1
|
Wu Y, Pernet E, Touqui L. Modulation of Airway Expression of the Host Bactericidal Enzyme, sPLA2-IIA, by Bacterial Toxins. Toxins (Basel) 2023; 15:440. [PMID: 37505708 PMCID: PMC10467128 DOI: 10.3390/toxins15070440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/23/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
Host molecules with antimicrobial properties belong to a large family of mediators including type-IIA secreted phospholipase A2 (sPLA2-IIA). The latter is a potent bactericidal agent with high selectivity against Gram-positive bacteria, but it may also play a role in modulating the host inflammatory response. However, several pathogen-associated molecular patterns (PAMPs) or toxins produced by pathogenic bacteria can modulate the levels of sPLA2-IIA by either inducing or inhibiting its expression in host cells. Thus, the final sPLA2-IIA concentration during the infection process is determined by the orchestration between the levels of toxins that stimulate and those that downregulate the expression of this enzyme. The stimulation of sPLA2-IIA expression is a process that participates in the clearance of invading bacteria, while inhibition of this expression highlights a mechanism by which certain bacteria can subvert the immune response and invade the host. Here, we will review the major functions of sPLA2-IIA in the airways and the role of bacterial toxins in modulating the expression of this enzyme. We will also summarize the major mechanisms involved in this modulation and the potential consequences for the pulmonary host response to bacterial infection.
Collapse
Affiliation(s)
- Yongzheng Wu
- Unité de Biologie Cellulaire de l’Infection Microbionne, CNRS UMR3691, Institut Pasteur, Université de Paris Cité, 75015 Paris, France;
| | - Erwan Pernet
- Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - Lhousseine Touqui
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
- Institut Pasteur, Université de Paris Cité, Mucoviscidose et Bronchopathies Chroniques, 75015 Paris, France
| |
Collapse
|
2
|
Doré E, Joly-Beauparlant C, Morozumi S, Mathieu A, Lévesque T, Allaeys I, Duchez AC, Cloutier N, Leclercq M, Bodein A, Payré C, Martin C, Petit-Paitel A, Gelb MH, Rangachari M, Murakami M, Davidovic L, Flamand N, Arita M, Lambeau G, Droit A, Boilard E. The interaction of secreted phospholipase A2-IIA with the microbiota alters its lipidome and promotes inflammation. JCI Insight 2022; 7:152638. [PMID: 35076027 PMCID: PMC8855825 DOI: 10.1172/jci.insight.152638] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022] Open
Abstract
Secreted phospholipase A2-IIA (sPLA2-IIA) hydrolyzes phospholipids to liberate lysophospholipids and fatty acids. Given its poor activity toward eukaryotic cell membranes, its role in the generation of proinflammatory lipid mediators is unclear. Conversely, sPLA2-IIA efficiently hydrolyzes bacterial membranes. Here, we show that sPLA2-IIA affects the immune system by acting on the intestinal microbial flora. Using mice overexpressing transgene-driven human sPLA2-IIA, we found that the intestinal microbiota was critical for both induction of an immune phenotype and promotion of inflammatory arthritis. The expression of sPLA2-IIA led to alterations of the intestinal microbiota composition, but housing in a more stringent pathogen-free facility revealed that its expression could affect the immune system in the absence of changes to the composition of this flora. In contrast, untargeted lipidomic analysis focusing on bacteria-derived lipid mediators revealed that sPLA2-IIA could profoundly alter the fecal lipidome. The data suggest that a singular protein, sPLA2-IIA, produces systemic effects on the immune system through its activity on the microbiota and its lipidome.
Collapse
Affiliation(s)
- Etienne Doré
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| | - Charles Joly-Beauparlant
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Satoshi Morozumi
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Alban Mathieu
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Tania Lévesque
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| | - Isabelle Allaeys
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| | - Anne-Claire Duchez
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
| | - Nathalie Cloutier
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
| | - Mickaël Leclercq
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Antoine Bodein
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Christine Payré
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Cyril Martin
- The Research Center of the University Institute of Cardiology and Pneumology of Quebec, Quebec City, Quebec, Canada
| | - Agnes Petit-Paitel
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Michael H. Gelb
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Manu Rangachari
- CHU de Québec-Université Laval Research Center, Neurosciences Axis, Quebec City, Quebec, Canada
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Laetitia Davidovic
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Nicolas Flamand
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
- The Research Center of the University Institute of Cardiology and Pneumology of Quebec, Quebec City, Quebec, Canada
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama-City University, Yokohama, Japan
| | - Gérard Lambeau
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Arnaud Droit
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Eric Boilard
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| |
Collapse
|
3
|
Kuefner MS, Stephenson E, Savikj M, Smallwood HS, Dong Q, Payré C, Lambeau G, Park EA. Group IIA secreted phospholipase A2 (PLA2G2A) augments adipose tissue thermogenesis. FASEB J 2021; 35:e21881. [PMID: 34478587 DOI: 10.1096/fj.202002481rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022]
Abstract
Group IIA secreted phospholipase A2 (PLA2G2A) hydrolyzes glycerophospholipids at the sn-2 position resulting in the release of fatty acids and lysophospholipids. C57BL/6 mice do not express Pla2g2a due to a frameshift mutation (wild-type [WT] mice). We previously reported that transgenic expression of human PLA2G2A in C57BL/6 mice (IIA+ mice) protects against weight gain and insulin resistance, in part by increasing total energy expenditure. Additionally, we found that brown and white adipocytes from IIA+ mice have increased expression of mitochondrial uncoupling markers, such as uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor-gamma coactivator, and PR domain containing 16, suggesting that the energy expenditure phenotype might be due to an increased thermogenic capacity in adipose tissue. Here, we further characterize the impact of PLA2G2A on thermogenic mechanisms in adipose tissue. Metabolic analysis of WT and IIA+ mice revealed that even when housed within their thermoneutral zone, IIA+ mice have elevated energy expenditure compared to WT littermates. Increased energy expenditure in IIA+ mice is associated with increased citrate synthase activity in brown adipose tissue (BAT) and increased mitochondrial respiration in both brown and white adipocytes. We also observed that direct addition of recombinant PLA2G2A enzyme to in vitro cultured adipocytes results in the marked induction of UCP1 protein expression. Finally, we report that PLA2G2A induces the expression of numerous transcripts related to energy substrate transport and metabolism in BAT, suggestive of an increase in substrate flux to fuel BAT activity. These data demonstrate that PLA2G2A enhances adipose tissue thermogenesis, in part, through elevated substrate delivery and increased mitochondrial content in BAT.
Collapse
Affiliation(s)
- Michael S Kuefner
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Erin Stephenson
- Department of Anatomy, College of Graduate Studies and Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois, USA
| | - Mladen Savikj
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Heather S Smallwood
- Department of Pharmacology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Qingming Dong
- Department of Pharmacology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Veterans Affairs Medical Center, Memphis, Tennessee, USA
| | - Christine Payré
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne Sophia Antipolis, France
| | - Gérard Lambeau
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne Sophia Antipolis, France
| | - Edwards A Park
- Department of Pharmacology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Veterans Affairs Medical Center, Memphis, Tennessee, USA
| |
Collapse
|
4
|
Kuefner MS. Secretory Phospholipase A2s in Insulin Resistance and Metabolism. Front Endocrinol (Lausanne) 2021; 12:732726. [PMID: 34512555 PMCID: PMC8429832 DOI: 10.3389/fendo.2021.732726] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023] Open
Abstract
The phospholipases A2 (PLA2) superfamily encompasses enzymes commonly found in mammalian tissues and snake venom. Many of these enzymes have unique tissue distribution, function, and substrate specificity suggesting distinct biological roles. In the past, much of the research on secretory PLA2s has analyzed their roles in inflammation, anti-bacterial actions, and atherosclerosis. In recent studies utilizing a variety of mouse models, pancreatic islets, and clinical trials, a role for many of these enzymes in the control of metabolism and insulin action has been revealed. In this review, this research, and the unique contributions of the PLA2 enzymes in insulin resistance and metabolism.
Collapse
|
5
|
Dore E, Boilard E. Roles of secreted phospholipase A 2 group IIA in inflammation and host defense. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:789-802. [PMID: 30905346 DOI: 10.1016/j.bbalip.2018.08.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 01/08/2023]
Abstract
Among all members of the secreted phospholipase A2 (sPLA2) family, group IIA sPLA2 (sPLA2-IIA) is possibly the most studied enzyme. Since its discovery, many names have been associated with sPLA2-IIA, such as "non-pancreatic", "synovial", "platelet-type", "inflammatory", and "bactericidal" sPLA2. Whereas the different designations indicate comprehensive functions or sources proposed for this enzyme, the identification of the precise roles of sPLA2-IIA has remained a challenge. This can be attributed to: the expression of the enzyme by various cells of different lineages, its limited activity towards the membranes of immune cells despite its expression following common inflammatory stimuli, its ability to interact with certain proteins independently of its catalytic activity, and its absence from multiple commonly used mouse models. Nevertheless, elevated levels of the enzyme during inflammatory processes and associated consistent release of arachidonic acid from the membrane of extracellular vesicles suggest that sPLA2-IIA may contribute to inflammation by using endogenous substrates in the extracellular milieu. Moreover, the remarkable potency of sPLA2-IIA towards bacterial membranes and its induced expression during the course of infections point to a role for this enzyme in the defense of the host against invading pathogens. In this review, we present current knowledge related to mammalian sPLA2-IIA and its roles in sterile inflammation and host defense.
Collapse
Affiliation(s)
- Etienne Dore
- Centre de Recherche du CHU de Québec, Université Laval, Department of Infectious Diseases and Immunity, Québec City, QC, Canada
| | - Eric Boilard
- Centre de Recherche du CHU de Québec, Université Laval, Department of Infectious Diseases and Immunity, Québec City, QC, Canada; Canadian National Transplantation Research Program, Edmonton, AB, Canada.
| |
Collapse
|
6
|
Kuefner MS, Pham K, Redd JR, Stephenson EJ, Harvey I, Deng X, Bridges D, Boilard E, Elam MB, Park EA. Secretory phospholipase A 2 group IIA modulates insulin sensitivity and metabolism. J Lipid Res 2017; 58:1822-1833. [PMID: 28663239 DOI: 10.1194/jlr.m076141] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/08/2017] [Indexed: 12/22/2022] Open
Abstract
Secretory phospholipase A2 group IIA (PLA2G2A) is a member of a family of secretory phospholipases that have been implicated in inflammation, atherogenesis, and antibacterial actions. Here, we evaluated the role of PLA2G2A in the metabolic response to a high fat diet. C57BL/6 (BL/6) mice do not express PLA2g2a due to a frameshift mutation. We fed BL/6 mice expressing the human PLA2G2A gene (IIA+ mice) a fat diet and assessed the physiologic response. After 10 weeks on the high fat diet, the BL/6 mice were obese, but the IIA+ mice did not gain weight or accumulate lipid. The lean mass in chow- and high fat-fed IIA+ mice was constant and similar to the BL/6 mice on a chow diet. Surprisingly, the IIA+ mice had an elevated metabolic rate, which was not due to differences in physical activity. The IIA+ mice were more insulin sensitive and glucose tolerant than the BL/6 mice, even when the IIA+ mice were provided the high fat diet. The IIA+ mice had increased expression of uncoupling protein 1 (UCP1), sirtuin 1 (SIRT1), and PPARγ coactivator 1α (PGC-1α) in brown adipose tissue (BAT), suggesting that PLA2G2A activates mitochondrial uncoupling in BAT. Our data indicate that PLA2G2A has a previously undiscovered impact on insulin sensitivity and metabolism.
Collapse
Affiliation(s)
- Michael S Kuefner
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Veterans Affairs Medical Center, Memphis, TN
| | - Kevin Pham
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Veterans Affairs Medical Center, Memphis, TN
| | - Jeanna R Redd
- Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI
| | - Erin J Stephenson
- Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Innocence Harvey
- Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI
| | - Xiong Deng
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Veterans Affairs Medical Center, Memphis, TN
| | - Dave Bridges
- Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI
| | - Eric Boilard
- Department of Infectious Diseases and Immunity, Faculté de Médecine de l'Université Laval, CHUQ Research Center and Division of Rheumatology, Quebec City, Canada
| | - Marshall B Elam
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Veterans Affairs Medical Center, Memphis, TN
| | - Edwards A Park
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN .,Department of Veterans Affairs Medical Center, Memphis, TN
| |
Collapse
|
7
|
Lu S, Dong Z. Overexpression of secretory phospholipase A2-IIa supports cancer stem cell phenotype via HER/ERBB-elicited signaling in lung and prostate cancer cells. Int J Oncol 2017; 50:2113-2122. [PMID: 28440478 DOI: 10.3892/ijo.2017.3964] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/10/2017] [Indexed: 11/06/2022] Open
Abstract
Resistance to conventional chemotherapies remains a significant clinical challenge in treatment of cancer. The cancer stem cells (CSCs) have properties necessary for tumor initiation, resistance to therapy, and progression. HER/ERBB‑elicited signaling supports CSC properties. Our previous studies revealed that secretory phospholipase A2 group IIa (sPLA2‑IIa) is overexpressed in both prostate and lung cancer cells, leading to an aberrant high level in the interstitial fluid, i.e., tumor microenvironment and blood. HER/ERBB-PI3K-Akt-NF-κB signaling stimulates sPLA2‑IIa overexpression, and in turn, sPLA2‑IIa activates EGFR family receptors and HER/ERBB-elicited signaling and stimulates sPLA2‑IIa overexpression in a positive feedback manner. The present study determined the molecular mechanisms of sPLA2‑IIa in stimulating HER/ERBB-elicited signaling and supporting CSC properties. We found that sPLA2‑IIa binds both EGFR and HER3 demonstrated by co-immunoprecipitation experiments and also indirectly interacts with HER2, suggesting that sPLA2‑IIa functions as a ligand for both EGFR and HER3. Furthermore, both side population CSCs from non-small cell lung cancer (NSCLC) A549 and H1975 cells and ALDH1‑high CSCs from castration-resistant prostate cancer (CRPC) 22Rv1 cells overexpress sPLA2‑IIa and produce tumors when inoculated into subcutis of nude mice. Given an aberrant high level of sPLA2‑IIa in the tumor microenvironment that should be much higher than that in the blood, our findings support the notion that sPLA2‑IIa functions as a ligand for EGFR family receptors and supports CSC properties via HER/ERBB-elicited signaling, which may contribute to resistance to therapy and cancer progression.
Collapse
Affiliation(s)
- Shan Lu
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Zhongyun Dong
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
8
|
Xie Q, Zhang D. Effects of Statins and Xuezhikang on the Expression of Secretory Phospholipase A2, Group IIA in Rat Vascular Smooth Muscle Cells. Int Heart J 2017; 58:115-124. [PMID: 28123160 DOI: 10.1536/ihj.16-163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Atherosclerosis is a multifactorial vascular disease characterized by formation of inflammatory lesions. Secretory phospholipase A2, group IIA (sPLA2-IIA) is involved in this process and plays a critical role. However, the exact role of sPLA2-IIA in cardiovascular inflammation is more complicated and remains unclear. Furthermore, both statins and Xuezhikang (XZK) are widely used in the prevention and treatment of cardiovascular disease risk because of their pleiotropic effects on the cardiovascular system. However, their effects on sPLA2-IIA are still controversial. We investigated the regulation of sPLA2-IIA by rat thoracic aorta smooth muscle cells (VSMCs) in culture. Cells were first incubated with IL-1β alone to induce expression of sPLA2-IIA and then treated with several concentrations of statins or XZK for different times in the absence or presence of IL-1β. We tested the expression of sPLA2-IIA, including sPLA2-IIA mRNA, protein, as well as activity. We found that statins or IL-1β increase the expression of sPLA2-IIA in VSMCs and the effect is based on a synergetic relationship between them. However, for the first time, we observed that XZK effectively reduces sPLA2-IIA expression in IL-1β-treated VSMCs. Our findings may shine a new light on the clinical use of XZK and statins in the prevention and treatment of atherosclerosis-related thrombosis.
Collapse
Affiliation(s)
- Qiang Xie
- Department of Cardiology, The First Hospital of Xiamen University
| | | |
Collapse
|
9
|
Zhang Y, Zhang Y. Pterostilbene, a novel natural plant conduct, inhibits high fat-induced atherosclerosis inflammation via NF-κB signaling pathway in Toll-like receptor 5 (TLR5) deficient mice. Biomed Pharmacother 2016; 81:345-355. [PMID: 27261612 DOI: 10.1016/j.biopha.2016.04.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/12/2016] [Accepted: 04/12/2016] [Indexed: 01/13/2023] Open
Abstract
Atherosclerosis is a specific form of an artery wall thickens, a syndrome affecting arterial blood vessels due to a chronic inflammatory response in the walls of arteries, which is promoted by fat accumulation. Toll-like receptors (TLRs) play prominent roles in inflammatory responses. And TLR5 is overexpressed in several diseases. Here in our study, we investigated the effect of TLR5 in high fat-induced atherosclerosis via NF-κB signaling pathway modulating pro-inflammatory cytokines releasing. Our results found that high fat induced atherosclerosis in wild type mice with fat accumulation and inflammatory response through NF-κB activation. Contrastly, TLR5 knockout mice displayed lower fat accumulation and ameliorated inflammation after high fat feeding with NF-κB inactivation. In addition, pterostilbene, as a natural dimethyl ether derivative of resveratrol mainly from blueberries, has diverse pharmacological activities, especially anti-inflammation. Our study also found that pterostilbene displayed inhibited role in suppressing inflammatory response through inactivating NF-κB signaling pathway regulated by TLR5 down-regulation in high fat-induced mice. Moreover, in vitro experiments of vascular smooth muscle cells (VSMCs) challenged with LPS or TNF-α, further indicated that NF-κB was involved in atherosclerosis progression, leading to high secretion of pro-inflammatory cytokines. However, VSMCs from TLR5 deficient mice inhibited phosphorylated levels of NF-κB signalilng pathway, finally resulting in down-regulation of inflammatory cytokines. Notably, pterostilbene also displayed suppressed role in inflammatory response via NF-κB inactivity in LPS or TNF-α-induced VSMCs by decreasing TLR5 expression. The results above indicated a novel therapeutic strategy of pterostilbene to protect against atherosclerosis via TLR5 regulation for clinic treatment in the future.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Cardiology, Huaihe Hospital, Henan University, 8 Baobei Rd., Kaifeng, 475000, China.
| | - Yi Zhang
- Department of Cardiology, The Fifth people's Hospital of Shenzhen City, 47 Youyi Rd., Shenzhen, 518001, China
| |
Collapse
|
10
|
Takada Y, Fujita M. Secreted Phospholipase A2 Type IIA (sPLA2-IIA) Activates Integrins in an Allosteric Manner. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 925:103-115. [PMID: 27864802 DOI: 10.1007/5584_2016_95] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Secreted phospholipase A2 type IIA (sPLA2-IIA) is a well-established pro-inflammatory protein and has been a major target for drug discovery. However, the mechanism of its signaling action has not been fully understood. We previously found that sPLA2-IIA binds to integrins αvβ3 and α4β1 in human and that this interaction plays a role in sPLA2-IIA's signaling action. Our recent studies found that sPLA2-IIA activates integrins in an allosteric manner through direct binding to a newly identified binding site of integrins (site 2), which is distinct from the classical RGD-binding site (site 1). The sPLA2-IIA-induced integrin activation may be related to the signaling action of sPLA2-IIA. Since sPLA2-IIA is present in normal human tears in addition to rheumatoid synovial fluid at high concentrations the sPLA2-IIA-mediated integrin activation on leukocytes may be involved in immune responses in normal and pathological conditions.
Collapse
Affiliation(s)
- Yoshikazu Takada
- Department of Dermatology, Biochemistry and Molecular Medicine, UC Davis School of Medicine, Research III Suite 3300, 4645 Second Avenue, Sacramento, CA, 95817, USA. .,The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan, Republic of China.
| | - Masaaki Fujita
- Department of Clinical Immunology and Rheumatology, The Tazuke-Kofukai Medical Research Institute, Kitano Hospital, 2-4-20 Ohgimachi, Kita-ku, Osaka, 530-8480, Japan
| |
Collapse
|
11
|
El Hadri K, Denoyelle C, Ravaux L, Viollet B, Foretz M, Friguet B, Rouis M, Raymondjean M. AMPK Signaling Involvement for the Repression of the IL-1β-Induced Group IIA Secretory Phospholipase A2 Expression in VSMCs. PLoS One 2015; 10:e0132498. [PMID: 26162096 PMCID: PMC4498592 DOI: 10.1371/journal.pone.0132498] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 06/15/2015] [Indexed: 12/25/2022] Open
Abstract
Secretory Phospholipase A2 of type IIA (sPLA2 IIA) plays a crucial role in the production of lipid mediators by amplifying the neointimal inflammatory context of the vascular smooth muscle cells (VSMCs), especially during atherogenesis. Phenformin, a biguanide family member, by its anti-inflammatory properties presents potential for promoting beneficial effects upon vascular cells, however its impact upon the IL-1β-induced sPLA2 gene expression has not been deeply investigated so far. The present study was designed to determine the relationship between phenformin coupling AMP-activated protein kinase (AMPK) function and the molecular mechanism by which the sPLA2 IIA expression was modulated in VSMCs. Here we find that 5-aminoimidazole-4-carboxamide-1-β-D-ribonucleotide (AICAR) treatment strongly repressed IL-1β-induced sPLA2 expression at least at the transcriptional level. Our study reveals that phenformin elicited a dose-dependent inhibition of the sPLA2 IIA expression and transient overexpression experiments of constitutively active AMPK demonstrate clearly that AMPK signaling is involved in the transcriptional inhibition of sPLA2-IIA gene expression. Furthermore, although the expression of the transcriptional repressor B-cell lymphoma-6 protein (BCL-6) was markedly enhanced by phenformin and AICAR, the repression of sPLA2 gene occurs through a mechanism independent of BCL-6 DNA binding site. In addition we show that activation of AMPK limits IL-1β-induced NF-κB pathway activation. Our results indicate that BCL-6, once activated by AMPK, functions as a competitor of the IL-1β induced NF-κB transcription complex. Our findings provide insights on a new anti-inflammatory pathway linking phenformin, AMPK and molecular control of sPLA2 IIA gene expression in VSMCs.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Aminoimidazole Carboxamide/analogs & derivatives
- Aminoimidazole Carboxamide/pharmacology
- Animals
- Binding Sites
- Cattle
- Cell Separation
- Cells, Cultured
- Enzyme Activation/drug effects
- Gene Expression Regulation, Enzymologic/drug effects
- Group II Phospholipases A2/genetics
- Group II Phospholipases A2/metabolism
- Interleukin-1beta/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- NF-kappa B/metabolism
- Phenformin/pharmacology
- Phosphorylation/drug effects
- Promoter Regions, Genetic/genetics
- Protein Subunits/metabolism
- Proto-Oncogene Proteins c-bcl-6/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Wistar
- Ribonucleotides/pharmacology
- Signal Transduction/drug effects
- Transcriptional Activation/drug effects
Collapse
Affiliation(s)
- Khadija El Hadri
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
- * E-mail:
| | - Chantal Denoyelle
- Université Paris Diderot, Sorbonne Paris Cité, BFA CNRS UMR8251, 4 Rue MA Lagroua Weill Hallé, 75013 Paris, France
| | - Lucas Ravaux
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
| | - Benoit Viollet
- Institut Cochin, Inserm U1016, Paris, France
- CNRS, UMR 8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marc Foretz
- Institut Cochin, Inserm U1016, Paris, France
- CNRS, UMR 8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bertrand Friguet
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
| | - Mustapha Rouis
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
| | - Michel Raymondjean
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
| |
Collapse
|
12
|
Pernet E, Brunet J, Guillemot L, Chignard M, Touqui L, Wu Y. Staphylococcus aureusAdenosine Inhibits sPLA2-IIA–Mediated Host Killing in the Airways. THE JOURNAL OF IMMUNOLOGY 2015; 194:5312-9. [DOI: 10.4049/jimmunol.1402665] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/17/2015] [Indexed: 12/19/2022]
|
13
|
DONG ZHONGYUN, MELLER JAROSLAW, SUCCOP PAUL, WANG JIANG, WIKENHEISER-BROKAMP KATHRYN, STARNES SANDRA, LU SHAN. Secretory phospholipase A2-IIa upregulates HER/HER2-elicited signaling in lung cancer cells. Int J Oncol 2014; 45:978-84. [PMID: 24913497 PMCID: PMC4121404 DOI: 10.3892/ijo.2014.2486] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/03/2014] [Indexed: 11/06/2022] Open
Abstract
Lung cancer is the leading cause of cancer death worldwide. There is an urgent need for early diagnostic tools and novel therapies in order to increase lung cancer survival. Secretory phospholipase A2 group IIa (sPLA2-IIa) is involved in inflammation, tumorigenesis and metastasis. We were the first to uncover that cancer cells secrete sPLA2‑IIa. sPLA2‑IIa is overexpressed in almost all specimens of human lung cancers examined and is significantly elevated in the plasma of lung cancer patients. High levels of plasma sPLA2-IIa are significantly associated with advanced stage and decreased overall cancer survival. In this study, we further showed that elevated HER/HER2‑PI3K-Akt-NF-κB signaling contributes to sPLA2-IIa overexpression in lung cancer cells. sPLA2-IIa in turn phosphorylates and activates HER2 and HER3 in a time- and dose‑dependent manner in lung cancer cells. The structure and sequence‑based docking analysis revealed that sPLA2-IIa β hairpin shares structural similarity with the corresponding EGF hairpin. sPLA2-IIa forms an extensive interface with EGFR and brings the two lobes of EGFR into an active conformation. sPLA2-IIa also enhances the NF-κB promoter activity. Anti-sPLA2-IIa antibody, but not the small molecule sPLA2-IIa inhibitor LY315920, significantly inhibits sPLA2‑IIa-induced activation of NF-κB promoter. Our findings support the notion that sPLA2-IIa functions as a ligand for the EGFR family of receptors leading to an elevated HER/HER2-elicited signaling. Plasma sPLA2-IIa can potentially serve as lung cancer biomarker and sPLA2‑IIa is a potential therapeutic target against lung cancer.
Collapse
Affiliation(s)
- ZHONGYUN DONG
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - JAROSLAW MELLER
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - PAUL SUCCOP
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - JIANG WANG
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | | | - SANDRA STARNES
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - SHAN LU
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| |
Collapse
|
14
|
Sharma P, Thakran S, Deng X, Elam MB, Park EA. Nuclear corepressors mediate the repression of phospholipase A2 group IIa gene transcription by thyroid hormone. J Biol Chem 2013; 288:16321-16333. [PMID: 23629656 DOI: 10.1074/jbc.m112.445569] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Secretory phospholipase A2 group IIa (PLA2g2a) is associated with inflammation, hyperlipidemia, and atherogenesis. Transcription of the PLA2g2a gene is induced by multiple cytokines. Here, we report the surprising observation that thyroid hormone (T3) inhibited PLA2g2a gene expression in human and rat hepatocytes as well as in rat liver. Moreover, T3 reduced the cytokine-mediated induction of PLA2g2a, suggesting that the thyroid status may modulate aspects of the inflammatory response. In an effort to dissect the mechanism of repression by T3, we cloned the PLA2g2a gene and identified a negative T3 response element in the promoter. This T3 receptor (TRβ)-binding site differed considerably from consensus T3 stimulatory elements. Using in vitro and in vivo binding assays, we found that TRβ bound directly to the PLA2g2a promoter as a heterodimer with the retinoid X receptor. Knockdown of nuclear corepressor or silencing mediator for retinoid and thyroid receptors by siRNA blocked the T3 inhibition of PLA2g2a. Using chromatin immunoprecipitation assays, we showed that nuclear corepressor and silencing mediator for retinoid and thyroid receptors were associated with the PLA2g2a gene in the presence of T3. In contrast with the established role of T3 to promote coactivator association with TRβ, our experiments demonstrate a novel inverse recruitment mechanism in which liganded TRβ recruits corepressors to inhibit PLA2g2a expression.
Collapse
Affiliation(s)
- Pragya Sharma
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Shalini Thakran
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Xiong Deng
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163; Department of Veterans Affairs Medical Center, Memphis, Tennessee 38163
| | - Marshall B Elam
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163; Department of Veterans Affairs Medical Center, Memphis, Tennessee 38163
| | - Edwards A Park
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163.
| |
Collapse
|
15
|
Pessolano LG, Sullivan CP, Seidl SE, Rich CB, Liscum L, Stone PJ, Sipe JD, Schreiber BM. Trafficking of endogenous smooth muscle cell cholesterol: a role for serum amyloid A and interleukin-1β. Arterioscler Thromb Vasc Biol 2012; 32:2741-50. [PMID: 22995521 DOI: 10.1161/atvbaha.112.300243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Intracellular cholesterol distribution impacts cell function; however, processes influencing endogenous cholesterol trafficking remain largely unknown. Atherosclerosis is associated with vascular inflammation and these studies address the role of inflammatory mediators on smooth muscle cell cholesterol trafficking. METHODS AND RESULTS Interestingly, in the absence of an exogenous cholesterol source, serum amyloid A increased [(14)C] oleic acid incorporation into cholesteryl ester in rat smooth muscle cells, suggesting endogenous cholesterol trafficking to the endoplasmic reticulum. [(3)H] cholesteryl ester accumulated in cells prelabeled with [(3)H] cholesterol, confirming that serum amyloid A mediated the movement of endogenous cholesterol. Cholesterol movement was dependent upon functional endolysosomes. The cholesterol oxidase-sensitive pool of cholesterol decreased in serum amyloid A-treated cells. Furthermore, the mechanism whereby serum amyloid A induced cholesterol trafficking was determined to be via activation of expression of secretory phospholipase A(2), group IIA (sPLA(2)) and sPLA(2)-dependent activation of sphingomyelinase. Interestingly, although neither tumor necrosis factor-α nor interferon-γ induced cholesterol trafficking, interleukin-1β induced [(14)C] cholesteryl ester accumulation that was also dependent upon sPLA(2) and sphingomyelinase activities. Serum amyloid A activates smooth muscle cell interleukin-1β expression, and although the interleukin-1-receptor antagonist inhibited the interleukin-1β-induced cholesterol trafficking, it had no effect on the movement of cholesterol mediated by serum amyloid A. CONCLUSIONS These data support a role for inflammation in endogenous smooth muscle cell cholesterol trafficking from the plasma membrane to the endoplasmic reticulum.
Collapse
Affiliation(s)
- Lawrence G Pessolano
- Department of Biochemistry, Boston University School of Medicine, K207, 72 East Concord Street, Boston, MA 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
Alzheimer's disease (AD) and bipolar disorder (BD) are progressive brain disorders. Upregulated mRNA and protein levels of neuroinflammatory and arachidonic acid (AA) markers with loss of synaptic markers (synaptophysin and drebrin) have been reported in brain tissue from AD and BD patients. We hypothesized that some of these changes are associated with epigenetic modifications of relevant genes. To test this, we measured gene-specific CpG methylation, global DNA methylation and histone modifications in postmortem frontal cortex from BD (n=10) and AD (n=10) patients and respective age-matched controls (10 per group). AD and BD brains showed several epigenetic similarities, including global DNA hypermethylation, and histone H3 phosphorylation. These changes were associated with hypo- and hypermethylation of CpG islands in cyclooxygenase-2 and brain-derived neurotrophic factor promoter regions, respectively. Only the AD brain showed hyper- and hypomethylated CpG islands in promoter regions for cAMP response element-binding protein and nuclear transcription factor kappa B genes, respectively. Only the BD brain demonstrated increased global histone H3 acetylation and hypermethylation of the promotor region for the drebrin-like protein gene. There was no significant epigenetic modification for 12-lipooxygenase or p450 epoxygenase in either illness. Many observed epigenetic changes were inversely related to respective changes in mRNA and protein levels. These epigenetic modifications involving neuroinflammatory, AA cascade and synaptic markers may contribute to progression in AD and BD and identify new targets for drug development.
Collapse
|
17
|
Oleksowicz L, Liu Y, Bracken RB, Gaitonde K, Burke B, Succop P, Levin L, Dong Z, Lu S. Secretory phospholipase A2-IIa is a target gene of the HER/HER2-elicited pathway and a potential plasma biomarker for poor prognosis of prostate cancer. Prostate 2012; 72:1140-9. [PMID: 22127954 PMCID: PMC3345320 DOI: 10.1002/pros.22463] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 11/01/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Our previous study showed that prostate cancer cells overexpress and secrete secretory phospholipases A2 group IIa (sPLA2-IIa) and plasma sPLA2-IIa was elevated in prostate cancer patients. The current study further explored the underlying mechanism of sPLA2-IIa overexpression and the potential role of sPLA2-IIa as a prostate cancer biomarker. METHODS Plasma and tissue specimens from prostate cancer patients were analyzed for sPLA2-IIa levels. Regulation of sPLA2-IIa expression by Heregulin-α was determined by Western blot and reporter assay. RESULTS We found that Heregulin-α enhanced expression of the sPLA2-IIa gene via the HER2/HER3-elicited pathway. The EGFR/HER2 dual inhibitor Lapatinib and the NF-kB inhibitor Bortezomib inhibited sPLA2-IIa expression induced by Heregulin-α. Heregulin-α upregulated expression of the sPLA2-IIa gene at the transcriptional level. We further confirmed that plasma sPLA2-IIa secreted by mouse bearing human prostate cancer xenografts reached detectable plasma concentrations. A receiver operating characteristic (ROC) analysis of patient plasma specimens revealed that high levels of plasma sPLA2-IIa, with the optimum cutoff value of 2.0 ng/ml, were significantly associated with high Gleason score (8-10) relative to intermediate Gleason score (6-7) prostate cancers and advanced relative to indolent cancers. The area under the ROC curve (area under curve, AUC) was 0.73 and 0.74, respectively. CONCLUSION We found that Heregulin-α, in addition to EGF, contributes to sPLA2-IIa overexpression in prostate cancer cells. Our findings support the notion that high levels of plasma sPLA2-IIa may serve as a poor prognostic biomarker capable of distinguishing aggressive from indolent prostate cancers, which may improve decision-making and optimize patient management.
Collapse
Affiliation(s)
- Leslie Oleksowicz
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Yin Liu
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - R. Bruce Bracken
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Krishnanath Gaitonde
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Barbara Burke
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Paul Succop
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Linda Levin
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Zhongyun Dong
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Shan Lu
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH 45237
- To whom correspondence should be addressed. Department of Pathology, University of Cincinnati College of Medicine, Building A, Room 259, 2120 East, Galbraith Road, Cincinnati, OH 45237-0507. Phone: 513-558-5109; Fax: 513-558-1312;
| |
Collapse
|
18
|
Kellom M, Basselin M, Keleshian VL, Chen M, Rapoport SI, Rao JS. Dose-dependent changes in neuroinflammatory and arachidonic acid cascade markers with synaptic marker loss in rat lipopolysaccharide infusion model of neuroinflammation. BMC Neurosci 2012; 13:50. [PMID: 22621398 PMCID: PMC3464147 DOI: 10.1186/1471-2202-13-50] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 05/08/2012] [Indexed: 11/16/2022] Open
Abstract
Background Neuroinflammation, caused by six days of intracerebroventricular infusion of bacterial lipopolysaccharide (LPS), stimulates rat brain arachidonic acid (AA) metabolism. The molecular changes associated with increased AA metabolism are not clear. We examined effects of a six-day infusion of a low-dose (0.5 ng/h) and a high-dose (250 ng/h) of LPS on neuroinflammatory, AA cascade, and pre- and post-synaptic markers in rat brain. We used artificial cerebrospinal fluid-infused brains as controls. Results Infusion of low- or high-dose LPS increased brain protein levels of TNFα, and iNOS, without significantly changing GFAP. High-dose LPS infusion upregulated brain protein and mRNA levels of AA cascade markers (cytosolic cPLA2-IVA, secretory sPLA2-V, cyclooxygenase-2 and 5-lipoxygenase), and of transcription factor NF-κB p50 DNA binding activity. Both LPS doses increased cPLA2 and p38 mitogen-activated protein kinase levels, while reducing protein levels of the pre-synaptic marker, synaptophysin. Post-synaptic markers drebrin and PSD95 protein levels were decreased with high- but not low-dose LPS. Conclusions Chronic LPS infusion has differential effects, depending on dose, on inflammatory, AA and synaptic markers in rat brain. Neuroinflammation associated with upregulated brain AA metabolism can lead to synaptic dysfunction.
Collapse
Affiliation(s)
- Matthew Kellom
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, 9000 Rockville Pike, Bldg. 9, 1S-126, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
19
|
Plasma secretory phospholipase A2-IIa as a potential biomarker for lung cancer in patients with solitary pulmonary nodules. BMC Cancer 2011; 11:513. [PMID: 22151235 PMCID: PMC3250967 DOI: 10.1186/1471-2407-11-513] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 12/09/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Five-year survival for lung cancer has remained at 16% over last several decades largely due to the fact that over 50% of patients are diagnosed with locally-advanced or metastatic disease. Diagnosis at an earlier and potentially curable stage is crucial. Solitary pulmonary nodules (SPNs) are common, but the difficulty lies in the determination of which SPN is malignant. Currently, there is no convenient and reliable biomarker effective for early diagnosis. Secretory phospholipase A2-IIa (sPLA2-IIa) is secreted into the circulation by cancer cells and may allow for an early detection of lung cancer. METHODS Plasma samples from healthy donors, patients with only benign SPN, and patients with lung cancer were analyzed. Expression of sPLA2-IIa protein in lung cancer tissues was also determined. RESULTS We found that the levels of plasma sPLA2-IIa were significantly elevated in lung cancer patients. The receiver operating characteristic curve analysis, comparing lung cancer patients to patients with benign nodules, revealed an optimum cutoff value for plasma sPLA2-IIa of 2.4 ng/ml to predict an early stage cancer with 48% sensitivity and 86% specificity and up to 67% sensitivity for T2 stage lung cancer. Combined sPLA2-IIa, CEA, and Cyfra21.1 tests increased the sensitivity for lung cancer prediction. High level of plasma sPLA2-IIa was associated with a decreased overall cancer survival. sPLA2-IIa was overexpressed in almost all non-small cell lung cancer and in the majority of small cell lung cancer by immunohistochemistry analysis. CONCLUSION Our finding strongly suggests that plasma sPLA2-IIa is a potential lung biomarker to distinguish benign nodules from lung cancer and to aid lung cancer diagnosis in patients with SPNs.
Collapse
|
20
|
Rao JS, Rapoport SI, Kim HW. Altered neuroinflammatory, arachidonic acid cascade and synaptic markers in postmortem Alzheimer's disease brain. Transl Psychiatry 2011; 1:e31. [PMID: 22832605 PMCID: PMC3309508 DOI: 10.1038/tp.2011.27] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 06/07/2011] [Accepted: 06/16/2011] [Indexed: 12/29/2022] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is the leading cause of dementia in the elderly. A recent positron emission tomography imaging study demonstrated upregulated brain arachidonic acid (AA) metabolism in AD patients. Further, a mouse model of AD shows an increase in AA-releasing cytosolic phospholipase A(2) (cPLA(2)) in brain, and a reduction in cPLA(2) activity ameliorated cognitive deficits. These observations led us to hypothesize that there is an upregulation of AA cascade and neuroinflammatory markers in the brain of AD patients. To test this hypothesis, we measured protein and mRNA levels of AA cascade, neuroinflammatory and synaptic markers in postmortem frontal cortex from 10 AD patients and 10 age-matched controls. Consistent with our hypothesis, AD frontal cortex showed significant increases in protein and mRNA levels of cPLA(2)-IVA, secretory sPLA(2)-IIA, cyclooxygenase-1 and -2, membrane prostaglandin (PG) synthase-1 and lipoxygenase-12 and -15. Calcium-independent iPLA(2)-VIA and cytosolic PGE(2) synthase were decreased. In addition, interleukin-1β, tumor necrosis factor-α, glial fibrillary acidic protein and CD11b were increased. AD postmortem brain also showed signs of cellular injury, including decreased synaptophysin and drebrin, pre- and postsynaptic markers. These results indicate that increased AA cascade and inflammatory markers could contribute to AD pathology. Altered brain AA cascade enzymes could be considered therapeutic targets for future drug development.
Collapse
Affiliation(s)
- J S Rao
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
21
|
Rao JS, Kim HW, Kellom M, Greenstein D, Chen M, Kraft AD, Harry GJ, Rapoport SI, Basselin M. Increased neuroinflammatory and arachidonic acid cascade markers, and reduced synaptic proteins, in brain of HIV-1 transgenic rats. J Neuroinflammation 2011; 8:101. [PMID: 21846384 PMCID: PMC3175175 DOI: 10.1186/1742-2094-8-101] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 08/16/2011] [Indexed: 12/23/2022] Open
Abstract
Background Cognitive impairment has been reported in human immune deficiency virus-1- (HIV-1-) infected patients as well as in HIV-1 transgenic (Tg) rats. This impairment has been linked to neuroinflammation, disturbed brain arachidonic acid (AA) metabolism, and synapto-dendritic injury. We recently reported upregulated brain AA metabolism in 7- to 9-month-old HIV-1 Tg rats. We hypothesized that these HIV-1 Tg rats also would show upregulated brain inflammatory and AA cascade markers and a deficit of synaptic proteins. Methods We measured protein and mRNA levels of markers of neuroinflammation and the AA cascade, as well as pro-apoptotic factors and synaptic proteins, in brains from 7- to 9-month-old HIV-1 Tg and control rats. Results Compared with control brain, HIV-1 Tg rat brain showed immunoreactivity to glycoprotein 120 and tat HIV-1 viral proteins, and significantly higher protein and mRNA levels of (1) the inflammatory cytokines interleukin-1β and tumor necrosis factor α, (2) the activated microglial/macrophage marker CD11b, (3) AA cascade enzymes: AA-selective Ca2+-dependent cytosolic phospholipase A2 (cPLA2)-IVA, secretory sPLA2-IIA, cyclooxygenase (COX)-2, membrane prostaglandin E2 synthase, 5-lipoxygenase (LOX) and 15-LOX, cytochrome p450 epoxygenase, and (4) transcription factor NF-κBp50 DNA binding activity. HIV-1 Tg rat brain also exhibited signs of cell injury, including significantly decreased levels of brain-derived neurotrophic factor (BDNF) and drebrin, a marker of post-synaptic excitatory dendritic spines. Expression of Ca2+-independent iPLA2-VIA and COX-1 was unchanged. Conclusions HIV-1 Tg rats show elevated brain markers of neuroinflammation and AA metabolism, with a deficit in several synaptic proteins. These changes are associated with viral proteins and may contribute to cognitive impairment. The HIV-1 Tg rat may be a useful model for understanding progression and treatment of cognitive impairment in HIV-1 patients.
Collapse
Affiliation(s)
- Jagadeesh Sridhara Rao
- Brain Physiology and Metabolism Section, National Institute on Aging, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Basselin M, Ramadan E, Chen M, Rapoport SI. Anti-inflammatory effects of chronic aspirin on brain arachidonic acid metabolites. Neurochem Res 2011; 36:139-45. [PMID: 20981485 PMCID: PMC3011042 DOI: 10.1007/s11064-010-0282-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2010] [Indexed: 12/22/2022]
Abstract
Pro-inflammatory and anti-inflammatory mediators derived from arachidonic acid (AA) modulate peripheral inflammation and its resolution. Aspirin (ASA) is a unique non-steroidal anti-inflammatory drug, which switches AA metabolism from prostaglandin E₂ (PGE₂) and thromboxane B₂ (TXB₂) to lipoxin A₄ (LXA₄) and 15-epi-LXA₄. However, it is unknown whether chronic therapeutic doses of ASA are anti-inflammatory in the brain. We hypothesized that ASA would dampen increases in brain concentrations of AA metabolites in a rat model of neuroinflammation, produced by a 6-day intracerebroventricular infusion of bacterial lipopolysaccharide (LPS). In rats infused with LPS (0.5 ng/h) and given ASA-free water to drink, concentrations in high-energy microwaved brain of PGE₂, TXB₂ and leukotriene B₄ (LTB₄) were elevated. In rats infused with artificial cerebrospinal fluid, 6 weeks of treatment with a low (10 mg/kg/day) or high (100 mg/kg/day) ASA dose in drinking water decreased brain PGE₂, but increased LTB₄, LXA₄ and 15-epi-LXA₄ concentrations. Both doses attenuated the LPS effects on PGE₂, and TXB₂. The increments in LXA₄ and 15-epi-LXA₄ caused by high-dose ASA were significantly greater in LPS-infused rats. The ability of ASA to increase anti-inflammatory LXA₄ and 15-epi-LXA₄ and reduce pro-inflammatory PGE₂ and TXB₂ suggests considering aspirin further for treating clinical neuroinflammation.
Collapse
Affiliation(s)
- Mireille Basselin
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Epolia Ramadan
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mei Chen
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stanley I. Rapoport
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
Varespladib (A-002), a secretory phospholipase A2 inhibitor, reduces atherosclerosis and aneurysm formation in ApoE-/- mice. J Cardiovasc Pharmacol 2010; 53:60-5. [PMID: 19129734 DOI: 10.1097/fjc.0b013e318195bfbc] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The family of secretory phospholipase A2 (sPLA2) enzymes has been associated with inflammatory diseases and tissue injury including atherosclerosis. A-001 is a novel inhibitor of sPLA2 enzymes discovered by structure-based drug design, and A-002 is the orally bioavailable prodrug currently in clinical development. A-001 inhibited human and mouse sPLA2 group IIA, V, and X enzymes with IC50 values in the low nM range. A-002 (1 mg/kg) led to high serum levels of A-001 and inhibited PLA2 activity in transgenic mice overexpressing human sPLA2 group IIA in C57BL/6J background. In addition, the effects of A-002 on atherosclerosis in 2 ApoE mouse models were evaluated using en face analysis. (1) In a high-fat diet model, A-002 (30 and 90 mg/kg twice a day for 16 weeks) reduced aortic atherosclerosis by 50% (P < 0.05). Plasma total cholesterol was decreased (P < 0.05) by 1 month and remained lowered throughout the study. (2) In an accelerated atherosclerosis model, with angiotensin II-induced aortic lesions and aneurysms, A-002 (30 mg/kg twice a day) reduced aortic atherosclerosis by approximately 40% (P < 0.05) and attenuated aneurysm formation (P = 0.0096). Thus, A-002 was effective at significantly decreasing total cholesterol, atherogenesis, and aneurysm formation in these 2 ApoE mouse models.
Collapse
|
24
|
Dong Z, Liu Y, Scott KF, Levin L, Gaitonde K, Bracken RB, Burke B, Zhai QJ, Wang J, Oleksowicz L, Lu S. Secretory phospholipase A2-IIa is involved in prostate cancer progression and may potentially serve as a biomarker for prostate cancer. Carcinogenesis 2010; 31:1948-55. [PMID: 20837598 DOI: 10.1093/carcin/bgq188] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The majority of prostate cancers are indolent, whereas a significant portion of patients will require systemic treatment during the course of their disease. To date, only high Gleason scores are best associated with a poor prognosis in prostate cancer. No validated serum biomarker has been identified with prognostic power. Previous studies showed that secretory phospholipase A2-IIa (sPLA2-IIa) is overexpressed in almost all human prostate cancer specimens and its elevated levels are correlated with high tumor grade. Here, we found that sPLA2-IIa is overexpressed in androgen-independent prostate cancer LNCaP-AI cells relative to their androgen-dependent LNCaP cell counterparts. LNCaP-AI cells also secrete significantly higher levels of sPLA2-IIa. Blocking sPLA2-IIa function compromises androgen-independent cell growth. Inhibition of the ligand-induced signaling output of the HER network, by blocking PI3K-Akt signaling and the nuclear factor-kappaB (NF-κB)-mediated pathway, compromises both sPLA2-IIa protein expression and secretion, as a result of downregulation of sPLA2-IIa promoter activity. More importantly, we demonstrated elevated serum sPLA2-IIa levels in prostate cancer patients. High serum sPLA2-IIa levels are associated significantly with high Gleason score and advanced disease stage. Increased sPLA2-IIa expression was confirmed in prostate cancer cells, but not in normal epithelium and stroma by immunohistochemistry analysis. We showed that elevated signaling of the HER/HER2-PI3K-Akt-NF-κB pathway contributes to sPLA2-IIa overexpression and secretion by prostate cancer cells. Given that sPLA2-IIa overexpression is associated with prostate development and progression, serum sPLA2-IIa may serve as a prognostic biomarker for prostate cancer and a potential surrogate prostate biomarker indicative of tumor burden.
Collapse
Affiliation(s)
- Zhongyun Dong
- Department of Medicine, University of Cincinnati College of Medicine, 2120 East Galbraith Road, Cincinnati, OH 45237-0507, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Raymond B, Ravaux L, Mémet S, Wu Y, Sturny-Leclère A, Leduc D, Denoyelle C, Goossens PL, Payá M, Raymondjean M, Touqui L. Anthrax lethal toxin down-regulates type-IIA secreted phospholipase A(2) expression through MAPK/NF-kappaB inactivation. Biochem Pharmacol 2009; 79:1149-55. [PMID: 19962969 DOI: 10.1016/j.bcp.2009.11.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 11/27/2009] [Accepted: 11/30/2009] [Indexed: 10/20/2022]
Abstract
Bacillus anthracis, the etiological agent of anthrax, produces lethal toxin (LT) that displays a metallo-proteolytic activity toward the N-terminus of the MAPK-kinases. We have previously shown that secreted type-IIA phospholipase A(2) (sPLA(2)-IIA) exhibits potent anthracidal activity. In vitro expression of sPLA(2)-IIA in guinea pig alveolar macrophages (AMs), the major source of this enzyme in lung tissues, is inhibited by LT. Here, we examined the mechanisms involved in sPLA(2)-IIA inhibition by LT. We first showed that chemical inhibitors of p38 and ERK MAPKs reduced sPLA(2)-IIA expression in AMs indicating that these kinases play a role in sPLA(2)-IIA expression. LT inhibited IL-1beta-induced p38 phosphorylation as well as sPLA(2)-IIA promoter activity in CHO cells. Inhibition of sPLA(2)-IIA promoter activity was mimicked by co-transfection with dominant negative construct of p38 (DN-p38) and reversed by the active form of p38-MAPK (AC-p38). Both LT and DN-p38 decreased IL-1beta-induced NF-kappaB luciferase activity. This contrasted with the effect of AC-p38, which enhanced this activity. However, neither LT nor specific p-38 inhibitor interfered with LPS-induced IkappaBalpha degradation or NF-kappaB nuclear translocation in AMs. Subcutaneous administration of LT to guinea pig before LPS challenge reduced sPLA(2)-IIA levels in broncho-alveolar lavages and ears. We conclude that sPLA(2)-IIA expression is induced via a sequential MAPK-NF-kappaB activation and that LT inhibits this expression likely by interfering with the transactivation of NF-kappaB in the nucleus. This inhibition, which is operating both in vitro and in vivo, may represent a mechanism by which B. anthracis subvert host defense.
Collapse
|
26
|
Sullivan CP, Seidl SE, Rich CB, Raymondjean M, Schreiber BM. Secretory phospholipase A2, group IIA is a novel serum amyloid A target gene: activation of smooth muscle cell expression by an interleukin-1 receptor-independent mechanism. J Biol Chem 2009; 285:565-75. [PMID: 19850938 DOI: 10.1074/jbc.m109.070565] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a multifactorial vascular disease characterized by formation of inflammatory lesions. Elevated circulating acute phase proteins indicate disease risk. Serum amyloid A (SAA) is one such marker but its function remains unclear. To determine the role of SAA on aortic smooth muscle cell gene expression, a preliminary screen of a number of genes was performed and a strong up-regulation of expression of secretory phospholipase A(2), group IIA (sPLA(2)) was identified. The SAA-induced increase in sPLA(2) was validated by real time PCR, Western blot analysis, and enzyme activity assays. Demonstrating that SAA increased expression of sPLA(2) heteronuclear RNA and that inhibiting transcription eliminated the effect of SAA on sPLA(2) mRNA suggested that the increase was transcriptional. Transient transfections and electrophoretic mobility shift assays identified CAAT enhancer-binding protein (C/EBP) and nuclear factor kappaB (NFkappaB) as key regulatory sites mediating the induction of sPLA(2). Moreover, SAA activated the inhibitor of NF-kappaB kinase (IKK) in cultured smooth muscle cells. Previous reports showed that interleukin (IL)-1beta up-regulates Pla2g2a gene transcription via C/EBPbeta and NFkappaB. Interestingly, SAA activated smooth muscle cell IL-1beta mRNA expression, however, blocking IL-1 receptors had no effect on SAA-mediated activation of sPLA(2) expression. Thus, the observed changes in sPLA(2) expression were not secondary to SAA-induced IL-1 receptor activation. The association of SAA with high density lipoprotein abrogated the SAA-induced increase in sPLA(2) expression. These data suggest that during atherogenesis, SAA can amplify the involvement of smooth muscle cells in vascular inflammation and that this can lead to deposition of sPLA(2) and subsequent local changes in lipid homeostasis.
Collapse
Affiliation(s)
- Christopher P Sullivan
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | |
Collapse
|
27
|
Divchev D, Schieffer B. The secretory phospholipase A2 group IIA: a missing link between inflammation, activated renin-angiotensin system, and atherogenesis? Vasc Health Risk Manag 2008; 4:597-604. [PMID: 18827909 PMCID: PMC2515419 DOI: 10.2147/vhrm.s2008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Inflammation, lipid peroxidation and chronic activation of the rennin – angiotensin system (RAS) are hallmarks of the development of atherosclerosis. Recent studies have suggested the involvement of the pro-inflammatory secretory phospholipase A2 (sPLA2)-IIA in atherogenesis. This enzyme is produced by different cell types through stimulation by pro-inflammatory cytokines. It is detectable in the intima and in media smooth muscle cells, not only in atherosclerotic lesions but also in the very early stages of atherogenesis. sPLA2-IIA can hydrolyse the phospholipid monolayers of low density lipoproteins (LDL). Such modified LDL show increased affinity to proteoglycans. The modified particles have a greater tendency to aggregate and an enhanced ability to insert cholesterol into cells. This modification may promote macrophage LDL uptake leading to the formation of foam cells. Furthermore, sPLA2-IIA is not only a mediator for localized inflammation but may be also used as an independent predictor of adverse outcomes in patients with stable coronary artery disease or acute coronary syndromes. An interaction between activated RAS and phospholipases has been indicated by observations showing that inhibitors of sPLA2 decrease angiotensin (Ang) II-induced macrophage lipid peroxidation. Meanwhile, various interactions between Ang II and oxLDL have been demonstrated suggesting a central role of sPLA2-IIA in these processes and offering a possible target for treatment. The role of sPLA2-IIA in the perpetuation of atherosclerosis appears to be the missing link between inflammation, activated RAS and lipidperoxidation.
Collapse
Affiliation(s)
- Dimitar Divchev
- Department of Cardiology and Angiology, Medizinische Hochschule Hannover, Germany
| | | |
Collapse
|
28
|
Raymond B, Leduc D, Ravaux L, Le Goffic R, Candela T, Raymondjean M, Goossens PL, Touqui L. Edema toxin impairs anthracidal phospholipase A2 expression by alveolar macrophages. PLoS Pathog 2008; 3:e187. [PMID: 18069891 PMCID: PMC2134952 DOI: 10.1371/journal.ppat.0030187] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Accepted: 10/26/2007] [Indexed: 01/14/2023] Open
Abstract
Bacillus anthracis, the etiological agent of anthrax, is a spore-forming Gram-positive bacterium. Infection with this pathogen results in multisystem dysfunction and death. The pathogenicity of B. anthracis is due to the production of virulence factors, including edema toxin (ET). Recently, we established the protective role of type-IIA secreted phospholipase A2 (sPLA2-IIA) against B. anthracis. A component of innate immunity produced by alveolar macrophages (AMs), sPLA2-IIA is found in human and animal bronchoalveolar lavages at sufficient levels to kill B. anthracis. However, pulmonary anthrax is almost always fatal, suggesting the potential impairment of sPLA2-IIA synthesis and/or action by B. anthracis factors. We investigated the effect of purified ET and ET-deficient B. anthracis strains on sPLA2-IIA expression in primary guinea pig AMs. We report that ET inhibits sPLA2-IIA expression in AMs at the transcriptional level via a cAMP/protein kinase A–dependent process. Moreover, we show that live B. anthracis strains expressing functional ET inhibit sPLA2-IIA expression, whereas ET-deficient strains induced this expression. This stimulatory effect, mediated partly by the cell wall peptidoglycan, can be counterbalanced by ET. We conclude that B. anthracis down-regulates sPLA2-IIA expression in AMs through a process involving ET. Our study, therefore, describes a new molecular mechanism implemented by B. anthracis to escape innate host defense. These pioneering data will provide new molecular targets for future intervention against this deathly pathogen. All mammals are susceptible to infection by Bacillus anthracis, the etiological agent of anthrax. Infection can occur either accidentally or as a potential consequence of a terrorism threat. Pulmonary infection is the most life-threatening form of the disease, causing a near 100% mortality. Despite appropriate therapy, all forms of infection may progress to fatal systemic anthrax, characterized by sepsis and respiratory failure. Thus, it is important to understand the mechanisms of host defense against B. anthracis. We have previously shown that alveolar macrophages produce an enzyme involved in innate defense that can kill B. anthracis: the enzyme is known as secreted phospholipase A2-IIA (sPLA2-IIA). The alveolar macrophage is one of the first cell types to come in contact with B. anthracis. In this study, we show that live B. anthracis spores stimulate the synthesis of sPLA2-IIA, this stimulation being counterbalanced by the inhibitory effect of the edema toxin produced by germinated spores and bacilli. Our study suggests that inhibition of sPLA2-IIA synthesis by edema toxin is a mechanism by which B. anthracis can escape innate host defense. These pioneering data provide new molecular targets for future intervention against this deadly pathogen.
Collapse
Affiliation(s)
- Benoit Raymond
- Institut Pasteur, Unité de Défense Innée et Inflammation, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhang F, Sha J, Wood TG, Galindo CL, Garner HR, Burkart MF, Suarez G, Sierra JC, Agar SL, Peterson JW, Chopra AK. Alteration in the activation state of new inflammation-associated targets by phospholipase A2-activating protein (PLAA). Cell Signal 2008; 20:844-61. [PMID: 18291623 PMCID: PMC2729133 DOI: 10.1016/j.cellsig.2008.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Revised: 12/29/2007] [Accepted: 01/01/2008] [Indexed: 01/08/2023]
Abstract
Phospholipase A(2) (PLA(2))-activating protein (PLAA) is a novel signaling molecule that regulates the production of prostaglandins (PGE(2)) and tumor necrosis factor (TNF)-alpha. To characterize the function of native PLAA in situ, we generated HeLa (Tet-off) cells overexpressing plaa (plaa(high)) and control (plaa(low)) cells, with the plaa gene in opposite orientation in the latter construct. The plaa(high) cells produced significantly more PGE(2) and interleukin (IL)-6 compared to plaa(low) cells in response to TNF-alpha. There was an increased activation and/or expression of cytosolic PLA(2), cyclooxgenase-2, and NF-kappaB after induction of plaa(high) cells with TNF-alpha compared to the respective plaa(low) cells. Microarray analysis of plaa(high) cells followed by functional assays revealed increased production of proinflammatory cytokine IL-32 and a decrease in the production of annexin A4 and clusterin compared to plaa(low) cells. We demonstrated the role of annexin A4 as an inhibitor of PLA(2) and showed that addition of exogeneous clusterin limited the production of PGE(2) from plaa(high) cells. To understand regulation of plaa gene expression, we used a luciferase reporter system in HeLa cells and identified one stimulatory element, with Sp1 binding sites, and one inhibitory element, in exon 1 of the plaa gene. By using decoy DNA oligonucleotides to Sp1 and competitive binding assays, we showed that Sp1 maintains basal expression of the plaa gene and binds to the above-mentioned stimulatory element. We demonstrated for the first time that the induction of native PLAA by TNF-alpha can perpetuate inflammation by enhancing activation of PLA(2) and NF-kappaB.
Collapse
Affiliation(s)
- Fan Zhang
- Departments of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555-1070
| | - Jian Sha
- Departments of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555-1070
| | - Thomas. G. Wood
- Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77555-1070
| | - Cristi. L. Galindo
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical School, Dallas, TX 75390-8591
| | - Harold R. Garner
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical School, Dallas, TX 75390-8591
| | - Mark. F. Burkart
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical School, Dallas, TX 75390-8591
| | - Giovanni Suarez
- Departments of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555-1070
| | - Johanna. C. Sierra
- Departments of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555-1070
| | - Stacy. L. Agar
- Departments of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555-1070
| | - Johnny W. Peterson
- Departments of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555-1070
| | - Ashok K. Chopra
- Departments of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555-1070
| |
Collapse
|
30
|
Clément N, Gueguen M, Glorian M, Blaise R, Andréani M, Brou C, Bausero P, Limon I. Notch3 and IL-1beta exert opposing effects on a vascular smooth muscle cell inflammatory pathway in which NF-kappaB drives crosstalk. J Cell Sci 2008; 120:3352-61. [PMID: 17881497 DOI: 10.1242/jcs.007872] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Atherogenesis begins with the transfer of monocytes from the lumen to the intimal layer of arteries. The paracrine activity acquired by these monocytes shifts vascular smooth muscle cells from a contractile-quiescent to a secretory-proliferative phenotype, allowing them to survive and migrate in the intima. Transformed and relocated, they also start to produce and/or secrete inflammatory enzymes, converting them into inflammatory cells. Activation of the Notch pathway, a crucial determinant of cell fate, regulates some of the new features acquired by these cells as it triggers vascular smooth muscle cells to grow and inhibits their death and migration. Here, we evaluate whether and how the Notch pathway regulates the cell transition towards an inflammatory or de-differentiated state. Activation of the Notch pathway by the notch ligand Delta1, as well as overexpression of the active form of Notch3, prevents this phenomenon [initiated by interleukin 1beta (IL-1beta)], whereas inhibiting the Notch pathway enhances the transition. IL-1beta decreases the expression of Notch3 and Notch target genes. As shown by using an IkappaBalpha-mutated form, the decrease of Notch3 signaling elements occurs subsequent to dissociation of the NF-kappaB complex. These results demonstrate that the Notch3 pathway is attenuated through NF-kappaB activation, allowing vascular smooth muscle cells to switch into an inflammatory state.
Collapse
MESH Headings
- Amyloid Precursor Protein Secretases/antagonists & inhibitors
- Amyloid Precursor Protein Secretases/metabolism
- Animals
- Aorta/anatomy & histology
- Aorta/metabolism
- Biomarkers/metabolism
- Cell Communication/physiology
- Cells, Cultured
- Dinoprostone/metabolism
- Gene Expression Regulation
- Humans
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism
- Inflammation/metabolism
- Interleukin-1beta/genetics
- Interleukin-1beta/metabolism
- Male
- Muscle Contraction/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- NF-kappa B/metabolism
- Phospholipases A2/metabolism
- Rats
- Rats, Wistar
- Receptor, Notch3
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Nathalie Clément
- UMR 7079 de Physiologie et Physiopathologie, Université Pierre et Marie Curie, CNRS, 7 quai Saint-Bernard 75252 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Menschikowski M, Hagelgans A, Hempel U, Lattke P, Ismailov I, Siegert G. On interaction of activated protein C with human aortic smooth muscle cells attenuating the secretory group IIA phospholipase A2 expression. Thromb Res 2007; 122:69-76. [PMID: 17936881 DOI: 10.1016/j.thromres.2007.08.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 08/14/2007] [Accepted: 08/30/2007] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Pharmacological restriction of secretory group IIA phospholipase A(2) (sPLA(2)-IIA) expression is thought to be beneficial in the treatment of inflammatory diseases such as sepsis and septic shock. In this study we investigated the effects of activated protein C (APC) on sPLA(2)-IIA expression, phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, and on DNA-binding activities of nuclear factor-kappaB (NF-kappaB) and CCAAT box enhancer binding protein-beta (C/EBP-beta) in human aortic smooth muscle cells (HASMC). MATERIALS AND METHODS To achieve elevated sPLA(2)-IIA production as occurring during inflammation, HASMC were stimulated with interferon-gamma (IFN-gamma) alone and in combination with other inductors, thus modeling the strong sPLA(2)-IIA elevation by inflammation. RESULTS AND CONCLUSIONS APC inhibited the stimulated expression of sPLA(2)-IIA in HASMC dose-dependently (1-300 nM). At the same time, APC increased the phosphorylation of ERK 1/2 and decreased NF-kappaB and C/EBP-beta DNA-binding activities in these cells, as compared with respective stimulated controls. Reverse transcriptase-polymerase chain reaction and cell-based ELISA reveal an endothelial protein C receptor (EPCR) expression in HASMC. Application of antibodies against EPCR and protease-activated receptor-1 (PAR-1) reduced the APC-induced ERK 1/2 activation and the treatment of cells with a PAR-1 antagonist diminished the sPLA(2)-IIA inhibition. The obtained results show that APC effectively suppresses the up-regulated sPLA(2)-IIA expression, which might contribute to the reported beneficial effects of APC in the treatment of severe inflammatory disorders.
Collapse
Affiliation(s)
- Mario Menschikowski
- Institute of Clinical Chemistry and Laboratory Medicine, Technical University of Dresden, Medical Faculty Carl Gustav Carus, Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
32
|
Ravaux L, Denoyelle C, Monne C, Limon I, Raymondjean M, El Hadri K. Inhibition of interleukin-1beta-induced group IIA secretory phospholipase A2 expression by peroxisome proliferator-activated receptors (PPARs) in rat vascular smooth muscle cells: cooperation between PPARbeta and the proto-oncogene BCL-6. Mol Cell Biol 2007; 27:8374-87. [PMID: 17908795 PMCID: PMC2169168 DOI: 10.1128/mcb.00623-07] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The inflammation that occurs during atherosclerosis is characterized by the release of large amounts of group IIA secretory phospholipase A2 (sPLA2-IIA). This study was designed to define the function of the three peroxisome proliferator-activated receptors (PPARs) on sPLA2 expression in vascular smooth muscle cells (VSMCs). We found that PPAR ligands decreased sPLA2-IIA activity and inhibited mRNA accumulation under inflammatory conditions. Furthermore, interleukin-1beta-induced sPLA2-IIA promoter activity was inhibited by the three PPAR ligands and in a similar way when cells were cotransfected with PPARalpha, PPARbeta, or PPARgamma, plus retinoid X receptor alpha (RXRalpha). Our study revealed that the regulation of sPLA2-IIA gene transcription by PPARalpha/RXR and PPARgamma/RXR heterodimers requires an interaction with a PPAR response element (PPRE) of the sPLA2-IIA promoter. In contrast, PPARbeta operates through a PPRE-independent mechanism. In addition, we demonstrated that VSMCs expressed the transcriptional repressor BCL-6. Overexpression of BCL-6 markedly reduced sPLA2-IIA promoter activity in VSMCs, while a dominant negative form of BCL-6 abrogated sPLA2 repression by PPARbeta. The PPARbeta agonist induced a BCL-6 binding to the sPLA2 promoter in VSMCs under inflammatory conditions. The knockdown of BCL-6 by short interfering RNA abolished the inhibitory effect of the PPARbeta ligand on sPLA2 activity and prostaglandin E2 release. Thus, the inhibition of sPLA2-IIA activity by PPARbeta agonists may provide a promising approach to impacting the initiation and progression of atherosclerosis.
Collapse
MESH Headings
- Animals
- Cattle
- Enzyme Induction/drug effects
- Gene Expression Regulation, Enzymologic/drug effects
- Group II Phospholipases A2/biosynthesis
- Group II Phospholipases A2/genetics
- Interleukin-1beta/pharmacology
- Ligands
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- PPAR-beta/metabolism
- Protein Binding/drug effects
- Proto-Oncogene Proteins c-bcl-6/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- Rats
- Rats, Wistar
- Repressor Proteins/metabolism
- Response Elements
- Retinoid X Receptors/metabolism
- Sequence Deletion
Collapse
Affiliation(s)
- Lucas Ravaux
- UMR Physiologie et Physiopathologie, Université Pierre et Marie Curie, CNRS, 7 quai Saint-Bernard, 75252 Paris, France
| | | | | | | | | | | |
Collapse
|
33
|
Jaulmes A, Thierry S, Janvier B, Raymondjean M, Maréchal V. Activation of sPLA2-IIA and PGE2 production by high mobility group protein B1 in vascular smooth muscle cells sensitized by IL-1beta. FASEB J 2006; 20:1727-9. [PMID: 16807371 DOI: 10.1096/fj.05-5514fje] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Lipid mediators such as prostaglandin E2 (PGE2) play a central role during atherogenesis as a consequence of inflammation. PGE2 is produced from phospholipids by a cascade of enzymatic reactions involving phospholipase A2 (PLA2), cyclooxygenase (COX), and prostaglandin E synthase (PGES). It is released by several cell types, including vascular smooth muscle cells (VSMCs). Recent work has shown that the secretory PLA2-IIA (sPLA2-IIA), the most abundant isoform of secreted PLA2 in VSMCs, acts as a potent cytokine and activates VSMCs through a positive feedback loop. High mobility group protein 1 (HMGB1), also known as amphoterin, is a ubiquitous protein that plays various roles in the nucleus. HMGB1 is released by necrotic cells and by immune cells in response to various inflammatory mediators and acts as a potent proinflammatory cytokine. The present study investigates the role of HMGB1 in the activation of sPLA2-IIA expression and PGE2 production in VSMCs. Recombinant HMGB1 slightly activated the sPLA2-IIA, COX-2, and mPGES-1 genes but dramatically stimulated these genes in VSMCs that had been incubated with the proinflammatory cytokine IL-1beta for 24 h. This effect was accompanied by significantly increased PGE2 release. Induction of the three known receptors of HMGB1, namely RAGE, TLR-2, and TLR-4, by IL-1beta suggests that proinflammatory cytokines sensitize VSMCs to HMGB1. This provides new insights into the role of HMGB1 in VSMCs, suggesting it may be essential for the progression of atherosclerosis.
Collapse
Affiliation(s)
- Amandine Jaulmes
- UMR Physiologie et Physiopathologie, Université Pierre et Marie Curie, CNRS, Paris, France
| | | | | | | | | |
Collapse
|
34
|
Menschikowski M, Hagelgans A, Siegert G. Secretory phospholipase A2 of group IIA: Is it an offensive or a defensive player during atherosclerosis and other inflammatory diseases? Prostaglandins Other Lipid Mediat 2006; 79:1-33. [PMID: 16516807 DOI: 10.1016/j.prostaglandins.2005.10.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Revised: 10/29/2005] [Accepted: 10/31/2005] [Indexed: 02/07/2023]
Abstract
Since its discovery in the serum of patients with severe inflammation and in rheumatoid arthritic fluids, the secretory phospholipase A2 of group IIA (sPLA2-IIA) has been chiefly considered as a proinflammatory enzyme, the result of which has been very intense interest in selective inhibitors of sPLA2-IIA in the hope of developing new and efficient therapies for inflammatory diseases. The recent discovery of the antibacterial properties of sPLA2-IIA, however, has raised the question of whether the upregulation of sPLA2-IIA during inflammation is to be considered uniformly negative and the hindrance of sPLA2-IIA in every instance beneficial. The aim of this review is for this reason, along with the results of various investigations which argue for the proinflammatory and proatherogenic effects of an upregulation of sPLA2-IIA, also to array data alongside which point to a protective function of sPLA2-IIA during inflammation. Thus, it could be shown that sPLA2-IIA, apart from the bactericidal effects, possesses also antithrombotic properties and indeed plays a possible role in the resolution of inflammation and the accelerated clearance of oxidatively modified lipoproteins during inflammation via the liver and adrenals. Based on these multipotent properties the knowledge of the function of sPLA2-IIA during inflammation is a fundamental prerequisite for the development and establishment of new therapeutic strategies to prevent and treat severe inflammatory diseases up to and including sepsis.
Collapse
Affiliation(s)
- Mario Menschikowski
- Technische Universität Dresden, Medizinische Fakultät Carl Gustav Carus, Institut für Klinische Chemie and Laboratoriumsmedizin, Fetscherstrasse 74, D-01307 Dresden, Germany.
| | | | | |
Collapse
|
35
|
Naito T, Tanihata Y, Nishimura H, Tanaka T, Higuchi C, Taguchi T, Sanaka T. Expression of matrix metalloproteinase-9 associated with ets-1 proto-oncogene in rat tubulointerstitial cells. Nephrol Dial Transplant 2005; 20:2333-48. [PMID: 16046515 DOI: 10.1093/ndt/gfi013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Ets-1 proto-oncogene exhibits multiple activities in the transcriptional regulation of numerous genes including metalloproteinase (MMP)-1, -3 and -9. MMPs play an important role in the remodelling of extracellular matrix in various renal diseases. However, the role of the Ets-1-MMP axis in advanced renal diseases is uncertain. In the present study, we investigated whether Ets-1 is involved in interleukin (IL)-1-mediated expression of MMPs in tubulointerstitial cells. METHODS Rat renal fibroblasts (NRK-49F) and tubular epithelial cells (NRK-52E) were cultured and allocated to an IL-1beta-treated group (10 ng/ml), a platelet-derived growth factor (PDGF)-BB-treated group (25 ng/ml) and a control group. Protein and mRNA were extracted after 1, 6, 12 and 24 h of treatment. Parallel flasks were treated with 2 muM ets-1 antisense oligodeoxynucleotides (ODNs) before exposure to IL-1beta. The expression of Ets-1 protein was evaluated by western blotting. The activities of MMPs were evaluated by gelatin zymography. The expression of ets-1 and/or MMP-9 mRNA was evaluated semiquantitatively by real-time reverse transcription-polymerase chain reaction (RT-PCR). RESULTS In NRK-49F cells, Ets-1 protein increased significantly by 6.8-fold at 6 h, and MMP-9 activity increased significantly by 9.9-fold at 12 h in the IL-1beta-treated group compared with controls. MMP-2 and -3 activities also increased significantly in the IL-1beta-treated group. In NRK-52E cells, Ets-1 protein was 3.1 times higher at 1 h, and the latent form of MMP-9 activity increased 3.4-fold at 6 h in the IL-1beta group compared with controls. However, MMP-2 or MMP-3 activities were not markedly altered by IL-1beta treatment compared with controls. When the cells were treated with ets-1 antisense ODNs before IL-1beta treatment, Ets-1 protein expression decreased at least 50%, and MMP-9 activity was clearly inhibited in both cells. We also confirmed that MMP-9 activity was upregulated on days 21 and 28 in renal cortex of rat crescentic glomerulonephritis. CONCLUSIONS The Ets-1 transcriptional factor may participate in IL-1beta-mediated MMP-9 expression in tubulointerstitial cells.
Collapse
Affiliation(s)
- Takashi Naito
- Department of Medicine, Tokyo Women's Medical University Daini Hospital, Arakawaku, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
36
|
Jaulmes A, Janvier B, Andreani M, Raymondjean M. Autocrine and Paracrine Transcriptional Regulation of Type IIA Secretory Phospholipase A2 Gene in Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2005; 25:1161-7. [PMID: 15802623 DOI: 10.1161/01.atv.0000164310.67356.a9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
The inflammation that occurs during the development of atherosclerosis is characterized by a massive release of sPLA2-IIA (group IIA secretory phospholipase A2) from vascular smooth muscle cells (VSMCs). We have investigated the autocrine function of sPLA2-IIA in rat aortic and human VSMCs.
Methods and Results—
We found that the transcription of the endogenous sPLA2-IIA gene increased by adding a cell supernatant containing human sPLA2-IIA proteins. We show that this effect was independent of the sPLA2 activity using sPLA2-IIA proteins lacking enzyme activity. Transient transfections with various sPLA2-IIA rat promoter-luciferase constructs demonstrated that the C/EBP, NK-κB, and Ets transcription factors are involved in the increase in sPLA2-IIA gene transcription. We also found the M-type sPLA2 receptor mRNA in VSMCs, and we showed that the sPLA2-luciferase reporter gene was induced by the specific agonist of the sPLA2 receptor, aminophenylmannopyranoside (APMP), and that this induction was mediated by the same transcription factor-binding sites. Finally, we used a sPLA2-IIA mutant unable to bind heparan-sulfate proteoglycans to show that the binding of wild-type sPLA2-IIA to proteoglycans is essential for the induction of an autocrine loop.
Conclusions—
We have thus identified new autocrine and paracrine pathways activating sPLA2-IIA gene expression in rat and human VSMCs.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Autocrine Communication/physiology
- CCAAT-Enhancer-Binding Proteins/metabolism
- Cells, Cultured
- Gene Expression Regulation, Enzymologic/physiology
- Group II Phospholipases A2
- Humans
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/enzymology
- NF-kappa B/metabolism
- Paracrine Communication/physiology
- Phospholipases A/genetics
- Phospholipases A/metabolism
- Phospholipases A2
- Protein Binding
- Proteoglycans/metabolism
- Rats
- Rats, Wistar
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Phospholipase A2
- Transcriptional Activation/physiology
- Winged-Helix Transcription Factors/metabolism
Collapse
Affiliation(s)
- Amandine Jaulmes
- UMR Physiologie et Physiopathologie, Université Pierre et Marie Curie, Paris, France
| | | | | | | |
Collapse
|
37
|
Menschikowski M, Hagelgans A, Heyne B, Hempel U, Neumeister V, Goez P, Jaross W, Siegert G. Statins potentiate the IFN-γ-induced upregulation of group IIA phospholipase A2 in human aortic smooth muscle cells and HepG2 hepatoma cells. Biochim Biophys Acta Mol Cell Biol Lipids 2005; 1733:157-71. [PMID: 15863363 DOI: 10.1016/j.bbalip.2005.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2004] [Revised: 12/20/2004] [Accepted: 01/10/2005] [Indexed: 01/06/2023]
Abstract
The present study shows that the incubation of human aortic smooth muscle cells (HASMC) and HepG2 cells with atorvastatin and mevastatin as HMG-CoA reductase inhibitors potentiated the interferon-gamma (INF-gamma)-induced group IIA phospholipase A(2) (sPLA(2)-IIA) expression in a dose- and time-dependent manner. The effect of statins on sPLA(2)-IIA expression was reduced by mevalonate, farnesyl pyrophosphate and geranylgeranyl pyrophosphate. Inversely, inhibitors of the farnesyl transferase and geranylgeranyl transferase-I mimicked the effects of statins. Clostridium difficile toxin B (TcdB), Y-27632 and H-1152, functioning as inhibitors of Rho proteins and Rho-associated kinase, also augmented the sPLA(2)-IIA expression in combination with IFN-gamma. The same effects were observed when inhibitors of mitogen-activated/extracellular response protein kinase kinase (MEK), PD98059 or U0126 were used. Further, the Janus kinase-2 (Jak2)-specific inhibitor, AG-490 and inhibitors of nuclear factor-kappaB (NFkappaB) abrogated the sPLA(2)-IIA elevating effects of statins, TcdB and PD98059 in the presence of IFN-gamma. This cytokine alone increased the NFkappaB p65 and CAAT-enhancer-binding protein-beta (C/EBP-beta) activity in HASMC nuclear extract, but only C/EBP-beta was further augmented when the cells were incubated in addition to IFN-gamma with atorvastatin, H-1152, PD98059 or U0126. Moreover, after the incubation of cells with atorvastatin and IFN-gamma the stability of sPLA-(2)IIA mRNA significantly increased in comparison to those after incubation with IFN-gamma alone. In conclusion, the obtained data suggest that (i) the expression of sPLA(2)-IIA is negatively regulated by RhoA/Rho-associated kinase and MEK/ERK signaling pathways and (ii) statins, because of their ability to down-regulate these pathways, can potentiate the IFN-gamma-induced sPLA(2)-II expression at transcriptional and post-transcriptional levels.
Collapse
Affiliation(s)
- Mario Menschikowski
- Technische Universität Dresden, Medizinische Fakultät Carl Gustav Carus, Institut für Klinische Chemie und Laboratoriumsmedizin, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Menschikowski M, Hagelgans A, Hempel U, Siegert G. Glycogen synthase kinase-3beta negatively regulates group IIA phospholipase A2 expression in human aortic smooth muscle and HepG2 hepatoma cells. FEBS Lett 2005; 577:81-6. [PMID: 15527765 DOI: 10.1016/j.febslet.2004.09.067] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2004] [Revised: 09/14/2004] [Accepted: 09/17/2004] [Indexed: 12/24/2022]
Abstract
The present study shows that the IFN-gamma-mediated upregulation of secretory phospholipase A2 of group IIA (sPLA2-IIA) in HASMC and HepG2 cells is synergistically increased after simultaneous inhibition of glycogen synthase kinase-3beta (GSK-3beta) by indirubin-3'-monoxime, 5-iodo or AR-A014418. The effect of GSK-3beta inhibition was dose- and time-dependent and can be further augmented by its concomitant incubation with Clostridium difficile toxin B, an inhibitor of small Rho proteins, or H-1152, an inhibitor of Rho-associated kinase. Using AG-490 and caffeic acid phenethyl ester (CAPE), it is further demonstrated that the effect of GSK-3beta inhibition on sPLA2-IIA expression depends on Janus kinase-2 and NF-kappaB-signaling.
Collapse
Affiliation(s)
- Mario Menschikowski
- Medizinische Fakultät Carl Gustav Carus, Institut für Klinische Chemie und Laboratoriumsmedizin, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany.
| | | | | | | |
Collapse
|
39
|
Antonio V, Janvier B, Brouillet A, Andreani M, Raymondjean M. Oxysterol and 9-cis-retinoic acid stimulate the group IIA secretory phospholipase A2 gene in rat smooth-muscle cells. Biochem J 2003; 376:351-60. [PMID: 12882648 PMCID: PMC1223770 DOI: 10.1042/bj20030098] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2003] [Revised: 07/09/2003] [Accepted: 07/28/2003] [Indexed: 01/26/2023]
Abstract
The inflammation that occurs during rheumatoid arthritis or atherosclerosis is characterized by the release of large amounts of sPLA(2) (group IIA secretory phospholipase A(2)). We have shown previously that the sPLA(2) promoter in SMC (smooth-muscle cells) is activated by interleukin-1beta and cAMP-signalling pathways, through the interplay of multiple transcription factors [Antonio, Brouillet, Janvier, Monne, Bereziat, Andreani, and Raymondjean (2002) Biochem. J. 368, 415-424]. In the present study, we have investigated the regulation of sPLA(2) gene expression in rat aortic SMCs by oxysterols. We found that oxysterol ligands that bind to the LXR (liver X receptor), including 25-HC (25-hydroxycholesterol) and 22( R )-HC, cause the accumulation of sPLA(2) mRNA and an increased enzyme activity. Transient transfection experiments demonstrated that the sPLA(2) promoter is synergistically activated by 22( R )-HC in combination with 9- cis -retinoic acid, a ligand for the LXR heterodimeric partner RXR (retinoid X receptor). Promoter activity was also increased in a sterol-responsive fashion when cells were co-transfected with LXRalpha/RXRalpha or LXRbeta/RXRalpha. Mutagenesis studies and gel mobility-shift assays revealed that LXR/RXR heterodimers regulate sPLA(2) transcription directly, by interacting with a degenerated LXRE (LXR response element) at position [-421/-406] of the sPLA(2) promoter. Chromatin immunoprecipitation revealed the in vivo occupancy of LXR on the sPLA(2) promoter. In addition, the orphan nuclear receptor LRH-1 (liver receptor homologue-1) potentiated the sterol-dependent regulation of the sPLA(2) promoter by binding to an identified promoter element (TCAAGGCTG). Finally, we have demonstrated that oxysterols act independent of interleukin-1beta and cAMP pathways to activate the sPLA(2) promoter. In the present study, we have identified a new pathway activating sPLA(2) gene expression in SMCs.
Collapse
MESH Headings
- Alitretinoin
- Animals
- Cells, Cultured
- Cyclic AMP/metabolism
- DNA-Binding Proteins
- Enzyme Induction
- Group II Phospholipases A2
- Hydroxycholesterols/pharmacology
- Interleukin-1/pharmacology
- Liver X Receptors
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Orphan Nuclear Receptors
- Phospholipases A/biosynthesis
- Phospholipases A/genetics
- Phospholipases A/metabolism
- Phospholipases A2
- Promoter Regions, Genetic
- RNA, Messenger/biosynthesis
- Rats
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Retinoic Acid/metabolism
- Response Elements
- Retinoid X Receptors
- Transcription Factors/metabolism
- Transcriptional Activation
- Tretinoin/pharmacology
Collapse
Affiliation(s)
- Valérie Antonio
- UMR (Unité Mixte de Recherche 7079) Physiologie et Physiopathologie, Université Pierre et Marie Curie, Case courrier 256, Bâtiment A, 5èmeétage, 7 quai St Bernard, 75252 Paris cedex 05, France
| | | | | | | | | |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Inflammation is an integral feature of atherosclerosis, in which inflammatory processes contribute to the initiation, progression and rupture of lipid-rich atherosclerotic plaques. Recent studies have suggested the involvement of the proinflammatory secretory phospholipase A2 (sPLA2)-IIA in the development of atherosclerosis. This enzyme has been proposed to hydrolyze phosphatidylcholine (PC) in lipoproteins to liberate lyso-PC and free fatty acids in the arterial wall, thereby facilitating the accumulation of bioactive lipids and modified lipoproteins in atherosclerotic foci. However, the recent discovery of several novel sPLA2 isozymes has raised the question of which types of sPLA2 truly contribute to the atherosclerotic process. RECENT FINDINGS Amongst the 10 mammalian sPLA2 isozymes, sPLA2-X, -V, -IIF and -III exhibit much more potent PC-hydrolyzing activity than do the others, and can release free fatty acids and lysophospholipids from the PC-rich outer leaflet of the cellular plasma membrane. In particular, sPLA2-X and sPLA2-V hydrolyze PC in lipoproteins far more efficiently than does sPLA2-IIA. Moreover, sPLA2-X promotes foam cell formation in vitro and is expressed in the atherosclerotic arterial walls of apolipoprotein E deficient mice in vivo. SUMMARY PC-hydrolyzing sPLA2 isozymes, particularly sPLA2-V and sPLA2-X, are attractive candidates for proatherosclerotic factors that may act in place of sPLA2-IIA. However, their expression in human atherosclerotic lesions requires confirmation by specific methods that can distinguish between the different sPLA2 isozymes.
Collapse
Affiliation(s)
- Makoto Murakami
- Department of Health Chemistry, School of Pharmaceutical Sciences, Showa University, Tokyo, Japan.
| | | |
Collapse
|
41
|
Ares MPS. Atherosclerosis: cell biology and lipoproteins. Curr Opin Lipidol 2003; 14:393-6. [PMID: 12865738 DOI: 10.1097/00041433-200308000-00010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
42
|
Hoshikawa Y, Nana-Sinkam P, Moore MD, Sotto-Santiago S, Phang T, Keith RL, Morris KG, Kondo T, Tuder RM, Voelkel NF, Geraci MW. Hypoxia induces different genes in the lungs of rats compared with mice. Physiol Genomics 2003; 12:209-19. [PMID: 12464684 DOI: 10.1152/physiolgenomics.00081.2001] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Different animal species have a varying response to hypoxia. Mice develop less pulmonary artery thickening after chronic hypoxia exposure than rats. We hypothesized that the lung tissue gene expression pattern displayed in hypoxic rats would differ from that of hypoxic mice. We exposed Sprague-Dawley rats and C57BL/6 mice to both 1 and 3 wk of hypobaric hypoxia. Although both species developed pulmonary hypertension, mice showed less pulmonary vascular remodeling than rats. Microarray gene analysis demonstrated a distinct pattern of gene expression between mice and rats when exposed to hypoxic conditions. In addition, some genes appeared to be more responsive at an earlier time point of 1 wk of hypoxia. Hypoxic conditions in the rat induce genes involved in endothelial cell proliferation, repression of apoptosis, and vasodilation. Mice exposed to hypoxic conditions decrease the expression of genes involved in vasodilation and in endothelial cell proliferation. Although we cannot determine whether the differential expression of genes during chronic hypoxia is cause or consequence of the differential pulmonary vascular remodeling, we propose that a balance between over- and under-expression of a selective group of genes may be responsible for lung vascular remodeling and vascular tone control.
Collapse
Affiliation(s)
- Yasushi Hoshikawa
- Pulmonary Hypertension Center, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|