and to contribute to tumor metastasis. However, the precise mechanism of this phenomenon remains unknown. We have previously reported that host stromal prostaglandin E(2) (PGE(2))-prostaglandin E2 receptor (EP)3 signaling appears critical for tumor-associated angiogenesis and tumor growth. Here we tested whether the EP3 receptor has a critical role in tumor metastasis. Lewis lung carcinoma (LLC) cells were intravenously injected into WT mice and mice treated with the COX-2 inhibitor NS-398. The nonselective COX inhibitor aspirin reduced lung metastasis, but the COX-1 inhibitor SC560 did not. The expression of matrix metalloproteinases (MMP)-9 and vascular endothelial growth factor (VEGF)-A was suppressed in NS-398-treated mice compared with PBS-treated mice. Lungs containing LLC colonies were markedly reduced in EP3 receptor knockout (EP3(-/-)) mice compared with WT mice. The expression of MMP-9 and VEGF-A was downregulated in metastatic lungs of EP3(-/-) mice. An immunohistochemical study revealed that MMP-9-expressing endothelial cells were markedly reduced in EP3(-/-) mice compared with WT mice. When HUVEC were treated with agonists for EP1, EP2, EP3, or EP4, only the EP3 agonist enhanced MMP-9 expression. These results suggested that EP3 receptor signaling on endothelial cells is essential for the MMP-9 upregulation that enhances tumor metastasis and angiogenesis. An EP3 receptor antagonist may be useful to protect against tumor metastasis.
Collapse
Affiliation(s)
- Hideki Amano
- Department of Pharmacology, Kitasato University School of Medicine, Kanagawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Côté SC, Pasvanis S, Bounou S, Dumais N. CCR7-specific migration to CCL19 and CCL21 is induced by PGE(2) stimulation in human monocytes: Involvement of EP(2)/EP(4) receptors activation. Mol Immunol 2009; 46:2682-93. [PMID: 19545899 DOI: 10.1016/j.molimm.2008.08.269] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 08/12/2008] [Accepted: 08/13/2008] [Indexed: 11/16/2022]
Abstract
The recent demonstration that newly recruited monocytes do not die at the site of inflammation, but migrate to draining lymph nodes, raises the question on the mechanism involved in this process. In this study, we demonstrate for the first time that prostaglandin E(2) (PGE(2)) regulates the expression and the activity of CCR7 in human blood-isolated monocytes as well as in the MONO-MAC-1 cell lineage. PGE(2) induces intracellular cAMP formation through engagement of the E-prostanoid 2/E-prostanoid 4 (EP(2)/EP(4)) receptors present on monocytes. Migration to chemokines CCL19 and CCL21 in the PGE(2)-stimulated monocytes is mediated through the augmentation of cAMP concentration and furthermore, the cAMP/PKA pathway appears to act as the major inducer of CCR7 transcription in MONO-MAC-1. While p38 MAPK was induced by PGE(2), we observed that PGE(2) can downregulate p42/p44 MAPK phosphorylation. At the transcription level, inhibition of p38 MAPK inhibits CCR7 mRNA expression. Finally, we demonstrated that transcription factors CREB-1 and C/EBPalpha and C/EBPbeta are translocated to the nucleus following PGE(2) stimulation and bind the potent CCR7 promoter region. Our findings may have important implication for HIV-1 migration to the lymph nodes since macrophages and monocytes, particularly CD16 positive subset, are susceptible to HIV-1 infection.
Collapse
Affiliation(s)
- Sandra C Côté
- Département de Biologie, Université de Sherbrooke, QC, Canada
| | | | | | | |
Collapse
|