1
|
Karpurapu M, Nie Y, Chung S, Yan J, Dougherty P, Pannu S, Wisler J, Harkless R, Parinandi N, Berdyshev E, Pei D, Christman JW. The calcineurin-NFATc pathway modulates the lipid mediators in BAL fluid extracellular vesicles, thereby regulating microvascular endothelial cell barrier function. Front Physiol 2024; 15:1378565. [PMID: 38812883 PMCID: PMC11133699 DOI: 10.3389/fphys.2024.1378565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/16/2024] [Indexed: 05/31/2024] Open
Abstract
Extracellular vesicles mediate intercellular communication by transporting biologically active macromolecules. Our prior studies have demonstrated that the nuclear factor of activated T cell cytoplasmic member 3 (NFATc3) is activated in mouse pulmonary macrophages in response to lipopolysaccharide (LPS). Inhibition of NFATc3 activation by a novel cell-permeable calcineurin peptide inhibitor CNI103 mitigated the development of acute lung injury (ALI) in LPS-treated mice. Although pro-inflammatory lipid mediators are known contributors to lung inflammation and injury, it remains unclear whether the calcineurin-NFATc pathway regulates extracellular vesicle (EV) lipid content and if this content contributes to ALI pathogenesis. In this study, EVs from mouse bronchoalveolar lavage fluid (BALF) were analyzed for their lipid mediators by liquid chromatography in conjunction with mass spectrometry (LC-MS/MS). Our data demonstrate that EVs from LPS-treated mice contained significantly higher levels of arachidonic acid (AA) metabolites, which were found in low levels by prior treatment with CNI103. The catalytic activity of lung tissue cytoplasmic phospholipase A2 (cPLA2) increased during ALI, correlating with an increased amount of arachidonic acid (AA) in the EVs. Furthermore, ALI is associated with increased expression of cPLA2, cyclooxygenase 2 (COX2), and lipoxygenases (5-LOX, 12-LOX, and 15-LOX) in lung tissue, and pretreatment with CNI103 inhibited the catalytic activity of cPLA2 and the expression of cPLA2, COX, and LOX transcripts. Furthermore, co-culture of mouse pulmonary microvascular endothelial cell (PMVEC) monolayer and NFAT-luciferase reporter macrophages with BALF EVs from LPS-treated mice increased the pulmonary microvascular endothelial cell (PMVEC) monolayer barrier permeability and luciferase activity in macrophages. However, EVs from CNI103-treated mice had no negative impact on PMVEC monolayer barrier integrity. In summary, BALF EVs from LPS-treated mice carry biologically active NFATc-dependent, AA-derived lipids that play a role in regulating PMVEC monolayer barrier function.
Collapse
Affiliation(s)
- Manjula Karpurapu
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Sangwoon Chung
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Jiasheng Yan
- Department of Pharmacology, Ohio State University, Columbus, OH, United States
| | - Patrick Dougherty
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| | - Sonal Pannu
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Jon Wisler
- Department of Surgery, Ohio State Wexner Medical Center, Columbus, OH, United States
| | - Ryan Harkless
- Department of Surgery, Ohio State Wexner Medical Center, Columbus, OH, United States
| | - Narasimham Parinandi
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Evgeny Berdyshev
- Division of Pulmonary Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, United States
| | - Dehua Pei
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| | - John W. Christman
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| |
Collapse
|
2
|
Li X, Bai Y, Ma Y, Li Y. Ameliorating effects of berberine on sepsis-associated lung inflammation induced by lipopolysaccharide: molecular mechanisms and preclinical evidence. Pharmacol Rep 2023:10.1007/s43440-023-00492-2. [PMID: 37184743 DOI: 10.1007/s43440-023-00492-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
As a life-threatening disorder, sepsis-associated lung injury is a dysregulated inflammatory response to microbial infection, characterized by the infiltration of inflammatory cells into lung tissues and excessive production of pro-inflammatory mediators. Therefore, immunomodulatory/anti-inflammatory agents are a potential treatment for sepsis-associated lung injury. Berberine, one of the well-studied medicinal plant-derived compounds, has shown promising anti-inflammatory potential in inflammatory conditions, through modulating excessive immune responses induced by various immune cells. A systematic literature search in electronic databases indicated several publications that studied the effect of berberine on lipopolysaccharide (LPS)-induced sepsis in preclinical investigations. The current review article aims to provide evidence on the effects of berberine against LPS-induced acute lung injury (ALI), together with underlying molecular mechanisms. The findings reveal that berberine through inhibiting the excessive production of multiple pro-inflammatory cytokines, suppressing the infiltration of immune cells into lung tissues, as well as preventing pulmonary edema and coagulation, can relieve pulmonary histopathological changes from LPS-mediated inflammation, thereby attenuating sepsis-associated lung injury and lethality in the experimental models. In conclusion, berberine shows great potential as a preventing and therapeutic agent for sepsis-associated lung injury, however, further proof-of-concept studies and clinical investigations are warranted for translating these preclinical findings into clinical practices.
Collapse
Affiliation(s)
- Xiaojuan Li
- Department of Critical Care Medicine, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Yi Bai
- Department of Critical Care Medicine, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Yulong Ma
- Department of Critical Care Medicine, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Yan Li
- Department of Critical Care Medicine, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China.
| |
Collapse
|
3
|
Chen Y, Liu H, Tian Y, Luo Z, Ran J, Miao Z, Zhang Q, Yin G, Xie Q. Fexofenadine protects against lipopolysaccharide-induced acute lung injury by targeting cytosolic phospholipase A2. Int Immunopharmacol 2023; 116:109637. [PMID: 36764283 DOI: 10.1016/j.intimp.2022.109637] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 02/11/2023]
Abstract
OBJECTIVE Acute lung injury (ALI) causes acute respiratory distress syndrome, with a high mortality rate of 40%, with currently available pharmacological treatments. Cytosolic phospholipase A2 (cPLA2) plays a critical role in the lipopolysaccharide (LPS)-induced pathology of ALI. This study assessed the therapeutic effects of fexofenadine (FFD), an on-market small-molecule drug that can target cPLA2 in LPS-induced ALI. METHODS Primary macrophages obtained from the bone marrow of wild-type and cPLA2 knockout mice and the alveolar macrophage cell line, MHS were used to test the inhibitory effect of FFD on the cPLA2/ERK/p65 signaling pathway, NF-κB p65 translocation, and cytokine and chemokine production. An LPS-induced ALI mouse model was used to assess the treatment effects of FFD. Flow cytometry detected subsets of macrophages and neutrophils. cPLA2 activity and downstream hydrolysates were detected. Treatment with a cPLA2 inhibitor or NF-κB p65 inhibitor confirmed that FFD functioned through the cPLA2/ERK/p65 signaling pathway by targeting cPLA2. RESULTS FFD reduced the infiltration of macrophages and neutrophils, decreased the protein secretion in bronchoalveolar lavage fluid, and reduced the production of TNFα, IL-1β, IL-6, MCP-1, and IL-8 in the lung, bronchoalveolar lavage fluid, and sera of LPS-induced ALI mice. FFD inhibited cPLA2 activity, suppressed the cPLA2/ERK/p65 signaling pathway, inhibited translocation of p65, and decreased the production of cytokines, chemokines, and downstream hydrolysates of cPLA2, arachidonic acid, and leukotriene B4. CONCLUSION FFD inhibits the cPLA2/ERK/p65 signaling pathway by targeting cPLA2. Therefore, FFD is promising as a therapeutic against cPLA2-involved diseases, particularly ALI.
Collapse
Affiliation(s)
- Yuehong Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Huan Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yunru Tian
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhongling Luo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jingjing Ran
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhiyong Miao
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qiuping Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Geng Yin
- Department of General Practice, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
4
|
Chen P, Liu H, Xin H, Cheng B, Sun C, Liu Y, Liu T, Wen Z, Cheng Y. Inhibiting the Cytosolic Phospholipase A2-Arachidonic Acid Pathway With Arachidonyl Trifluoromethyl Ketone Attenuates Radiation-Induced Lung Fibrosis. Int J Radiat Oncol Biol Phys 2023; 115:476-489. [PMID: 35450754 DOI: 10.1016/j.ijrobp.2022.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE Radiation-induced lung fibrosis (RILF) is a serious late complication of thoracic radiation therapy. Inflammation is crucial in fibroblast activation and RILF, and arachidonic acid (AA) is an important inflammatory mediator released by cytosolic phospholipase A2 (cPLA2) and reduced by arachidonyl trifluoromethyl ketone (ATK)-targeting of cPLA2. Here, we aimed to investigate the roles of the cPLA2/AA pathway in RILF and assess the potential of targeting cPLA2 to prevent RILF. METHODS AND MATERIALS A computed tomography scan was used to obtain the mean lung density, and hematoxylin-eosin, Masson's trichrome, and Sirius Red staining were used to assess the histopathologic conditions in mouse models. AA levels in mouse serum and cell supernatants were tested by enzyme-linked immunosorbent assay. Fibroblast phenotype alterations were examined by a Cell Counting Kit-8, manual cell counting, and a Transwell system. The protein levels were evaluated via Western blotting, immunofluorescence, and immunohistochemistry. RESULTS AA protected fibroblasts against radiation-induced growth inhibition and promoted fibroblast activation, which was characterized by enhanced α-smooth muscle actin expression and migration capacity. Radiation could activate fibroblasts by upregulating cPLA2 expression and AA production, which could be reversed by ATK. Moreover, inhibiting cPLA2 with ATK significantly attenuated collagen deposition and radiation-induced pulmonary fibrosis in mouse models. We further identified extracellular-signal regulated protein kinase (ERK) as the downstream target of the radiation-AA regulatory axis. Radiation-induced AA increased phosphorylated-ERK levels, promoting cyclinD1, cyclin-dependent kinase 6, and α-smooth muscle actin expression and contributing to fibroblast activation. Inhibiting P-ERK impaired radiation- and AA-induced fibroblast activation. The related molecular mechanisms were verified using specimens from animal models. CONCLUSIONS Our findings uncover the role of the cPLA2/AA-ERK regulatory axis in response to radiation in pulmonary fibroblast activation and recognize cPLA2 as the key regulatory molecule during RILF for the first time. Targeting cPLA2 may be a promising protective strategy against RILF.
Collapse
Affiliation(s)
- Pengxiang Chen
- Department of Radiation Oncology; Laboratory of Basic Medical Sciences
| | - Hui Liu
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | | | - Bo Cheng
- Shandong Cancer Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Changhua Sun
- Shandong Institute for Food and Drug Control, Jinan, People's Republic of China
| | | | | | | | | |
Collapse
|
5
|
Kitsiouli E, Tenopoulou M, Papadopoulos S, Lekka ME. Phospholipases A2 as biomarkers in ARDS. Biomed J 2021; 44:663-670. [PMID: 34478892 PMCID: PMC8847824 DOI: 10.1016/j.bj.2021.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a multifactorial life-threatening lung injury, characterized by diffuse lung inflammation and increased alveolocapillary barrier permeability. The different stages of ARDS have distinctive biochemical and clinical profiles. Despite the progress of our understanding on ARDS pathobiology, the mechanisms underlying its pathogenesis are still obscure. Herein, we review the existing literature about the implications of phospholipases 2 (PLA2s), a large family of enzymes that catalyze the hydrolysis of fatty acids at the sn-2 position of glycerophospholipids, in ARDS-related pathology. We emphasize on the versatile way of participation of different PLA2s isoforms in the distinct ARDS subgroup phenotypes by either potentiating lung inflammation and damage or by preserving the normal lung. Current research supports that PLA2s are associated with the progression and the outcome of ARDS. We herein discuss the transcellular communication of PLA2s through secreted extracellular vesicles and suggest it as a new mechanism of PLA2s involvement in ARDS. Thus, the elucidation of the spatiotemporal features of PLA2s expression may give new insights and provide valuable information about the risk of an individual to develop ARDS or advance to more severe stages, and potentially identify PLA2 isoforms as biomarkers and target for pharmacological intervention.
Collapse
Affiliation(s)
- Eirini Kitsiouli
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Margarita Tenopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Stylianos Papadopoulos
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Marilena E Lekka
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
6
|
Kashour T, Halwani R, Arabi YM, Sohail MR, O'Horo JC, Badley AD, Tleyjeh IM. Statins as an adjunctive therapy for COVID-19: the biological and clinical plausibility. Immunopharmacol Immunotoxicol 2021; 43:37-50. [PMID: 33406943 DOI: 10.1080/08923973.2020.1863984] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes the coronavirus disease 2019 (COVID-19) has infected millions of individuals and has claimed hundreds of thousands of human lives worldwide. Patients with underlying cardiovascular conditions are at high risk for SARS-CoV-2 infection, and COVID-19 patients have high incidence of cardiovascular complications such as acute cardiac injury, arrhythmias, heart failure, and thromboembolism. The disease has no approved proven effective therapy and hence repurposing of existing approved drugs has been considered as the fastest treatment approach. Statins have been shown to exhibit lipid lowering dependent and independent cardiovascular protective effects as well as favorable effects in various other pathophysiological states. These beneficial properties of statins are a result of their multiple pleotropic effects that include, anti-inflammatory, immunomodulatory, antithrombotic and antimicrobial properties. In this review, we provide a comprehensive description of the mechanisms of the pleotropic effects of statins, the relevant pre-clinical and clinical data pertinent to their role in infections and acute lung injury, the possible cardiovascular benefits of statins in COVID-19, and the implications of the therapeutic potential of statins in COVID-19 disease. We conclude with the rationale for conducting randomized controlled trials of statins in COVID-19 disease.
Collapse
Affiliation(s)
- Tarek Kashour
- Department of Cardiac Sciences, King Fahad Cardiac Center, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Rabih Halwani
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Yaseen M Arabi
- Intensive Care Department, Ministry of National Guard Health Affairs, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - M Rizwan Sohail
- Section of Infectious Diseases, Baylor College of Medicine Houston, TX, USA.,Division of Infectious Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - John C O'Horo
- Division of Infectious Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Department of Molecular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Imad M Tleyjeh
- Division of Infectious Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Division of Epidemiology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Department of Medical Specialties, Infectious Diseases Section, King Fahad Medical City, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Yu G, Mo S, Gao L, Wen X, Chen S, Long X, Xie X, Deng Y, Ren L, Zang N, Chen S, Liu E. Club cell 10-kDa protein (CC10) inhibits cPLA2/COX2 pathway to alleviate RSV-induced airway inflammation and AHR. Int Immunopharmacol 2020; 83:106327. [DOI: 10.1016/j.intimp.2020.106327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/14/2020] [Accepted: 02/14/2020] [Indexed: 11/25/2022]
|
8
|
Tzouvelekis A, Karampitsakos T, Bouros E, Tzilas V, Liossis SN, Bouros D. Autoimmune Biomarkers, Antibodies, and Immunologic Evaluation of the Patient with Fibrotic Lung Disease. Clin Chest Med 2019; 40:679-691. [DOI: 10.1016/j.ccm.2019.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
9
|
Gowda R, Dinavahi SS, Iyer S, Banerjee S, Neves RI, Pameijer CR, Robertson. GP. Nanoliposomal delivery of cytosolic phospholipase A 2 inhibitor arachidonyl trimethyl ketone for melanoma treatment. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2018; 14:863-873. [PMID: 29317343 PMCID: PMC5899023 DOI: 10.1016/j.nano.2017.12.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/15/2017] [Accepted: 12/27/2017] [Indexed: 12/19/2022]
Abstract
Drug resistance and toxicity are major limitations of cancer treatment and frequently occurs during melanoma therapy. Nanotechnology can decrease drug resistance by improving drug delivery, with limited toxicity. This study details the development of nanoparticles containing arachidonyl trifluoromethyl ketone (ATK), a cytosolic phospholipase A2 inhibitor, which can inhibit multiple key pathways responsible for the development of recurrent resistant disease. Free ATK is toxic, limiting its efficacy as a therapeutic agent. Hence, a novel nanoliposomal delivery system called NanoATK was developed, which loads 61.7% of the compound and was stable at 4oC for 12 weeks. The formulation decreased toxicity-enabling administration of higher doses, which was more effective at inhibiting melanoma cell growth compared to free-ATK. Mechanistically, NanoATK decreased cellular proliferation and triggered apoptosis to inhibit melanoma xenograft tumor growth without affecting animal weight. Functionally, it inhibited the cPLA2, AKT, and STAT3 pathways. Our results suggest the successful preclinical development of a unique nanoliposomal formulation containing ATK for the treatment of melanoma.
Collapse
Affiliation(s)
- Raghavendra Gowda
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033,The Penn State Melanoma and Skin Cancer Center The Pennsylvania State University College of Medicine, Hershey, PA 17033,Penn State Melanoma Therapeutics Program The Pennsylvania State University College of Medicine, Hershey, PA 17033,Foreman Foundation for Melanoma Research The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Saketh S. Dinavahi
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Soumya Iyer
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Shubhadeep Banerjee
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Rogerio I. Neves
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Dermatology and The Pennsylvania State University College of Medicine, Hershey, PA 17033 The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Surgery The Pennsylvania State University College of Medicine, Hershey, PA 17033,The Penn State Melanoma and Skin Cancer Center The Pennsylvania State University College of Medicine, Hershey, PA 17033,Penn State Melanoma Therapeutics Program The Pennsylvania State University College of Medicine, Hershey, PA 17033,Foreman Foundation for Melanoma Research The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Colette R. Pameijer
- Department of Surgery The Pennsylvania State University College of Medicine, Hershey, PA 17033,The Penn State Melanoma and Skin Cancer Center The Pennsylvania State University College of Medicine, Hershey, PA 17033,Penn State Melanoma Therapeutics Program The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Gavin P. Robertson.
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Pathology The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Dermatology and The Pennsylvania State University College of Medicine, Hershey, PA 17033 The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Surgery The Pennsylvania State University College of Medicine, Hershey, PA 17033,The Penn State Melanoma and Skin Cancer Center The Pennsylvania State University College of Medicine, Hershey, PA 17033,Penn State Melanoma Therapeutics Program The Pennsylvania State University College of Medicine, Hershey, PA 17033,Foreman Foundation for Melanoma Research The Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
10
|
Wu H, Liu H, Zuo F, Zhang L. Adenoviruses-mediated RNA interference targeting cytosolic phospholipase A2α attenuates focal ischemic brain damage in mice. Mol Med Rep 2018; 17:5601-5610. [PMID: 29484397 PMCID: PMC5866000 DOI: 10.3892/mmr.2018.8610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 06/19/2017] [Indexed: 01/11/2023] Open
Abstract
Cerebral ischemia injury is a clinical, frequently occurring disease, which causes a heavy burden on society and families. It has been demonstrated that cytosolic phospholipase A2α (cPLA2α) is significant in neurological injury caused by ischemic brain injury, and inhibition of cPLA2α may reduce stroke injury. In the present study, the role of cPLA2α was investigated in a mouse model of middle cerebral artery occlusion and/or reperfusion (MCAO/R) using an effective cPLA2α inhibitor and adenoviruses-mediated RNA interference. The most effective recombinant adenovirus encoding cPLA2α small interfering RNA (pAd-siRNA-cPLA2α) was constructed and selected. MCAO/R surgery is used to construct the model of focal ischemic brain damage in mice. Adenoviruses-mediated RNA interference targeting cPLA2α was administered by stereotactic surgery 2 h before the MCAO/R. The expression/activity of cPLA2α and cPLA2α-derived injurious lipid mediators was assessed. pAd-siRNA-cPLA2α-treated animals (RNA interference; RNAi group) were compared with pAd-siRNA-control-treated animals (negative group) with regard to neurological deficit, motor function, pathological changes, apoptosis, and infarct volume. The RNAi group animals reduced the expression level of cPLA2α, as determined by western blotting and reverse transcription-quantitative polymerase chain reaction, the improvement of locomotor function was evaluated by rotarod test, and the decrease of apoptosis was evaluated by terminal deoxynucleotidyl transferase dUTP nick end-labeling staining. The decreased infarct areas were evaluated by 2,3,5-triphenyltetrazolium chloride staining. The expression levels of prostaglandin E2, leukotrienes B4, lysophosphatidylcholine and free fatty acids were reduced in the RNAi group when compared with the negative control group. Thus, the data indicates that the expression level of cPLA2α was effectively controlled by pAd-siRNA-cPLA2α treatment. pAd-siRNA-cPLA2α treatment, in reducing the levels of inflammatory factors, neurological deficit and tissue damage, represents an effective potential therapeutic strategy. pAd-siRNA-cPLA2α reduces cPLA2α expression levels with long-term efficacy, thereby improving functional deficits and effectively attenuating ischemic brain damage. Thus, pAd-siRNA-cPLA2α shows potential value for therapeutic evaluation in ischemic brain damage.
Collapse
Affiliation(s)
- Huijun Wu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Hui Liu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Fengtong Zuo
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Lihua Zhang
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
11
|
Hardman CS, Chen YL, Salimi M, Jarrett R, Johnson D, Järvinen VJ, Owens RJ, Repapi E, Cousins DJ, Barlow JL, McKenzie ANJ, Ogg G. CD1a presentation of endogenous antigens by group 2 innate lymphoid cells. Sci Immunol 2017; 2:eaan5918. [PMID: 29273672 PMCID: PMC5826589 DOI: 10.1126/sciimmunol.aan5918] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/07/2017] [Indexed: 01/02/2023]
Abstract
Group 2 innate lymphoid cells (ILC2) are effectors of barrier immunity, with roles in infection, wound healing, and allergy. A proportion of ILC2 express MHCII (major histocompatibility complex II) and are capable of presenting peptide antigens to T cells and amplifying the subsequent adaptive immune response. Recent studies have highlighted the importance of CD1a-reactive T cells in allergy and infection, activated by the presentation of endogenous neolipid antigens and bacterial components. Using a human skin challenge model, we unexpectedly show that human skin-derived ILC2 can express CD1a and are capable of presenting endogenous antigens to T cells. CD1a expression is up-regulated by TSLP (thymic stromal lymphopoietin) at levels observed in the skin of patients with atopic dermatitis, and the response is dependent on PLA2G4A. Furthermore, this pathway is used to sense Staphylococcus aureus by promoting Toll-like receptor-dependent CD1a-reactive T cell responses to endogenous ligands. These findings define a previously unrecognized role for ILC2 in lipid surveillance and identify shared pathways of CD1a- and PLA2G4A-dependent ILC2 inflammation amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Clare S Hardman
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Yi-Ling Chen
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Maryam Salimi
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Rachael Jarrett
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - David Johnson
- Department of Plastic and Reconstructive Surgery, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Valtteri J Järvinen
- Oxford Protein Production Facility-UK, Harwell and Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Raymond J Owens
- Oxford Protein Production Facility-UK, Harwell and Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Emmanouela Repapi
- Computational Biology Research Group, Weatherall Institute of Molecular Medicine, Oxford, UK
| | - David J Cousins
- Department of Infection, Immunity and Inflammation, NIHR Leicester Respiratory Biomedical Research Unit, University of Leicester, Leicester, UK
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | | | | | - Graham Ogg
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
12
|
杨 泳, 郭 欣, 俞 志, 马 加, 刘 星, 李 丽, 肖 高, 刘 晓, 李 鑫, 沈 劲, 李 艳, 刘 睿. [Relationship between cytoplasmic phospholipase A2 and nuclear factor κB in one lung ventilation-induced lung injury in rabbits]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1345-1350. [PMID: 29070464 PMCID: PMC6743963 DOI: 10.3969/j.issn.1673-4254.2017.10.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To elucidate the mechanisms of up regulated expression of cytoplasmic phospholipase A2 (CPLA2) induced by one lung ventilation (OLV) by investigating the interactions between nuclear factor kappaB (NF-κB) and C-PLA2. METHODS Forty-eight healthy Japanese white rabbits were randomized into control group, solvent treatment group (group S), NF-κB inhibitor (PDTC)/solvent treatment group ( group PS), C-PLA2 inhibitor (AACOCF3)/solvent treatment group (group AS), OLV group (group O), solvent treatment plus OLV group (SO group), NFκB inhibitor (PDTC)/solvent treatment plus OLV group (group PSO) and CPLA2 inhibitor (AACOCF3)/solvent treatment plus OLV group (group ASO). ELISA was used to detect arachidonic acid (AA) content in the lung tissues, and NFκB and CPLA2 expressions were detected by Western blotting and quantitative PCR. Lung injuries were assessed based on the lung histological score, and the polymorphonuclear leukocyte count in the bronchial alveolar lavage fluid, myeloperoxidase (MPO) content in the lung tissues, and lung wet/dry weight (W/D) raito were determined. RESULTS Treatment of the rabbits with the solvent did not produce any adverse effects. OLV caused obvious lung injury in the rabbits and up regulated the expressions of CPLA2 and NFκB in the lung tissues (P<0.05). In rabbits without OLV, treatment with AACOCF3 or PDTC significantly down regulated both CPLA2 and NFκB expressions without affecting the other parameters. In rabbits with OLV, treatment with AACOCF3 or PDTC obviously lowered CPLA2 and NFκB expressions and lessened the OLV-induced lung injuries. CONCLUSION Both C-PLA2 and NF-κB play important roles and show interactions in OLV-induced lung injury in rabbits.
Collapse
Affiliation(s)
- 泳 杨
- 昆明医科大学医学机能实验中心, 云南 昆明 650500Experimental Center of Medical Function, Kunming Medical University, Kunming 650500, China
| | - 欣 郭
- 云南省第一人民医院麻醉科, 云南 昆明 650032Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming 650032, China
| | - 志成 俞
- 昆明医科大学医学机能实验中心, 云南 昆明 650500Experimental Center of Medical Function, Kunming Medical University, Kunming 650500, China
| | - 加庆 马
- 昆明医科大学医学机能实验中心, 云南 昆明 650500Experimental Center of Medical Function, Kunming Medical University, Kunming 650500, China
| | - 星玲 刘
- 云南省第一人民医院麻醉科, 云南 昆明 650032Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming 650032, China
| | - 丽莎 李
- 云南省第一人民医院麻醉科, 云南 昆明 650032Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming 650032, China
| | - 高鹏 肖
- 云南省第一人民医院麻醉科, 云南 昆明 650032Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming 650032, China
| | - 晓萌 刘
- 昆明医科大学医学机能实验中心, 云南 昆明 650500Experimental Center of Medical Function, Kunming Medical University, Kunming 650500, China
| | - 鑫楠 李
- 云南省第一人民医院麻醉科, 云南 昆明 650032Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming 650032, China
| | - 劲松 沈
- 云南省第一人民医院麻醉科, 云南 昆明 650032Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming 650032, China
| | - 艳华 李
- 云南省第一人民医院麻醉科, 云南 昆明 650032Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming 650032, China
| | - 睿 刘
- 云南省第一人民医院麻醉科, 云南 昆明 650032Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming 650032, China
| |
Collapse
|
13
|
Cytosolic Phospholipase A 2α Promotes Pulmonary Inflammation and Systemic Disease during Streptococcus pneumoniae Infection. Infect Immun 2017; 85:IAI.00280-17. [PMID: 28808157 DOI: 10.1128/iai.00280-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023] Open
Abstract
Pulmonary infection by Streptococcus pneumoniae is characterized by a robust alveolar infiltration of neutrophils (polymorphonuclear cells [PMNs]) that can promote systemic spread of the infection if not resolved. We previously showed that 12-lipoxygenase (12-LOX), which is required to generate the PMN chemoattractant hepoxilin A3 (HXA3) from arachidonic acid (AA), promotes acute pulmonary inflammation and systemic infection after lung challenge with S. pneumoniae As phospholipase A2 (PLA2) promotes the release of AA, we investigated the role of PLA2 in local and systemic disease during S. pneumoniae infection. The group IVA cytosolic isoform of PLA2 (cPLA2α) was activated upon S. pneumoniae infection of cultured lung epithelial cells and was critical for AA release from membrane phospholipids. Pharmacological inhibition of this enzyme blocked S. pneumoniae-induced PMN transepithelial migration in vitro Genetic ablation of the cPLA2 isoform cPLA2α dramatically reduced lung inflammation in mice upon high-dose pulmonary challenge with S. pneumoniae The cPLA2α-deficient mice also suffered no bacteremia and survived a pulmonary challenge that was lethal to wild-type mice. Our data suggest that cPLA2α plays a crucial role in eliciting pulmonary inflammation during pneumococcal infection and is required for lethal systemic infection following S. pneumoniae lung challenge.
Collapse
|
14
|
Wan M, Tang X, Stsiapanava A, Haeggström JZ. Biosynthesis of leukotriene B 4. Semin Immunol 2017; 33:3-15. [DOI: 10.1016/j.smim.2017.07.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 05/29/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
|
15
|
Menden HL, Xia S, Mabry SM, Navarro A, Nyp MF, Sampath V. Nicotinamide Adenine Dinucleotide Phosphate Oxidase 2 Regulates LPS-Induced Inflammation and Alveolar Remodeling in the Developing Lung. Am J Respir Cell Mol Biol 2017; 55:767-778. [PMID: 27438994 DOI: 10.1165/rcmb.2016-0006oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In premature infants, sepsis is associated with alveolar simplification manifesting as bronchopulmonary dysplasia. The redox-dependent mechanisms underlying sepsis-induced inflammation and alveolar remodeling in the immature lung remain unclear. We developed a neonatal mouse model of sepsis-induced lung injury to investigate whether nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) regulates Toll-like receptor (TLR)-mediated inflammation and alveolar remodeling. Six-day-old NOX2+/+ and NOX2-/- mice were injected with intraperitoneal LPS to induce sepsis. Lung inflammation and canonical TLR signaling were assessed 24 hours after LPS. Alveolar development was examined in 15-day-old mice after LPS on Day 6. The in vivo efficacy of a NOX2 inhibitor (NOX2-I) on NOX2 complex assembly and sepsis-induced lung inflammation were examined. Lung cytokine expression and neutrophil influx induced with sepsis in NOX2+/+ mice was decreased by >50% in NOX2-/- mice. LPS-induced TLR4 signaling evident by inhibitor of NF-κB kinase-β and mitogen-activated protein kinase phosphorylation, and nuclear factor-κB/AP-1 translocation were attenuated in NOX2-/- mice. LPS increased matrix metalloproteinase 9 while decreasing elastin and keratinocyte growth factor levels in NOX2+/+ mice. An LPS-induced increase in matrix metalloproteinase 9 and decrease in fibroblast growth factor 7 and elastin were not evident in NOX2-/- mice. An LPS-induced reduction in radial alveolar counts and increased mean linear intercepts were attenuated in NOX2-/- mice. LPS-induced NOX2 assembly evident by p67phox/gp91phox coimmunoprecipitation was disrupted with NOX2-I. NOX2-I also mitigated LPS-induced cytokine expression, TLR pathway signaling, and alveolar simplification. In a mouse model of neonatal sepsis, NOX2 regulates proinflammatory TLR signaling and alveolar remodeling induced by a single dose of LPS. Our results provide mechanistic insight into the regulation of sepsis-induced alveolar remodeling in the developing lung.
Collapse
Affiliation(s)
- Heather L Menden
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Sheng Xia
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Sherry M Mabry
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Angels Navarro
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Michael F Nyp
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Venkatesh Sampath
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| |
Collapse
|
16
|
Paoletti L, Domizi P, Marcucci H, Montaner A, Krapf D, Salvador G, Banchio C. Lysophosphatidylcholine Drives Neuroblast Cell Fate. Mol Neurobiol 2015; 53:6316-6331. [DOI: 10.1007/s12035-015-9528-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/05/2015] [Indexed: 12/31/2022]
|
17
|
Khan M, Shunmugavel A, Dhammu TS, Matsuda F, Singh AK, Singh I. Oral administration of cytosolic PLA2 inhibitor arachidonyl trifluoromethyl ketone ameliorates cauda equina compression injury in rats. J Neuroinflammation 2015; 12:94. [PMID: 25971887 PMCID: PMC4436116 DOI: 10.1186/s12974-015-0311-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/28/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Phospholipase A2 (PLA2)-derived proinflammatory lipid mediators such as prostaglandin E2 (PGE2), leukotrienes B4 (LTB4), lysophosphatidylcholine (LPC), and free fatty acids (FFA) are implicated in spinal cord injury (SCI) pathologies. Reducing the levels of these injurious bioactive lipid mediators is reported to ameliorate SCI. However, the specific role of the group IVA isoform of PLA2 cytosolic PLA2 (cPLA2) in lumbar spinal canal stenosis (LSS) due to cauda equina compression (CEC) injury is not clear. In this study, we investigated the role of cPLA2 in a rat model of CEC using a non-toxic cPLA2-preferential inhibitor, arachidonyl trifluoromethyl ketone (ATK). METHODS LSS was induced in adult female rats by CEC procedure using silicone blocks within the epidural spaces of L4 to L6 vertebrae. cPLA2 inhibitor ATK (7.5 mg/kg) was administered by oral gavage at 2 h following the CEC. cPLA2-derived injurious lipid mediators and the expression/activity of cPLA2, 5-lipoxygenase (5-LOX), and cyclooxygenase-2 (COX-2) were assessed. ATK-treated (CEC + ATK) were compared with vehicle-treated (CEC + VEH) animals in terms of myelin levels, pain threshold, and motor function. RESULTS ATK treatment of CEC animals reduced the phosphorylation of cPLA2 (pcPLA2) determined by Western blot, improved locomotor function evaluated by rotarod task, and reduced pain threshold evaluated by mechanical hyperalgesia method. Levels of FFA and LPC, along with PGE2 and LTB4, were reduced in CEC + ATK compared with CEC + VEH group. However, ATK treatment reduced neither the activity/expression of 5-LOX nor the expression of COX-2 in CEC + VEH animals. Increased cPLA2 activity in the spinal cord from CEC + VEH animals correlated well with decreased spinal cord as well as cauda equina fiber myelin levels, which were restored after ATK treatment. CONCLUSION The data indicate that cPLA2 activity plays a significant role in tissue injury and pain after LSS. Reducing the levels of proinflammatory and tissue damaging eicosanoids and the deleterious lipid mediator LPC shows therapeutic potential. ATK inhibits cPLA2 activity, thereby decreasing the levels of injurious lipid mediators, reducing pain, improving functional deficits, and conferring protection against LSS injury. Thus, it shows potential for preclinical evaluation in LSS.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | | | - Tajinder S Dhammu
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Fumiyo Matsuda
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA. .,School of Health Science, Kagoshima University, Kagoshima, Japan.
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA. .,Ralph H. Johnson VA Medical Center, Charleston, SC, USA.
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
18
|
Liu L, He H, Liu A, Xu J, Han J, Chen Q, Hu S, Xu X, Huang Y, Guo F, Yang Y, Qiu H. Therapeutic Effects of Bone Marrow-Derived Mesenchymal Stem Cells in Models of Pulmonary and Extrapulmonary Acute Lung Injury. Cell Transplant 2015; 24:2629-42. [PMID: 25695285 DOI: 10.3727/096368915x687499] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) offer a promising therapy for acute lung injury (ALI). However, whether the same MSC treatments possess similar potential for different ALI models is not fully clear. The present study evaluated the distribution and therapeutic effects of intravenous MSC administration for the treatment of intratracheal lipopolysaccharide (LPS)-induced intrapulmonary ALI and intravenous LPS/zymosan-induced extrapulmonary ALI, matched with lung injury severity, at 30 min and 1, 3, and 7 days. We found that MSC transplantation attenuated lung injury and inhibited lung inflammation in both ALI models. The benefits of MSCs were more significant in the intrapulmonary ALI mice. In vivo and ex vivo fluorescence imaging showed that MSCs primarily homed into the lung. However, more MSCs were recruited into the lungs of the intrapulmonary ALI mice than those of the extrapulmonary ALI mice over the time course. A few MSCs were also detected in the liver and spleen at days 3 and 7. In addition, the two ALI models showed different extrapulmonary organ dysfunction. A lower percentage of cell apoptosis and SDF-1α levels was found in the liver and spleen of the intrapulmonary ALI mice than in those of the extrapulmonary ALI mice. These results suggested that the two ALI models were accompanied with different degrees of extrapulmonary organ damage, which resulted in differences in the trafficking and accumulation of MSCs to the injured lung and consequently accounted for different therapeutic effects of MSCs for lung repair in the two ALI models. These data suggest that intravenous administration of MSCs has a greater potential for the treatment of intrapulmonary ALI than extrapulmonary ALI matched with lung injury severity; these differences were due to more recruitment of MSCs in the lungs of intrapulmonary ALI mice than those of extrapulmonary ALI mice. This finding may contribute to the clinical use of MSCs for the treatment of ALI.
Collapse
Affiliation(s)
- Ling Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Aggarwal S, Gross CM, Kumar S, Dimitropoulou C, Sharma S, Gorshkov BA, Sridhar S, Lu Q, Bogatcheva NV, Jezierska-Drutel AJ, Lucas R, Verin AD, Catravas JD, Black SM. Dimethylarginine dimethylaminohydrolase II overexpression attenuates LPS-mediated lung leak in acute lung injury. Am J Respir Cell Mol Biol 2014; 50:614-25. [PMID: 24134589 DOI: 10.1165/rcmb.2013-0193oc] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Acute lung injury (ALI) is a severe hypoxemic respiratory insufficiency associated with lung leak, diffuse alveolar damage, inflammation, and loss of lung function. Decreased dimethylaminohydrolase (DDAH) activity and increases in asymmetric dimethylarginine (ADMA), together with exaggerated oxidative/nitrative stress, contributes to the development of ALI in mice exposed to LPS. Whether restoring DDAH function and suppressing ADMA levels can effectively ameliorate vascular hyperpermeability and lung injury in ALI is unknown, and was the focus of this study. In human lung microvascular endothelial cells, DDAH II overexpression prevented the LPS-dependent increase in ADMA, superoxide, peroxynitrite, and protein nitration. DDAH II also attenuated the endothelial barrier disruption associated with LPS exposure. Similarly, in vivo, we demonstrated that the targeted overexpression of DDAH II in the pulmonary vasculature significantly inhibited the accumulation of ADMA and the subsequent increase in oxidative/nitrative stress in the lungs of mice exposed to LPS. In addition, augmenting pulmonary DDAH II activity before LPS exposure reduced lung vascular leak and lung injury and restored lung function when DDAH activity was increased after injury. Together, these data suggest that enhancing DDAH II activity may prove a useful adjuvant therapy to treat patients with ALI.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta, Georgia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Chen N, Sun G, Yuan X, Hou J, Wu Q, Soromou LW, Feng H. Inhibition of lung inflammatory responses by bornyl acetate is correlated with regulation of myeloperoxidase activity. J Surg Res 2014; 186:436-45. [DOI: 10.1016/j.jss.2013.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 08/16/2013] [Accepted: 09/05/2013] [Indexed: 11/26/2022]
|
21
|
Parker JC. Acute lung injury and pulmonary vascular permeability: use of transgenic models. Compr Physiol 2013; 1:835-82. [PMID: 23737205 DOI: 10.1002/cphy.c100013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acute lung injury is a general term that describes injurious conditions that can range from mild interstitial edema to massive inflammatory tissue destruction. This review will cover theoretical considerations and quantitative and semi-quantitative methods for assessing edema formation and increased vascular permeability during lung injury. Pulmonary edema can be quantitated directly using gravimetric methods, or indirectly by descriptive microscopy, quantitative morphometric microscopy, altered lung mechanics, high-resolution computed tomography, magnetic resonance imaging, positron emission tomography, or x-ray films. Lung vascular permeability to fluid can be evaluated by measuring the filtration coefficient (Kf) and permeability to solutes evaluated from their blood to lung clearances. Albumin clearances can then be used to calculate specific permeability-surface area products (PS) and reflection coefficients (σ). These methods as applied to a wide variety of transgenic mice subjected to acute lung injury by hyperoxic exposure, sepsis, ischemia-reperfusion, acid aspiration, oleic acid infusion, repeated lung lavage, and bleomycin are reviewed. These commonly used animal models simulate features of the acute respiratory distress syndrome, and the preparation of genetically modified mice and their use for defining specific pathways in these disease models are outlined. Although the initiating events differ widely, many of the subsequent inflammatory processes causing lung injury and increased vascular permeability are surprisingly similar for many etiologies.
Collapse
Affiliation(s)
- James C Parker
- Department of Physiology, University of South Alabama, Mobile, Alabama, USA.
| |
Collapse
|
22
|
Choi JM, Choi YH, Kim SK, Ahn KH, Won JH, Lim JH, Jang YJ, Lee S, Kim DH, Kim DK. (S)-tetrahydroisoquinoline alkaloid inhibits LPS-induced arachidonic acid release through downregulation of cPLA2 expression. Mol Cells 2013; 36:400-9. [PMID: 24293010 PMCID: PMC3887938 DOI: 10.1007/s10059-013-0078-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 09/10/2013] [Accepted: 09/15/2013] [Indexed: 01/24/2023] Open
Abstract
Sepsis, a systemic inflammatory response syndrome, remains a potentially lethal condition. (S)-1-α-Naphthylmethyl-6,7-dihydroxy-1,2,3,4-tetrahydroisoquinoline (CKD712) is noted as a drug candidate for sepsis. Many studies have demonstrated its significant anti-inflammatory effects. Here we first examined whether CKD712 inhibits lipopolysaccharide (LPS)-induced arachidonic acid (AA) release in the RAW 264.7 mouse monocyte cell line, and subsequently, its inhibitory mechanisms. CKD712 reversed LPS-associated morphological changes in the RAW 264.7 cells, and inhibited LPS-induced release of AA in a concentrationdependent manner. The inhibition was apparently due to the diminished expression of a cytosolic form of phospholipase A2 (cPLA2) by CKD712, resulting from reduced NF-κB activation. Furthermore, CKD712 inhibited the activation of ERK1/2 and SAP/JNK, but not of p38 MAPK. CKD712 had no effect on the activity or phosphorylation of cPLA2 and on calcium influx. Our results collectively suggest that CKD712 inhibits LPS-induced AA release through the inhibition of a MAPKs/NF-κB pathway leading to reduced cPLA2 expression in RAW 264.7 cells.
Collapse
Affiliation(s)
- Jong Min Choi
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Young Hwa Choi
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Seok Kyun Kim
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Kyong Hoon Ahn
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Jong Hoon Won
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Joo Hyuk Lim
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - You Jin Jang
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | | | | | - Dae Kyong Kim
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| |
Collapse
|
23
|
Di Gennaro A, Haeggström JZ. Targeting leukotriene B4 in inflammation. Expert Opin Ther Targets 2013; 18:79-93. [PMID: 24090264 DOI: 10.1517/14728222.2013.843671] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Leukotriene (LT) B(4) is a powerful proinflammatory lipid mediator and triggers adherence to the endothelium, activates and recruits leukocytes to the site of injury. When formed in excess, LTB(4) plays a pathogenic role and may sustain chronic inflammation in diseases such as asthma, rheumatoid arthritis, and inflammatory bowel disease. Recent investigations have also indicated that LTB(4) is involved in cardiovascular diseases. AREAS COVERED As the 5-lipoxygenase pathway involves several discrete, tightly coupled, enzymes, which convert the substrate, 'step by step', into bioactive products, several different strategies have been used to target LTB(4) as a means to treat inflammation. Here, we discuss recent findings regarding the development of selective enzyme inhibitors and antagonists for LTB(4) receptors, as well as their application in preclinical and clinical studies. EXPERT OPINION Components of the 5-lipoxygenase pathway have received considerable attention as candidate drug targets resulting in one new class of medications against asthma, that is, the antileukotrienes. However, efforts to specifically target LTB(4) have not yet been fruitful in the clinical setting, in spite of very promising preclinical data. Recently, crystal structures along with hitherto unknown functions of key enzymes in the leukotriene cascade have emerged, offering new opportunities for drug development and, with time, pharmacological intervention in LTB(4)-mediated pathologies.
Collapse
Affiliation(s)
- Antonio Di Gennaro
- Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Chemistry 2 , Scheeles väg 2, Stockholm, S-171 77 , Sweden
| | | |
Collapse
|
24
|
Huwiler A, Feuerherm AJ, Sakem B, Pastukhov O, Filipenko I, Nguyen T, Johansen B. The ω3-polyunsaturated fatty acid derivatives AVX001 and AVX002 directly inhibit cytosolic phospholipase A(2) and suppress PGE(2) formation in mesangial cells. Br J Pharmacol 2013; 167:1691-701. [PMID: 22831644 DOI: 10.1111/j.1476-5381.2012.02114.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 06/27/2012] [Accepted: 07/07/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE ω3-polyunsaturated fatty acids (ω3-PUFAs) are known to exert anti-inflammatory effects in various disease models although their direct targets are only poorly characterized. EXPERIMENTAL APPROACH Here we report on two new cPLA(2) inhibitors, the ω3-derivatives AVX001 and AVX002, and their effects on inflammatory PGE(2) production in cultures of renal mesangial cells. KEY RESULTS AVX001 and AVX002 dose-dependently inhibited the group IVA cytosolic phospholipase A(2) (cPLA(2) ) in an in vitro activity assay with similar IC(50) values for AVX001 and AVX002, whereas the known cPLA(2) inhibitor AACOCF(3) was less potent and docosahexaenoic acid (DHA) was inactive. In renal mesangial cells, AVX001 and AVX002 suppressed IL-1β-induced PGE(2) synthesis. Mechanistically, this effect occurred by a down-regulation of IL-1β-induced group IIA-sPLA(2) protein expression, mRNA expression and promoter activity. A similar but less potent effect was seen with AACOCF(3) and no effect was seen with DHA. As gene expression of sPLA(2) is known to be regulated by the transcription factor NF-κB, we further investigated NF-κB activation. Both compounds prevented NF-κB activation by blocking degradation of the inhibitor of κB. CONCLUSIONS AND IMPLICATIONS These data show for the first time that the novel cPLA(2) inhibitors AVX001 and AVX002 exert an anti-inflammatory effect in cultures of renal mesangial cells and reduce the pro-inflammatory mediator PGE(2) through an inhibitory effect on NF-κB activation. Therefore, these compounds may represent promising novel drugs for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Andrea Huwiler
- Institut für Pharmakologie, Universität Bern, Bern, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
25
|
Liu MS, Liu CH, Wu G, Zhou Y. Antisense inhibition of secretory and cytosolic phospholipase A2 reduces the mortality in rats with sepsis*. Crit Care Med 2012; 40:2132-40. [PMID: 22564957 DOI: 10.1097/ccm.0b013e31824e1e20] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Phospholipase A(2) has been implicated to play a pivotal role in the pathogenesis of sepsis syndrome. The two major forms of phospholipase A(2) isoenzymes, secretory phospholipase A(2) and cytosolic phospholipase A(2), are overexpressed during sepsis. The objective of this study was to test the hypothesis that inhibition of the overexpressed secretory phospholipase A(2) and cytosolic phospholipase A(2) during sepsis benefits the disease's eventual outcome. DESIGN Short-chain antisense oligonucleotide molecules were designed with the aid of computer software programs, and their in vitro efficacies were assessed in cell culture systems based on inhibition of target protein expression. The in vivo efficacies were determined in intact sepsis rats using 35-day survival rate as a primary efficacy end point. SETTING Animal research laboratory at a university. SUBJECTS Male Sprague-Dawley rats (180-200 g). INTERVENTIONS Sepsis was induced by cecal ligation and puncture. Antibiotics were administered subcutaneously once daily at 12 mg/kg, for 20 days. Oligonucleotides (antisense or mismatch) were administered intravenously once daily at 2 mg/kg to 0.8 mg/kg in a decreasing order, for 20 days. MEASUREMENTS AND MAIN RESULTS In cell culture systems, 21 of the 105 antisense constructs were found to be efficacious in inhibiting secretory phospholipase A(2) IIa and cytosolic phospholipase A(2) IVa protein expression. In sepsis rats, antisense oligonucleotides were capable of reducing their target protein expression by 18%-61% in major organs such as liver, heart, and kidney. In animal experiments, sepsis without any treatment (Group 1) had a median survival time of 2 days and a zero (0) percent survival rate at day 14. Sepsis with antibiotic treatment (Group 2) had a median survival time of 6 days and a 35-day survival rate of 28%. Sepsis with cotreatment of antibiotics and antisense oligonucleotides (one against secretory phospholipase A2 IIa and the other against cytosolic phospholipase A(2) IVa) (Group 4) increased the median survival time from 6 to 35 days and the 35-day survival rate from 28% to 58.8% as compared with antibiotics alone (Group 4 vs. Group 2; p <.05). Sepsis with cotreatment of antibiotics and mismatch oligonucleotides (Group 3) did not affect the median survival time and the 35-day survival rate as compared to antibiotics alone (Group 3 vs. Group 2; p >.05). CONCLUSIONS The results demonstrate that antisense strategy against secretory phospholipase A(2) IIa and cytosolic phospholipase A(2) IVa can inhibit their target protein expression in major organs and greatly improve the clinical outcome, i.e., an absolute reduction in 35-day mortality of 30.8%, in rats with sepsis. Our studies, thus, provide an improved method for the treatment of sepsis by targeting multiple forms of phospholipase A(2) isoenzymes with DNA antisense oligomers.
Collapse
Affiliation(s)
- Maw-Shung Liu
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St Louis, MO, USA.
| | | | | | | |
Collapse
|
26
|
Muñoz NM, Desai A, Meliton LN, Meliton AY, Zhou T, Leff AR, Dudek SM. Group V phospholipase A(2) increases pulmonary endothelial permeability through direct hydrolysis of the cell membrane. Pulm Circ 2012; 2:182-92. [PMID: 22837859 PMCID: PMC3401872 DOI: 10.4103/2045-8932.97604] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Acute lung injury (ALI) is characterized by inflammatory disruption of the alveolar–vascular barrier, resulting in severe respiratory compromise. Inhibition of the intercellular messenger protein, Group V phospholipase A2 (gVPLA2), blocks vascular permeability caused by LPS both in vivo and in vitro. In this investigation we studied the mechanism by which recombinant gVPLA2 increases permeability of cultured human pulmonary endothelial cells (EC). Exogenous gVPLA2 (500 nM), a highly hydrolytic enzyme, caused a significant increase in EC permeability that began within minutes and persisted for >10 hours. However, the major hydrolysis products of gVPLA2 (Lyso-PC, Lyso-PG, LPA, arachidonic acid) did not cause EC structural rearrangement or loss of barrier function at concentrations <10 μM. Higher concentrations (≥ 30 μM) of these membrane hydrolysis products caused some increased permeability but were associated with EC toxicity (measured by propidium iodide incorporation) that did not occur with barrier disruption by gVPLA2 (500 nM). Pharmacologic inhibition of multiple intracellular signaling pathways induced by gVPLA2 activity (ERK, p38, PI3K, cytosolic gIVPLA2) also did not prevent EC barrier disruption by gVPLA2. Finally, pretreatment with heparinase to prevent internalization of gVPLA2 did not inhibit EC barrier disruption by gVPLA2. Our data thus indicate that gVPLA2 increases pulmonary EC permeability directly through action as a membrane hydrolytic agent. Disruption of EC barrier function does not depend upon membrane hydrolysis products, gVPLA2 internalization, or upregulation of downstream intracellular signaling.
Collapse
Affiliation(s)
- Nilda M Muñoz
- Philippine Foundation for Lung Health, Research and Development, Inc. and Research and Biotechnology Division, St. Luke's Medical Center, Quezon City, Philippines
| | | | | | | | | | | | | |
Collapse
|
27
|
Changes in group II phospholipase A2 gene expression in rat heart during sepsis. J Surg Res 2012; 181:272-8. [PMID: 22835950 DOI: 10.1016/j.jss.2012.06.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/15/2012] [Accepted: 06/21/2012] [Indexed: 11/22/2022]
Abstract
BACKGROUND This study was undertaken to investigate alterations of group II phospholipase A2 (PLA2) gene expression and its underlying mechanism in rat heart during different phases of sepsis. MATERIALS AND METHODS Sepsis was induced by cecal ligation and puncture (CLP). Experiments were divided into three groups, control, early sepsis, and late sepsis. Early and late sepsis refers to those animals sacrificed at 9 and 18 h, respectively, after CLP. PLA2 enzyme activity, group II PLA2 protein level, messenger RNA (mRNA) abundance, transcription rate, and half-life were measured. RESULTS PLA2 activity was decreased by 29% during early sepsis but it was increased by 49% during late sepsis. Group II PLA2 protein level was decreased by 27% during early sepsis but it was increased by 35.3% during late sepsis. Group II PLA2 mRNA was decreased by 21% during early sepsis but it was increased by 141% during late sepsis. The transcription rate of group II PLA2 mRNA was reduced by 25% during early sepsis but it was elevated by 67% during late sepsis. The half-life of group II PLA2 mRNA remained unaltered during early and late phases of sepsis. CONCLUSIONS These results demonstrate that PLA2 activity, group II PLA2 protein level, the mRNA abundance, and transcription rate were concurrently underexpressed during early sepsis, while they were overexpressed during late sepsis, with no change in the degradation of gene transcript. These data indicate that the biphasic changes in group II PLA2 gene expression are regulated transcriptionally during sepsis.
Collapse
|
28
|
Aggarwal S, Dimitropoulou C, Lu Q, Black SM, Sharma S. Glutathione supplementation attenuates lipopolysaccharide-induced mitochondrial dysfunction and apoptosis in a mouse model of acute lung injury. Front Physiol 2012; 3:161. [PMID: 22654772 PMCID: PMC3361071 DOI: 10.3389/fphys.2012.00161] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/07/2012] [Indexed: 11/29/2022] Open
Abstract
Acute lung injury (ALI) is a life threatening condition associated with hypoxemia, diffuse alveolar damage, inflammation, and loss of lung function. Lipopolysaccharide (LPS; endotoxin) from the outer membrane of Gram-negative bacteria is a major virulence factor involved in the development of ALI. The depletion of glutathione (GSH), an essential intra- and extra-cellular protective antioxidant, by LPS is an important event that contributes to the elevation in reactive oxygen species. Whether restoring GSH homeostasis can effectively ameliorate mitochondrial dysfunction and cellular apoptosis in ALI is unknown and therefore, was the focus of this study. In peripheral lung tissue of LPS-treated mice, hydrogen peroxide and protein nitration levels were significantly increased. Pre-treatment with GSH-ethyl ester (GSH-EE) prevented this increase in oxidative stress. LPS also increased the lactate/pyruvate ratio, attenuated SOD2 protein levels, and decreased ATP levels in the mouse lung indicative of mitochondrial dysfunction. Again, GSH-EE treatment preserved the mitochondrial function. Finally, our studies showed that LPS induced an increase in the mitochondrial translocation of Bax, caspase 3 activation, and nuclear DNA fragmentation and these parameters were all prevented with GSH-EE. Thus, this study suggests that GSH-EE supplementation may reduce the mitochondrial dysfunction associated with ALI.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Vascular Biology Center, Georgia Health Sciences University Augusta, GA, USA
| | | | | | | | | |
Collapse
|
29
|
Noha SM, Jazzar B, Kuehnl S, Rollinger JM, Stuppner H, Schaible AM, Werz O, Wolber G, Schuster D. Pharmacophore-based discovery of a novel cytosolic phospholipase A(2)α inhibitor. Bioorg Med Chem Lett 2012; 22:1202-7. [PMID: 22192589 PMCID: PMC3268354 DOI: 10.1016/j.bmcl.2011.11.093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 11/18/2011] [Accepted: 11/19/2011] [Indexed: 01/26/2023]
Abstract
The release of arachidonic acid, a precursor in the production of prostaglandins and leukotrienes, is achieved by activity of the cytosolic phospholipase A(2)α (cPLA(2)α). Signaling mediated by this class of bioactive lipids, which are collectively referred to as eicosanoids, has numerous effects in physiological and pathological processes. Herein, we report the development of a ligand-based pharmacophore model and pharmacophore-based virtual screening of the National Cancer Institute (NCI) database, leading to the identification of 4-(hexadecyloxy)-3-(2-(hydroxyimino)-3-oxobutanamido)benzoic acid (NSC 119957) as cPLA(2)α inhibitor in cell-free and cell-based in vitro assays.
Collapse
Affiliation(s)
- Stefan M. Noha
- Institute of Pharmacy/Pharmaceutical Chemistry, Computer-Aided Molecular Design (CAMD) Group and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, AT-6020 Innsbruck, Austria
| | - Bianca Jazzar
- Department of Pharmaceutical Analytics, Pharmaceutical Institute, University of Tuebingen, Auf der Morgenstelle 8, DE-72076 Tuebingen, Germany
| | - Susanne Kuehnl
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, AT-6020 Innsbruck, Austria
| | - Judith M. Rollinger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, AT-6020 Innsbruck, Austria
| | - Hermann Stuppner
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, AT-6020 Innsbruck, Austria
| | - Anja M. Schaible
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, DE-07743 Jena, Germany
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, DE-07743 Jena, Germany
| | - Gerhard Wolber
- Institute of Pharmacy/Pharmaceutical Chemistry, Königin-Luise-Str. 2+4, DE-14195 Berlin, Germany
| | - Daniela Schuster
- Institute of Pharmacy/Pharmaceutical Chemistry, Computer-Aided Molecular Design (CAMD) Group and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, AT-6020 Innsbruck, Austria
| |
Collapse
|
30
|
Dennis EA, Cao J, Hsu YH, Magrioti V, Kokotos G. Phospholipase A2 enzymes: physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem Rev 2011; 111:6130-85. [PMID: 21910409 PMCID: PMC3196595 DOI: 10.1021/cr200085w] [Citation(s) in RCA: 820] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Edward A. Dennis
- Department of Chemistry and Biochemistry and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093-0601
| | - Jian Cao
- Department of Chemistry and Biochemistry and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093-0601
| | - Yuan-Hao Hsu
- Department of Chemistry and Biochemistry and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093-0601
| | - Victoria Magrioti
- Laboratory of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - George Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Athens 15771, Greece
| |
Collapse
|
31
|
Murakami M, Taketomi Y, Miki Y, Sato H, Hirabayashi T, Yamamoto K. Recent progress in phospholipase A₂ research: from cells to animals to humans. Prog Lipid Res 2010; 50:152-92. [PMID: 21185866 DOI: 10.1016/j.plipres.2010.12.001] [Citation(s) in RCA: 368] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mammalian genomes encode genes for more than 30 phospholipase A₂s (PLA₂s) or related enzymes, which are subdivided into several classes including low-molecular-weight secreted PLA₂s (sPLA₂s), Ca²+-dependent cytosolic PLA₂s (cPLA₂s), Ca²+-independent PLA₂s (iPLA₂s), platelet-activating factor acetylhydrolases (PAF-AHs), lysosomal PLA₂s, and a recently identified adipose-specific PLA. Of these, the intracellular cPLA₂ and iPLA₂ families and the extracellular sPLA₂ family are recognized as the "big three". From a general viewpoint, cPLA₂α (the prototypic cPLA₂ plays a major role in the initiation of arachidonic acid metabolism, the iPLA₂ family contributes to membrane homeostasis and energy metabolism, and the sPLA₂ family affects various biological events by modulating the extracellular phospholipid milieus. The cPLA₂ family evolved along with eicosanoid receptors when vertebrates first appeared, whereas the diverse branching of the iPLA₂ and sPLA₂ families during earlier eukaryote development suggests that they play fundamental roles in life-related processes. During the past decade, data concerning the unexplored roles of various PLA₂ enzymes in pathophysiology have emerged on the basis of studies using knockout and transgenic mice, the use of specific inhibitors, and information obtained from analysis of human diseases caused by mutations in PLA₂ genes. This review focuses on current understanding of the emerging biological functions of PLA₂s and related enzymes.
Collapse
Affiliation(s)
- Makoto Murakami
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | | | | | | | | | | |
Collapse
|
32
|
Huang W, Bhavsar A, Ward RE, Hall JCE, Priestley JV, Michael-Titus AT. Arachidonyl trifluoromethyl ketone is neuroprotective after spinal cord injury. J Neurotrauma 2010; 26:1429-34. [PMID: 19371144 DOI: 10.1089/neu.2008.0835] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In spinal cord injury (SCI), neuronal and oligodendroglial loss occurs as a result of the initial trauma and the secondary damage that is triggered by excitotoxicity, free radicals, and inflammation. There is evidence that SCI ellicits increased cytosolic phospholipase A(2) (cPLA(2)) activity. The cleavage of phospholipids by cPLA(2) leads to release of fatty acids, and in particular arachidonic acid (AA), the metabolites of which have been associated with increased inflammation and oxidative stress. The aim of our study was to investigate whether the inhibition of cPLA(2) following SCI leads to tissue protection and an improved functional outcome. Adult rats received compression SCI and 30 min after injury they were treated intravenously with either saline or the cPLA(2) inhibitor arachidonyl trifluoromethyl ketone (AACOCF3) (7.13 mg/kg). The animals were sacrificed at 7 days post-injury and the lesioned tissue was labeled using markers for neurons, oligodendrocytes, and macrophages/activated microglia. We also assessed locomotor recovery using the Basso-Beattie-Bresnahan (BBB) score. The number of surviving neurons and oligodendrocytes was significantly increased in animals treated with the cPLA(2) inhibitor compared to saline controls. The behavioral analysis mirrored the neuroprotective effects and showed that the inhibitor-treated group had better locomotor recovery compared to saline controls. Our results show that AACOCF3 has neuroprotective potential, and support the idea that cPLA(2) is critically involved in acute spinal injury.
Collapse
Affiliation(s)
- Wenlong Huang
- Neuroscience Centre, Institute of Cell and Molecular Science, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
33
|
Wu YZ, Abolhassani M, Ollero M, Dif F, Uozumi N, Lagranderie M, Shimizu T, Chignard M, Touqui L. Cytosolic phospholipase A2alpha mediates Pseudomonas aeruginosa LPS-induced airway constriction of CFTR -/- mice. Respir Res 2010; 11:49. [PMID: 20429932 PMCID: PMC2873258 DOI: 10.1186/1465-9921-11-49] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 04/29/2010] [Indexed: 11/17/2022] Open
Abstract
Background Lungs of cystic fibrosis (CF) patients are chronically infected with Pseudomonas aeruginosa. Increased airway constriction has been reported in CF patients but underplaying mechanisms have not been elucidated. Aim: to examine the effect of P. aeruginosa LPS on airway constriction in CF mice and the implication in this process of cytosolic phospholipase A2α (cPLA2α), an enzyme involved in arachidonic acid (AA) release. Methods Mice were instilled intra-nasally with LPS. Airway constriction was assessed using barometric plethysmograph. MIP-2, prostaglandin E2 (PGE2), leukotrienes and AA concentrations were measured in BALF using standard kits and gas chromatography. Results LPS induced enhanced airway constriction and AA release in BALF of CF compared to littermate mice. This was accompanied by increased levels of PGE2, but not those of leukotrienes. However, airway neutrophil influx and MIP-2 production remained similar in both mouse strains. The cPLA2α inhibitor arachidonyl trifluoro-methyl-ketone (ATK), but not aspirin which inhibit PGE2 synthesis, reduced LPS-induced airway constriction. LPS induced lower airway constriction and PGE2 production in cPLA2α -/- mice compared to corresponding littermates. Neither aspirin nor ATK interfered with LPS-induced airway neutrophil influx or MIP-2 production. Conclusions CF mice develop enhanced airway constriction through a cPLA2α-dependent mechanism. Airway inflammation is dissociated from airway constriction in this model. cPLA2α may represent a suitable target for therapeutic intervention in CF. Attenuation of airway constriction by cPLA2α inhibitors may help to ameliorate the clinical status of CF patients.
Collapse
Affiliation(s)
- Yong-Zheng Wu
- Unité de Défense Innée et Inflammation, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Pergola C, Werz O. 5-Lipoxygenase inhibitors: a review of recent developments and patents. Expert Opin Ther Pat 2010; 20:355-75. [DOI: 10.1517/13543771003602012] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
35
|
Lv XJ, Li YY, Zhang YJ, Mao M, Qian GS. Over-expression of caveolin-1 aggravate LPS-induced inflammatory response in AT-1 cells via up-regulation of cPLA2/p38 MAPK. Inflamm Res 2010; 59:531-41. [PMID: 20099006 DOI: 10.1007/s00011-010-0157-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 12/08/2009] [Accepted: 01/07/2010] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE AND DESIGN The aim of this study was to study the effect of caveolin-1 on the cytosolic phospholipase A2 (cPLA2), p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor kappaB (NF-kappaB) in mouse lung alveolar type-1 cells' (AT-1 cells) inflammatory response induced by LPS. MATERIALS AND METHODS Gene clone technique was used to over-express caveolin-1 in AT-1 cells by lentivirus vector. The level of tumor necrosis factor alpha (TNF-alpha), interleukin 6 (IL-6), cPLA2, p38 MAPK and NF-kappaB was measured by ELISA, western blotting and EMSA. TREATMENT AT-1 cells were treated with LPS. RESULTS Over-expression of caveolin-1 not only increased the production of pro-inflammatory cytokine TNF-alpha and IL-6, but also enhanced the expression of the cPLA2, p38 MAPK, and NF-kappaB. CONCLUSIONS Our data demonstrated that over-expression of caveolin-1 aggravates the AT-1 injury induced by LPS, involving in modulation of the cPLA2 mediated by the cPLA2/p38 MAPK pathway.
Collapse
Affiliation(s)
- Xue-Jun Lv
- Institute of Respiratory Disease, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
36
|
Lefebvre JS, Marleau S, Milot V, Lévesque T, Picard S, Flamand N, Borgeat P. Toll-like receptor ligands induce polymorphonuclear leukocyte migration: key roles for leukotriene B4 and platelet-activating factor. FASEB J 2009; 24:637-47. [PMID: 19843712 DOI: 10.1096/fj.09-135624] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Activation of toll-like receptors (TLRs) and polymorphonuclear leukocyte (PMN) accumulation at infection sites are critical events of host defense. The involvement of leukotriene (LT) B(4) and platelet-activating factor (PAF) in TLR ligand-induced activation of inflammatory cell functions is essentially unknown. Using an in vitro model of human PMN migration through human endothelial cell monolayers, we demonstrate that prototypic ligands of TLR1/2, 2/6, 3, 4, 5, and 7/8 promote PMN migration, an effect markedly inhibited by 3 LTB(4) receptor antagonists (70-80% inhibition at 100 nM compared to vehicle-treated cells), 3 PAF receptor antagonists (20-50% inhibition at 10 nM), 3 LT biosynthesis inhibitors (75-85% inhibition at 100 nM), and 1 cytosolic phospholipase A(2)alpha (cPLA(2)alpha) inhibitor (90% inhibition at 1 microM). Accordingly, selected TLR ligands caused Ser-505-phosphorylation of cPLA(2)alpha and measurable LTB(4) and PAF biosynthesis in the transmigration assay. As negative controls, interleukin-8- and formyl-methionyl-leucyl-phenylalanine-elicited migration in vitro was not inhibited either by an LTB(4) receptor antagonist or by the cPLA(2)alpha inhibitor. Finally, LTB(4) and PAF receptor antagonists inhibited (up to approximately 65% at optimal doses) TLR ligand-induced PMN infiltration in the mouse air-pouch model. These studies unravel the critical involvement of de novo LTB(4) and PAF biosynthesis in PMN migration elicited by TLR ligands.
Collapse
Affiliation(s)
- Julie S Lefebvre
- Centre de Recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec, 2705 Laurier Blvd., Rm. T1-49, Québec, QC, Canada, G1V 4G2
| | | | | | | | | | | | | |
Collapse
|
37
|
Kitsiouli E, Nakos G, Lekka ME. Phospholipase A2 subclasses in acute respiratory distress syndrome. Biochim Biophys Acta Mol Basis Dis 2009; 1792:941-53. [PMID: 19577642 DOI: 10.1016/j.bbadis.2009.06.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 06/25/2009] [Accepted: 06/25/2009] [Indexed: 01/12/2023]
Abstract
Phospholipases A2 (PLA2) catalyse the cleavage of fatty acids esterified at the sn-2 position of glycerophospholipids. In acute lung injury-acute respiratory distress syndrome (ALI-ARDS) several distinct isoenzymes appear in lung cells and fluid. Some are capable to trigger molecular events leading to enhanced inflammation and lung damage and others have a role in lung surfactant recycling preserving lung function: Secreted forms (groups sPLA2-IIA, -V, -X) can directly hydrolyze surfactant phospholipids. Cytosolic PLA2 (cPLA2-IVA) requiring Ca2+ has a preference for arachidonate, the precursor of eicosanoids which participate in the inflammatory response in the lung. Ca(2+)-independent intracellular PLA2s (iPLA2) take part in surfactant phospholipids turnover within alveolar cells. Acidic Ca(2+)-independent PLA2 (aiPLA2), of lysosomal origin, has additionally antioxidant properties, (peroxiredoxin VI activity), and participates in the formation of dipalmitoyl-phosphatidylcholine in lung surfactant. PAF-AH degrades PAF, a potent mediator of inflammation, and oxidatively fragmented phospholipids but also leads to toxic metabolites. Therefore, the regulation of PLA2 isoforms could be a valuable approach for ARDS treatment.
Collapse
Affiliation(s)
- Eirini Kitsiouli
- Department of Biological Applications and Technologies, School of Sciences and Technologies, University of Ioannina, Greece
| | | | | |
Collapse
|
38
|
Lipids as targets for novel anti-inflammatory therapies. Pharmacol Ther 2009; 124:96-112. [PMID: 19576246 DOI: 10.1016/j.pharmthera.2009.06.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Accepted: 06/12/2009] [Indexed: 02/01/2023]
Abstract
Lipids serve important functions as membrane constituents and also as energy storing molecules. Besides these functions certain lipid species have now been recognized as signalling molecules that regulate a multitude of cellular responses including cell growth and death, and also inflammatory reactions. Bioactive lipids are generated by hydrolysis from membrane lipids mainly by phospholipases giving rise to fatty acids and lysophospholipids that either directly exert their function or are further converted to active mediators. This review will summarize the present knowledge about bioactive lipids that either promote or attenuate inflammatory reactions. These lipids include polyunsaturated fatty acids (PUFA), eicosanoids including the epoxyeicosatrienoic acids (EET), peroxisome proliferation activating receptor (PPAR) activators, cannabinoids and the sphingolipids ceramide, sphingosine 1-phosphate and sphingosylphosphorylcholine.
Collapse
|
39
|
Shimizu T. Lipid mediators in health and disease: enzymes and receptors as therapeutic targets for the regulation of immunity and inflammation. Annu Rev Pharmacol Toxicol 2009; 49:123-50. [PMID: 18834304 DOI: 10.1146/annurev.pharmtox.011008.145616] [Citation(s) in RCA: 430] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Prostaglandins, leukotrienes, platelet-activating factor, lysophosphatidic acid, sphingosine 1-phosphate, and endocannabinoids, collectively referred to as lipid mediators, play pivotal roles in immune regulation and self-defense, and in the maintenance of homeostasis in living systems. They are produced by multistep enzymatic pathways, which are initiated by the de-esterification of membrane phospholipids by phospholipase A2s or sphingo-myelinase. Lipid mediators exert their biological effects by binding to cognate receptors, which are members of the G protein-coupled receptor superfamily. The synthesis of the lipid mediators and subsequent induction of receptor activity is tightly regulated under normal physiological conditions, and enzyme and/or receptor dysfunction can lead to a variety of disease conditions. Thus, the manipulation of lipid mediator signaling, through either enzyme inhibitors or receptor antagonists and agonists, has great potential as a therapeutic approach to disease. In this review, I summarize our current state of knowledge of the synthesis of lipid mediators and the function of their cognate receptors, and discuss the effects of genetic or pharmacological ablation of enzyme or receptor function on various pathophysiological processes.
Collapse
Affiliation(s)
- Takao Shimizu
- Department of Biochemistry and Molecular Biology, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
40
|
Xu M, Dong MQ, Cao FL, Liu ML, Wang YX, Dong HY, Huang YF, Liu Y, Wang XB, Zhang B, Zhao PT, Luo Y, Niu W, Cui Y, Li ZC. Tanshinone IIA reduces lethality and acute lung injury in LPS-treated mice by inhibition of PLA2 activity. Eur J Pharmacol 2009; 607:194-200. [DOI: 10.1016/j.ejphar.2009.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Muñoz NM, Meliton AY, Meliton LN, Dudek SM, Leff AR. Secretory group V phospholipase A2 regulates acute lung injury and neutrophilic inflammation caused by LPS in mice. Am J Physiol Lung Cell Mol Physiol 2009; 296:L879-87. [PMID: 19286925 DOI: 10.1152/ajplung.90580.2008] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
We investigated the regulatory role of 14-kDa secretory group V phospholipase A(2) (gVPLA(2)) in the development of acute lung injury (ALI) and neutrophilic inflammation (NI) caused by intratracheal administration of LPS. Experiments were conducted in gVPLA(2) knockout (pla2g5(-/-)) mice, which lack the gene, and gVPLA(2) wild-type littermate control (pla2g5(+/+)) mice. Indices of pulmonary injury were evaluated 24 h after intratracheal administration of LPS. Expression of gVPLA(2) in microsections of airways and mRNA content in lung homogenates were increased substantially in pla2g5(+/+) mice after LPS-administered compared with saline-treated pla2g5(+/+) mice. By contrast, expression of gVPLA(2) was neither localized in LPS- nor saline-treated pla2g5(-/-) mice. LPS also caused 1) reduced transthoracic static compliance, 2) lung edema, 3) neutrophilic infiltration, and 4) increased neutrophil myeloperoxidase activity in pla2g5(+/+) mice. These events were attenuated in pla2g5(-/-) mice exposed to LPS or in pla2g5(+/+) mice receiving MCL-3G1, a neutralizing MAb directed against gVPLA(2), before LPS administration. Our data demonstrate that gVPLA(2) is an inducible protein in pla2g5(+/+) mice but not in pla2g5(-/-) mice within 24 h after LPS treatment. Specific inhibition of gVPLA(2) with MCL-3G1 or gene-targeted mice lacking gVPLA(2) blocks ALI and attenuates NI caused by LPS.
Collapse
Affiliation(s)
- Nilda M Muñoz
- Dept. of Medicine, M6076, Univ. of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
42
|
Rastogi P, McHowat J. Inhibition of calcium-independent phospholipase A2 prevents inflammatory mediator production in pulmonary microvascular endothelium. Respir Physiol Neurobiol 2009; 165:167-74. [PMID: 19059366 PMCID: PMC2845306 DOI: 10.1016/j.resp.2008.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 11/07/2008] [Accepted: 11/07/2008] [Indexed: 01/11/2023]
Abstract
Inhalation of allergens can result in mast cell degranulation and release of granule contents, including tryptase, in the lung. Injury to human pulmonary microvascular endothelial cells (HMVEC-L) can also result in activation of the coagulation cascade and thrombin generation. We hypothesize that these proteases activate calcium-independent phospholipase A2 (iPLA2), in HMVEC-L, leading to the production of membrane phospholipids-derived inflammatory mediators. Both thrombin and tryptase stimulation of HMVEC-L increased iPLA2 activity that was inhibited by pretreatment with the iPLA2 selective inhibitor bromoenol lactone (BEL). Arachidonic acid and prostaglandin I2 (PGI2) release were also increased in tryptase and thrombin stimulated cells and inhibited by BEL pretreatment. Pretreating the endothelial cells with AACOCF3 a cytosolic PLA2 inhibitor did not inhibit tryptase or thrombin induced arachidonic acid and PGI2 release. In addition thrombin and tryptase also increased HMVEC-L platelet activating factor (PAF) production that significantly contributes to the recruitment and initial adherence of polymorphonuclear neutrophils (PMN) to the endothelium. Tryptase or thrombin stimulated increase in PMN adherence to the endothelium was inhibited by pretreatment of HMVEC-L with BEL or pretreatment of PMN with CV3988, a PAF receptor specific antagonist. Collectively, these data support our hypothesis that iPLA2 activity is responsible for membrane phospholipid hydrolysis in response to tryptase or thrombin stimulation in HMVEC-L. Therefore selective inhibition of iPLA2 may be a pharmacological target to inhibit the early inflammation in pulmonary vasculature that occurs as a consequence of mast cell degranulation or acute lung injury.
Collapse
Affiliation(s)
- Prerna Rastogi
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO 63104, United States
| | | |
Collapse
|
43
|
Avgerinos ED, Kostopanagiotou G, Costopanagiotou C, Kopanakis N, Andreadou I, Lekka M, Nakos G, Smyrniotis V. Intestinal preconditioning ameliorates ischemia-reperfusion induced acute lung injury in rats: an experimental study. J Surg Res 2009; 160:294-301. [PMID: 19439321 DOI: 10.1016/j.jss.2008.12.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 11/25/2008] [Accepted: 12/11/2008] [Indexed: 12/13/2022]
Abstract
BACKGROUND Phospholipases A(2) (PLA(2)) have been implicated in the pathogenesis of acute respiratory distress syndrome (ARDS) induced by intestinal ischemia-reperfusion (IIR). Intestinal ischemic preconditioning (IIP) has been shown to improve intestinal tolerance to subsequent sustained ischemia and limit the systemic inflammatory response. We tested the effect of IIP on the intestinal ischemia-reperfusion-induced ARDS, with particular focus on PLA(2). METHODS Rats were randomized into three groups: (1) sham surgery group (sGroup), 45 min sham intestinal ischemia-4 h reperfusion, (2) IIR group (IIRGroup), 45 min intestinal ischemia-4 h reperfusion, (3) IIP group (ipGroup), three cycles of intestinal ischemia for 4 min and reperfusion for 10 min followed by 45 min intestinal ischemia-4 h reperfusion. At the end of each experiment, blood gases were obtained and bronchoalveolar lavage (BAL) followed. Biochemical (total protein, PLA(2), PAF-AcH) and cytological parameters of the BAL fluid were quantified. Plasma MDA was measured as an indicator of systemic oxidative stress. Comparisons between groups were made using one-way ANOVA followed by post hoc comparison with a Tukey test or Mann-Whitney test when appropriate. Differences were considered significant if P < 0.05. RESULTS Alveolar-arterial O(2) gradient values and wet to dry lung ratio were significantly (P < 0.05) increased in the IIRGroup and this increase was prevented in the ipGroup. Following the same pattern, BAL total protein, PLA(2), and PAF-AcH were significantly lower in the ipGroup. Ischemic preconditioning significantly abolished neutrophil count in BAL fluid. Plasma MDA was significantly lower in the ipGroup. Despite a significant tissue polymorphonuclear reduction, no significant lung or intestinal histologic damage score changes were revealed. CONCLUSIONS Intestinal preconditioning protects IIR-induced lung injury, partly by modulating the arachidonic acid cascade.
Collapse
Affiliation(s)
- Efthimios D Avgerinos
- 2nd Department of Surgery, Medical School, University of Athens, Aretaieion Hospital, Athens, Greece
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Han C, Lim K, Xu L, Li G, Wu T. Regulation of Wnt/beta-catenin pathway by cPLA2alpha and PPARdelta. J Cell Biochem 2009; 105:534-45. [PMID: 18636547 DOI: 10.1002/jcb.21852] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cytosolic phospholipase A(2)alpha (cPLA(2)alpha) is a rate-limiting key enzyme that releases arachidonic acid (AA) from membrane phospholipid for the production of biologically active lipid mediators including prostaglandins, leukotrienes and platelet-activating factor. cPLA(2)alpha is translocated to nuclear envelope in response to intracellular calcium increase and the enzyme is also present inside the cell nucleus; however, the biological function of cPLA(2)alpha in the nucleus remains unknown. Here we show a novel role of cPLA(2)alpha for activation of peroxisome proliferator-activated receptor-delta (PPARdelta) and beta-catenin in the nuclei. Overexpression of cPLA(2)alpha in human cholangiocarcinoma cells induced the binding of PPARdelta to beta-catenin and increased their association with the TCF/LEF response element. These effects are inhibited by the cPLA(2)alpha siRNA and inhibitors as well as by siRNA knockdown of PPARdelta. Overexpression of PPARdelta or treatment with the selective PPARdelta ligand, GW501516, also increased beta-catenin binding to TCF/LEF response element and increased its reporter activity. Addition of AA and GW501516 to nuclear extracts induced a comparable degree of beta-catenin binding to TCF/LEF response element. Furthermore, cPLA(2)alpha protein is present in the PPARdelta and beta-catenin binding complex. Thus the close proximity between cPLA(2)alpha and PPARdelta provides a unique advantage for their efficient functional coupling in the nucleus, where AA produced by cPLA(2)alpha becomes immediately available for PPARdelta binding and subsequent beta-catenin activation. These results depict a novel interaction linking cPLA(2)alpha, PPARdelta and Wnt/beta-catenin signaling pathways and provide insight for further understanding the roles of these key molecules in human cells and diseases.
Collapse
Affiliation(s)
- Chang Han
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | |
Collapse
|
45
|
Abstract
Premature infants often develop serious clinical complications associated with respiratory failure and hyperoxic lung injury that includes lung inflammation and alterations in lung development. The goal of these studies is to test the hypothesis that there are differences in the course of lung injury in newborn mice exposed to 85% or >95% oxygen that provide models to address the differential effects of oxidation and inflammation. Our results indicate differences between the 85% and >95% O2 exposure groups by day 14 in weight gain and lung alveolarization. Inflammation, assessed by neutrophil counts, was observed in both hyperoxia groups by day 3 but was dramatically greater in the >95% O2-exposed groups by day 14 and associated with greater developmental deficits. Cytoplasmic phospholipase A2, cyclooxygenase-2, and 5-lipoxygenase levels were elevated but no patterns of differences were observed between exposure groups. Prostaglandins D2, E2, and F2alpha were increased in the tissues from mouse pups exposed to >95% O2 at 7 d indicating a differential expression of cyclooxygenase-2 products. Our data indicate that there are differences in the models of 85% or >95% O2 exposure and these differences may provide mechanistic insights into hyperoxic lung injury in an immature system.
Collapse
Affiliation(s)
- Lynette K Rogers
- The Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, Ohio State University, Columbus, Ohio 43205, USA.
| | | | | | | |
Collapse
|
46
|
Zhang HQ, Wang HD, Lu DX, Qi RB, Wang YP, Yan YX, Fu YM. Berberine inhibits cytosolic phospholipase A2 and protects against LPS-induced lung injury and lethality independent of the alpha2-adrenergic receptor in mice. Shock 2008; 29:617-22. [PMID: 18414236 DOI: 10.1097/shk.0b013e318157ea14] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Acute lung injury is still a significant clinical problem having a high mortality rate despite significant advances in antimicrobial therapy and supportive care made in the past few years. Our previous study demonstrated that berberine (Ber) remarkably decreased mortality and attenuated the lung injury in mice challenged with LPS, but the mechanism behind this remains unclear. Here, we report that pretreatment with Ber significantly reduced pulmonary edema, neutrophil infiltration, and histopathological alterations; inhibited protein expression and phosphorylation of cytosolic phospholipase A2; and decreased thromboxane A2 release induced by LPS. Yohimbine, an alpha2-adrenergic receptor antagonist, did not antagonize these actions of Ber. Furthermore, pretreatment with Ber decreased TNF-alpha production and mortality in mice challenged with LPS, which were enhanced by yohimbine, and Ber combined with yohimbine also improved survival rate in mice subjected to cecal ligation and puncture. Taken together, these observations indicate that Ber attenuates LPS-induced lung injury by inhibiting TNF-alpha production and cytosolic phospholipase A2 expression and activation in an alpha2-adrenoceptor-independent manner. Berberine combined with yohimbine might provide an effective therapeutic approach to acute lung injury during sepsis.
Collapse
Affiliation(s)
- Hao-qing Zhang
- Department of Pathophysiology, Medical College of Jinan University, Guangzhou, Guangdong, China
| | | | | | | | | | | | | |
Collapse
|
47
|
Multiple roles of phospholipase A2 during lung infection and inflammation. Infect Immun 2008; 76:2259-72. [PMID: 18411286 DOI: 10.1128/iai.00059-08] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
48
|
Miyahara T, Hamanaka K, Weber DS, Anghelescu M, Frost JR, King JA, Parker JC. Cytosolic phospholipase A2 and arachidonic acid metabolites modulate ventilator-induced permeability increases in isolated mouse lungs. J Appl Physiol (1985) 2007; 104:354-62. [PMID: 18006865 DOI: 10.1152/japplphysiol.00959.2006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously reported that the cytosolic phospholipase A(2) (cPLA2) pathway is involved in ventilator-induced lung injury (VILI) produced by high peak inflation pressures (PIP) (J Appl Physiol 98: 1264-1271, 2005), but the relative contributions of the various downstream products of cPLA2 on the acute permeability response were not determined. Therefore, we investigated the role of cPLA2 and the downstream products of arachidonic acid metabolism in the high-PIP ventilation-induced increase in vascular permeability. We perfused isolated mouse lungs and measured the capillary filtration coefficient (K(fc)) after 30 min of ventilation with 9, 25, and 35 cmH2O PIP. In high-PIP-ventilated lungs, K(fc) increased significantly, 2.7-fold, after ventilation with 35 cmH2O PIP compared with paired baseline values and low-PIP-ventilated lungs. Also, increased phosphorylation of lung cPLA2 suggested enzyme activation after high-PIP ventilation. However, treatment with 40 mg/kg arachidonyl trifluoromethyl ketone (an inhibitor of cPLA2) or a combination of 30 microM ibuprofen [a cyclooxygenase (COX) inhibitor], 100 microM nordihydroguaiaretic acid [a lipoxygenase (LOX) inhibitor], and 10 microM 17-octadecynoic acid (a cytochrome P-450 epoxygenase inhibitor) prevented the high-PIP-induced increase in K(fc). Combinations of the inhibitors of COX, LOX, or cytochrome P-450 epoxygenase did not prevent significant increases in K(fc), even though bronchoalveolar lavage levels of the COX or LOX products were significantly reduced. These results suggest that multiple mediators from each pathway contribute to the acute ventilator-induced permeability increase in isolated mouse lungs by mutual potentiation.
Collapse
Affiliation(s)
- Takashige Miyahara
- Dept. of Physiology, MSB 3074, College of Medicine, Univ. of South Alabama, Mobile, AL 36688, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
AOKI-NAGASE T, NAGASE T, OH-HASHI Y, KURIHARA Y, YAMAGUCHI Y, YAMAMOTO H, NAGATA T, KURIHARA H, OUCHI Y. Calcitonin gene-related peptide mediates acid-induced lung injury in mice. Respirology 2007; 12:807-13. [DOI: 10.1111/j.1440-1843.2007.01172.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
50
|
Abstract
The role of oxidative stress has been well appreciated in the development of sepsis-induced acute lung injury (ALI). Oxidative stress in sepsis-induced ALI is believed to be initiated by products of activated lung macrophages and infiltrated neutrophils, promptly propagating to lung epithelial and endothelial cells. This leads to tissue damage and organ dysfunction. On stimulation, neutrophils (PMNs) enable their migration machinery. The lung undergoes changes favoring adhesion and transmigration of PMNs, resulting in PMN accumulation in lung, which is a characteristic of sepsis-induced ALI. Oxidative stress turns on the redox-sensitive transcription factors (NF-kappaB, AP-1), resulting in a large output of proinflammatory cytokines and chemokines, which further aggravate inflammation and oxidative stress. During the process, transcription factor nuclear factor-erythroid 2-p45-related factor 2 (Nrf2) and heme oxygenase (HO) appear to play the counterbalancing roles to limit the propagation of oxidative stress and inflammatory responses in lung. Many antioxidants have been tested to treat sepsis-induced ALI in animal models and in patients with sepsis. However, the results are inconclusive. In this article, we focus on the current understanding of the pathogenesis of sepsis-induced ALI and novel antioxidant strategies for therapeutic purposes.
Collapse
Affiliation(s)
- Ren-Feng Guo
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0602, USA.
| | | |
Collapse
|