1
|
Xing-Xing C, Ri-Jin H, Xin-Ge W, Cai-Ying Y, Qing Y, Ying C, Qi L, Xiao-Xin Z, Lihong Y, Long C, Yu D. Mechanistic exploration of the shenlian formula in the suppression of atherosclerosis progression via network pharmacology and in vivo experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118347. [PMID: 38801914 DOI: 10.1016/j.jep.2024.118347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/21/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Shenlian formula (SL) is a Chinese medicine formula used to curb the development of atherosclerosis (AS) and cardiovascular disease in clinical practice. However, owing to the complexity of compounds and their related multiple targets in traditional Chinese medicine (TCM), it remains difficult and urgent to elucidate the underlying mechanisms at a holistic level. AIM To investigate the intrinsic mechanisms by which SL suppresses AS progression and to gain new insight into its clinical use. METHODS We proposed a network pharmacology-based workflow to evaluate the mechanism by which SL affects AS via data analysis, target prediction, PPI network construction, GO and KEGG analyses, and a "drug-core ingredient-potential target-key pathway" network. Then, non-targeted lipidomic analysis was performed to explore the differential lipid metabolites in AS rats, revealing the possible mechanism by which SL affects atherosclerotic progression. Moreover, an AS rabbit model was constructed and gavaged for SL intervention. Serum lipid profiles and inflammatory cytokine indices were tested as an indication of the mitigating effect of SL on AS. RESULTS A total of 89 bioactive compounds and 298 targets related to SL and AS, which play essential roles in this process, were identified, and a component-target-disease network was constructed. GO and KEGG analyses revealed that SL regulated metabolic pathway, lipids and atherosclerosis, the PI3K-Akt pathway, the MAPK pathway and so on. In vivo experimental validation revealed that a total of 43 different lipid metabolites regulated by SL were identified by non-targeted lipidomics, and glycerophospholipid metabolism was found to be an important mechanism for SL to interfere with AS. SL reduced the plaque area and decreased the levels of inflammatory cytokines (TNF-α and IL-4) and blood lipids (TC, TG, LDL-C, and ApoB) in HFD-induced AS models. In addition, HDL and ApoA1 levels are increased. PLA2 and Lipin1 are highly expressed in AS model, indicating their role in destabilizing glycerophosphatidylcholine metabolism and contributing to the onset and progression of ankylosing spondylitis. Moreover, SL intervention significantly reduced the level of pro-inflammatory cytokines; significantly down-regulated NF-kB/p65 expression, exhibiting anti-inflammatory activity. CONCLUSION The Shenlian formula (SL) plays a pivotal role in the suppression of AS progression by targeting multiple pathways and mechanisms. This study provides novel insights into the essential genes and pathways associated with the prognosis and pathogenesis of AS.
Collapse
Affiliation(s)
- Chen Xing-Xing
- Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New Area, Honghuagang District, Zunyi, 563003, PR China.
| | - Hao Ri-Jin
- Shanxi Pharmaceuticals Vocational College, No. 16, Minhangnanlu, Taiyuan, 030031, PR China.
| | - Wang Xin-Ge
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 100053, No. 5, Beixiange, Xicheng District, Beijing, PR China; Chengdu University of Traditional Chinese Medicine, No. 1166, West Liutai Avenue, Chengdu, 611137, PR China.
| | - Yan Cai-Ying
- Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New Area, Honghuagang District, Zunyi, 563003, PR China.
| | - Yang Qing
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Dongcheng District, Beijing, 100700, PR China.
| | - Chen Ying
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Dongcheng District, Beijing, 100700, PR China.
| | - Li Qi
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Dongcheng District, Beijing, 100700, PR China.
| | - Zhu Xiao-Xin
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Dongcheng District, Beijing, 100700, PR China.
| | - Yang Lihong
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Dongcheng District, Beijing, 100700, PR China.
| | - Cheng Long
- College of Nursing, Chifeng University, 024000, No. 1, Yingbing Road, Hongshan District, Chifeng, PR China.
| | - Dong Yu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 100053, No. 5, Beixiange, Xicheng District, Beijing, PR China.
| |
Collapse
|
2
|
Ma B, Shi J, Zhang Y, Li Z, Yong H, Zhou YN, Liu S, A S, Zhou D. Enzymatically Activatable Polymers for Disease Diagnosis and Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306358. [PMID: 37992728 DOI: 10.1002/adma.202306358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/03/2023] [Indexed: 11/24/2023]
Abstract
The irregular expression or activity of enzymes in the human body leads to various pathological disorders and can therefore be used as an intrinsic trigger for more precise identification of disease foci and controlled release of diagnostics and therapeutics, leading to improved diagnostic accuracy, sensitivity, and therapeutic efficacy while reducing systemic toxicity. Advanced synthesis strategies enable the preparation of polymers with enzymatically activatable skeletons or side chains, while understanding enzymatically responsive mechanisms promotes rational incorporation of activatable units and predictions of the release profile of diagnostics and therapeutics, ultimately leading to promising applications in disease diagnosis and treatment with superior biocompatibility and efficiency. By overcoming the challenges, new opportunities will emerge to inspire researchers to develop more efficient, safer, and clinically reliable enzymatically activatable polymeric carriers as well as prodrugs.
Collapse
Affiliation(s)
- Bin Ma
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiahao Shi
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuhe Zhang
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhili Li
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Haiyang Yong
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ya-Nan Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shuai Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Sigen A
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
3
|
Sasidharan K, Caddeo A, Jamialahmadi O, Noto FR, Tomasi M, Malvestiti F, Ciociola E, Tavaglione F, Mancina RM, Cherubini A, Bianco C, Mirarchi A, Männistö V, Pihlajamäki J, Kärjä V, Grimaudo S, Luukkonen PK, Qadri S, Yki-Järvinen H, Petta S, Manfrini S, Vespasiani-Gentilucci U, Bruni V, Valenti L, Romeo S. IL32 downregulation lowers triglycerides and type I collagen in di-lineage human primary liver organoids. Cell Rep Med 2024; 5:101352. [PMID: 38232700 PMCID: PMC10829727 DOI: 10.1016/j.xcrm.2023.101352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 09/26/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024]
Abstract
Steatotic liver disease (SLD) prevails as the most common chronic liver disease yet lack approved treatments due to incomplete understanding of pathogenesis. Recently, elevated hepatic and circulating interleukin 32 (IL-32) levels were found in individuals with severe SLD. However, the mechanistic link between IL-32 and intracellular triglyceride metabolism remains to be elucidated. We demonstrate in vitro that incubation with IL-32β protein leads to an increase in intracellular triglyceride synthesis, while downregulation of IL32 by small interfering RNA leads to lower triglyceride synthesis and secretion in organoids from human primary hepatocytes. This reduction requires the upregulation of Phospholipase A2 group IIA (PLA2G2A). Furthermore, downregulation of IL32 results in lower intracellular type I collagen levels in di-lineage human primary hepatic organoids. Finally, we identify a genetic variant of IL32 (rs76580947) associated with lower circulating IL-32 and protection against SLD measured by non-invasive tests. These data suggest that IL32 downregulation may be beneficial against SLD.
Collapse
Affiliation(s)
- Kavitha Sasidharan
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Andrea Caddeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Francesca Rita Noto
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Melissa Tomasi
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Malvestiti
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Ester Ciociola
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Federica Tavaglione
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Rosellina M Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Alessandro Cherubini
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristiana Bianco
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Angela Mirarchi
- Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Clinical Nutrition and Obesity Centre, Kuopio University Hospital, Kuopio, Finland
| | - Vesa Kärjä
- Department of Pathology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Stefania Grimaudo
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Panu K Luukkonen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Sami Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Silvia Manfrini
- Operative Unit of Endocrinology and Diabetes, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Endocrinology and Diabetes, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Umberto Vespasiani-Gentilucci
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Vincenzo Bruni
- Operative Unit of Bariatric Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Luca Valenti
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy.
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy; Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
4
|
Lyu B, Li J, Niemeyer B, Anderson D, Beerntsen B, Song Q. Identification, structural modeling, gene expression analysis and RNAi effect of putative phospholipase A 2 in the lone star tick Amblyomma americanum. Ticks Tick Borne Dis 2024; 15:102256. [PMID: 37734164 DOI: 10.1016/j.ttbdis.2023.102256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/15/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
Amblyomma americanum, also known as the lone star tick, is a small arachnid that feeds on blood and can spread disease to humans and other animals. Despite the overlapped ecological niche, geographic distribution, and host selection, there is no proof that A. americanum transmits the pathogen Borrelia burgdorferi that causes Lyme disease. Studies have shown that phospholipase A2 (PLA2) may act as a tool to eliminate B. burgdorferi, but particular PLA2 genes in A. americanum have not been identified and functionally characterized. Using the de novo sequencing method, we identified 42 putative A. americanum PLA2 (pAaPLA2) homologs in the present study, of which three pAaPLA2 had calcium binding sites and canonical histidine catalytic sites. Then, we determined phylogenetic relationships, sequence alignments, and conserved protein motifs of these pAaPLA2s. Protein structural analysis demonstrated that pAaPLA2s primarily consisted of α-helices, β-sheets, and random coils. These genes were predicted to be engaged in the phospholipid metabolic process, arachidonic acid secretion, and PLA2 activity by functional annotation analysis. A transcriptional factor (Bgb) was discovered that interacted with pAaPLA2 proteins that may have unrecognized roles in regulating neuronal development. Based on the RNA-seq data, we surveyed expression profiles of key pAaPLA2-related genes to reveal putative modulatory networks of these genes. RNAi knockdown of pAaPLA2_1, a dominant isoform in A. americanum, led to decreased bacterial inhibition ability, suggesting pAaPLA2 may play an important role in mediating immune responses. Collectively, this study provides essential evidence of the identification, gene structure, phylogeny, and expression analysis of pAaPLA2 genes in A. americanum, and offers a deeper understanding of the putative borreliacidal roles in the lone star tick.
Collapse
Affiliation(s)
- Bo Lyu
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA
| | - Jingjing Li
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA
| | - Brigid Niemeyer
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA
| | - Deborah Anderson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Brenda Beerntsen
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA; Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Qisheng Song
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
5
|
Mehta D, Shaikh S, Mohanty B, Chaudhari P, Waghmare SK. Secretory phospholipase (sPLA 2-IIA) regulates breast cancer stem cells differentiation and metastatic potential. Biochem Biophys Res Commun 2023; 677:98-104. [PMID: 37566923 DOI: 10.1016/j.bbrc.2023.07.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023]
Abstract
Breast cancer is the second most cancer worldwide in females. The primary factor responsible for tumor recurrence is the presence of breast cancer stem cells (BCSCs), which escape the chemo-radiotherapy. In this study, we have investigated the role of Secretory phospholipase-A2 Group 2A (sPLA2-IIA) that is overexpressed in BCSCs of MCF7 and MDA-MB-231 breast cancer cell lines. Further, overexpression of sPLA2-IIA revealed an increased EGFR/JNK/c-JUN/c-FOS signaling in BCSCs, while sPLA2-IIA knockdown significantly reduced the percentage of BCSCs and decreased signaling in both the cell lines. Importantly, sPLA2-IIA knockdown showed differentiation of BCSCs. Strikingly, PET imaging showed a decreased metastatic potential of BCSCs. Our study revealed a novel role of sPLA2-IIA in regulating BCSCs, which play a crucial role in regulating the differentiation and metastatic potential of BCSCs.
Collapse
Affiliation(s)
- Darshan Mehta
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Sana Shaikh
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, Maharashtra, India
| | - Bhabani Mohanty
- Small Animal Imaging Facility (SAIF), Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, Maharashtra, India
| | - Pradip Chaudhari
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India; Small Animal Imaging Facility (SAIF), Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, Maharashtra, India
| | - Sanjeev K Waghmare
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India.
| |
Collapse
|
6
|
Wu Y, Pernet E, Touqui L. Modulation of Airway Expression of the Host Bactericidal Enzyme, sPLA2-IIA, by Bacterial Toxins. Toxins (Basel) 2023; 15:440. [PMID: 37505708 PMCID: PMC10467128 DOI: 10.3390/toxins15070440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/23/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
Host molecules with antimicrobial properties belong to a large family of mediators including type-IIA secreted phospholipase A2 (sPLA2-IIA). The latter is a potent bactericidal agent with high selectivity against Gram-positive bacteria, but it may also play a role in modulating the host inflammatory response. However, several pathogen-associated molecular patterns (PAMPs) or toxins produced by pathogenic bacteria can modulate the levels of sPLA2-IIA by either inducing or inhibiting its expression in host cells. Thus, the final sPLA2-IIA concentration during the infection process is determined by the orchestration between the levels of toxins that stimulate and those that downregulate the expression of this enzyme. The stimulation of sPLA2-IIA expression is a process that participates in the clearance of invading bacteria, while inhibition of this expression highlights a mechanism by which certain bacteria can subvert the immune response and invade the host. Here, we will review the major functions of sPLA2-IIA in the airways and the role of bacterial toxins in modulating the expression of this enzyme. We will also summarize the major mechanisms involved in this modulation and the potential consequences for the pulmonary host response to bacterial infection.
Collapse
Affiliation(s)
- Yongzheng Wu
- Unité de Biologie Cellulaire de l’Infection Microbionne, CNRS UMR3691, Institut Pasteur, Université de Paris Cité, 75015 Paris, France;
| | - Erwan Pernet
- Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - Lhousseine Touqui
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
- Institut Pasteur, Université de Paris Cité, Mucoviscidose et Bronchopathies Chroniques, 75015 Paris, France
| |
Collapse
|
7
|
Interplay between C1-inhibitor and group IIA secreted phospholipase A 2 impairs their respective function. Immunol Res 2023; 71:70-82. [PMID: 36385678 PMCID: PMC9845149 DOI: 10.1007/s12026-022-09331-7] [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: 07/22/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022]
Abstract
High levels of human group IIA secreted phospholipase A2 (hGIIA) have been associated with various inflammatory disease conditions. We have recently shown that hGIIA activity and concentration are increased in the plasma of patients with hereditary angioedema due to C1-inhibitor deficiency (C1-INH-HAE) and negatively correlate with C1-INH plasma activity. In this study, we analyzed whether the presence of both hGIIA and C1-INH impairs their respective function on immune cells. hGIIA, but not recombinant and plasma-derived C1-INH, stimulates the production of IL-6, CXCL8, and TNF-α from peripheral blood mononuclear cells (PBMCs). PBMC activation mediated by hGIIA is blocked by RO032107A, a specific hGIIA inhibitor. Interestingly, C1-INH inhibits the hGIIA-induced production of IL-6, TNF-α, and CXCL8, while it does not affect hGIIA enzymatic activity. On the other hand, hGIIA reduces the capacity of C1-INH at inhibiting C1-esterase activity. Spectroscopic and molecular docking studies suggest a possible interaction between hGIIA and C1-INH but further experiments are needed to confirm this hypothesis. Together, these results provide evidence for a new interplay between hGIIA and C1-INH, which may be important in the pathophysiology of hereditary angioedema.
Collapse
|
8
|
Varricchi G, Poto R, Ferrara AL, Gambino G, Marone G, Rengo G, Loffredo S, Bencivenga L. Angiopoietins, vascular endothelial growth factors and secretory phospholipase A 2 in heart failure patients with preserved ejection fraction. Eur J Intern Med 2022; 106:111-119. [PMID: 36280524 DOI: 10.1016/j.ejim.2022.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Heart failure (HF) is a growing public health burden, with high prevalence and mortality rates. A proportion of patients with HF have a normal ventricular ejection fraction (EF), referred to as HF with preserved EF (HFpEF), as opposed to patients with HF with reduced ejection fraction (HFrEF). HFpEF currently accounts for about 50% of all HF patients, and its prevalence is rising. Angiopoietins (ANGPTs), vascular endothelial growth factors (VEGFs) and secretory phospholipases A2 (sPLA2s) are proinflammatory mediators and key regulators of endothelial cells. METHODS The aim of this study was to analyze the plasma concentrations of angiogenic (ANGPT1, ANGPT2, VEGF-A) and lymphangiogenic (VEGF-C, VEGF-D) factors and the plasma activity of sPLA2 in patients with HFpEF and HFrEF compared to healthy controls. RESULTS The concentration of ANGPT1 was reduced in HFrEF compared to HFpEF patients and healthy controls. ANGPT2 levels were increased in both HFrEF and HFpEF subjects compared to controls. The ANGPT2/ANGPT1 ratio was increased in HFrEF patients compared to controls. The concentrations of both VEGF-A and VEGF-C did not differ among the three groups examined. VEGF-D was increased in both HFrEF and HFpEF patients compared to controls. Plasma activity of sPLA2 was increased in HFrEF but not in HFpEF patients compared to controls. CONCLUSIONS Our results indicate that three different classes of proinflammatory regulators of vascular permeability and smoldering inflammation are selectively altered in HFrEF or HFpEF patients. Studies involving larger cohorts of these patients will be necessary to demonstrate the clinical implications of our findings.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuseppina Gambino
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Istituti Clinici Scientifici Maugeri SpA Società Benefit, 82037, Telese, (BN), Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Leonardo Bencivenga
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131, Naples, Italy; Gèrontopole de Toulouse, Institut du Vieillissement, CHU de Toulouse, 31000, Toulouse, France
| |
Collapse
|
9
|
Treatment of Mouse Sperm with a Non-Catalytic Mutant of PLA2G10 Reveals That PLA2G10 Improves In Vitro Fertilization through Both Its Enzymatic Activity and as Ligand of PLA2R1. Int J Mol Sci 2022; 23:ijms23148033. [PMID: 35887380 PMCID: PMC9320362 DOI: 10.3390/ijms23148033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 12/04/2022] Open
Abstract
The group X secreted phospholipase A2 (PLA2G10) is present at high levels in mouse sperm acrosome. The enzyme is secreted during capacitation and amplifies the acrosome reaction and its own secretion via an autocrine loop. PLA2G10 also improves the rate of fertilization. In in vitro fertilization (IVF) experiments, sperm from Pla2g10-deficient mice produces fewer two-cell embryos, and the absence of PLA2G10 is rescued by adding recombinant enzymes. Moreover, wild-type (WT) sperm treated with recombinant PLA2G10 produces more two-cell embryos. The effects of PLA2G10 on mouse fertility are inhibited by sPLA2 inhibitors and rescued by products of the enzymatic reaction such as free fatty acids, suggesting a role of catalytic activity. However, PLA2G10 also binds to mouse PLA2R1, which may play a role in fertility. To determine the relative contribution of enzymatic activity and PLA2R1 binding in the profertility effect of PLA2G10, we tested H48Q-PLA2G10, a catalytically-inactive mutant of PLA2G10 with low enzymatic activity but high binding properties to PLA2R1. Its effect was tested in various mouse strains, including Pla2r1-deficient mice. H48Q-PLA2G10 did not trigger the acrosome reaction but was as potent as WT-PLA2G10 to improve IVF in inbred C57Bl/6 mice; however, this was not the case in OF1 outbred mice. Using gametes from these mouse strains, the effect of H48Q-PLA2G10 appeared dependent on both spermatozoa and oocytes. Moreover, sperm from C57Bl/6 Pla2r1-deficient mice were less fertile and lowered the profertility effects of H48Q-PLA2G10, which were completely suppressed when sperm and oocytes were collected from Pla2r1-deficient mice. Conversely, the effect of WT-PLA2G10 was not or less sensitive to the absence of PLA2R1, suggesting that the effect of PLA2G10 is polymodal and complex, acting both as an enzyme and a ligand of PLA2R1. This study shows that the action of PLA2G10 on gametes is complex and can simultaneously activate the catalytic pathway and the PLA2R1-dependent receptor pathway. This work also shows for the first time that PLA2G10 binding to gametes’ PLA2R1 participates in fertilization optimization.
Collapse
|
10
|
Kartha S, Ghimire P, Winkelstein BA. Inhibiting spinal secretory phospholipase A 2 after painful nerve root injury attenuates established pain and spinal neuronal hyperexcitability by altering spinal glutamatergic signaling. Mol Pain 2021; 17:17448069211066221. [PMID: 34919471 PMCID: PMC8721705 DOI: 10.1177/17448069211066221] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Neuropathic injury is accompanied by chronic inflammation contributing to the onset and maintenance of pain after an initial insult. In addition to their roles in promoting immune cell activation, inflammatory mediators like secretory phospholipase A2 (sPLA2) modulate nociceptive and excitatory neuronal signaling during the initiation of pain through hydrolytic activity. Despite having a known role in glial activation and cytokine release, it is unknown if sPLA2 contributes to the maintenance of painful neuropathy and spinal hyperexcitability later after neural injury. Using a well-established model of painful nerve root compression, this study investigated if inhibiting spinal sPLA2 7 days after painful injury modulates the behavioral sensitivity and/or spinal dorsal horn excitability that is typically evident. The effects of sPLA2 inhibition on altered spinal glutamatergic signaling was also probed by measuring spinal intracellular glutamate levels and spinal glutamate transporter (GLAST and GLT1) and receptor (mGluR5, GluR1, and NR1) expression. Spinal sPLA2 inhibition at day 7 abolishes behavioral sensitivity, reduces both evoked and spontaneous neuronal firing in the spinal cord, and restores the distribution of neuronal phenotypes to those of control conditions. Inhibiting spinal sPLA2 also increases intracellular glutamate concentrations and restores spinal expression of GLAST, GLT1, mGluR5, and GluR1 to uninjured expression with no effect on NR1. These findings establish a role for spinal sPLA2 in maintaining pain and central sensitization after neural injury and suggest this may be via exacerbating glutamate excitotoxicity in the spinal cord.
Collapse
Affiliation(s)
- Sonia Kartha
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Prabesh Ghimire
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Beth A Winkelstein
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.,Department of Neurosurgery, University of Pennsylvania, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Lipid Droplets, Phospholipase A 2, Arachidonic Acid, and Atherosclerosis. Biomedicines 2021; 9:biomedicines9121891. [PMID: 34944707 PMCID: PMC8699036 DOI: 10.3390/biomedicines9121891] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/01/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023] Open
Abstract
Lipid droplets, classically regarded as static storage organelles, are currently considered as dynamic structures involved in key processes of lipid metabolism, cellular homeostasis and signaling. Studies on the inflammatory state of atherosclerotic plaques suggest that circulating monocytes interact with products released by endothelial cells and may acquire a foamy phenotype before crossing the endothelial barrier and differentiating into macrophages. One such compound released in significant amounts into the bloodstream is arachidonic acid, the common precursor of eicosanoids, and a potent inducer of neutral lipid synthesis and lipid droplet formation in circulating monocytes. Members of the family of phospholipase A2, which hydrolyze the fatty acid present at the sn-2 position of phospholipids, have recently emerged as key controllers of lipid droplet homeostasis, regulating their formation and the availability of fatty acids for lipid mediator production. In this paper we discuss recent findings related to lipid droplet dynamics in immune cells and the ways these organelles are involved in regulating arachidonic acid availability and metabolism in the context of atherosclerosis.
Collapse
|
12
|
Beaulieu D, Attwe A, Breau M, Lipskaia L, Marcos E, Born E, Huang J, Abid S, Derumeaux G, Houssaini A, Maitre B, Lefevre M, Vienney N, Bertolino P, Jaber S, Noureddine H, Goehrig D, Vindrieux D, Bernard D, Adnot S. Phospholipase A2 receptor 1 promotes lung cell senescence and emphysema in obstructive lung disease. Eur Respir J 2021; 58:13993003.00752-2020. [PMID: 33509955 DOI: 10.1183/13993003.00752-2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 12/28/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Cell senescence is a key process in age-associated dysfunction and diseases, notably chronic obstructive pulmonary disease (COPD). We previously identified phospholipase A2 receptor 1 (PLA2R1) as a positive regulator of cell senescence acting via Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signalling. Its role in pathology, however, remains unknown. Here, we assessed PLA2R1-induced senescence in COPD and lung emphysema pathogenesis. METHODS We assessed cell senescence in lungs and cultured lung cells from patients with COPD and controls subjected to PLA2R1 knockdown, PLA2R1 gene transduction and treatment with the JAK1/2 inhibitor ruxolitinib. To assess whether PLA2R1 upregulation caused lung lesions, we developed transgenic mice overexpressing PLA2R1 (PLA2R1-TG) and intratracheally injected wild-type mice with a lentiviral vector carrying the Pla2r1 gene (LV-PLA2R1 mice). RESULTS We found that PLA2R1 was overexpressed in various cell types exhibiting senescence characteristics in COPD lungs. PLA2R1 knockdown extended the population doubling capacity of these cells and inhibited their pro-inflammatory senescence-associated secretory phenotype (SASP). PLA2R1-mediated cell senescence in COPD was largely reversed by treatment with the potent JAK1/2 inhibitor ruxolitinib. Five-month-old PLA2R1-TG mice exhibited lung cell senescence, and developed lung emphysema and lung fibrosis together with pulmonary hypertension. Treatment with ruxolitinib induced reversal of lung emphysema and fibrosis. LV-PLA2R1-treated mice developed lung emphysema within 4 weeks and this was markedly attenuated by concomitant ruxolitinib treatment. CONCLUSIONS Our data support a major role for PLA2R1 activation in driving lung cell senescence and lung alterations in COPD. Targeting JAK1/2 may represent a promising therapeutic approach for COPD.
Collapse
Affiliation(s)
- Delphine Beaulieu
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France.,These two authors contributed equally
| | - Aya Attwe
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France.,Environmental Health Research Laboratory (EHRL), Faculty of Sciences V, Lebanese University, Nabatieh, Lebanon.,These two authors contributed equally
| | - Marielle Breau
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Larissa Lipskaia
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Elisabeth Marcos
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Emmanuelle Born
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Jin Huang
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Shariq Abid
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Geneviève Derumeaux
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Amal Houssaini
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Bernard Maitre
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Marine Lefevre
- Département Anatomopathologie, Institut Mutualiste Montsouris, Paris, France
| | - Nora Vienney
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Sara Jaber
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Hiba Noureddine
- Environmental Health Research Laboratory (EHRL), Faculty of Sciences V, Lebanese University, Nabatieh, Lebanon
| | - Delphine Goehrig
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - David Vindrieux
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Centre Léon Bérard, Lyon, France.,These two authors are joint senior authors
| | - Serge Adnot
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France .,Institute for Lung Health, University of Giessen, Giessen, Germany.,These two authors are joint senior authors
| |
Collapse
|
13
|
Park HK, Park SH, Lee M, Kim GR, Park M, Yang SC, Kim YS, Lim HJ, Kim HR, Song H. Secretory phospholipase A2-X (Pla2g10) is a novel progesterone receptor target gene exclusively induced in uterine luminal epithelium for uterine receptivity in mice. Cell Biosci 2020; 10:132. [PMID: 33292460 PMCID: PMC7678068 DOI: 10.1186/s13578-020-00495-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/09/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Aberration of estrogen (E2) and/or progesterone (P4) signaling pathways affects expression of their target genes, which may lead to failure of embryo implantation and following pregnancy. Although many target genes of progesterone receptors (PRs) have been identified in uterine stroma, only a few PR targets have been reported in the epithelium. Secretory phospholipase A2-(PLA2)-X, a member of the PLA2 family that releases arachidonic acids for the synthesis of prostaglandins that are important for embryo implantation, is dysregulated in the endometrium of patients suffering from repeated implantation failure. However, it is not clear whether sPLA2-X is directly regulated by ovarian steroid hormones for embryo implantation in the uterus. RESULT P4 induced the Pla2g10 encoding of secretory PLA2-X in the apical region of uterine LE of ovariectomized mice via PR in both time- and dose-dependent manners, whereas E2 significantly inhibited it. This finding is consistent with the higher expression of Pla2g10 at the diestrus stage, when P4 is elevated during the estrous cycle, and at P4-treated delayed implantation. The level of Pla2g10 on day 4 of pregnancy (day 4) was dramatically decreased on day 5, when PRs are absent in the LE. Luciferase assays of mutagenesis in uterine epithelial cells demonstrated that four putative PR response elements in a Pla2g10 promoter region are transcriptionally active for Pla2g10. Intrauterine delivery of small interfering RNA for Pla2g10 on day 3 significantly reduced the number of implantation sites, reinforcing the critical function(s) of Pla2g10 for uterine receptivity in mice. CONCLUSIONS Pla2g10 is a novel PR target gene whose expression is exclusively localized in the apical region of the uterine LE for uterine receptivity for embryo implantation in mice.
Collapse
Affiliation(s)
- Hee Kyoung Park
- Department of Biomedical Science, CHA University, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Collage of Medicine, Gil Medical Center, Gachon University, Incheon, 21565, Republic of Korea
| | - So Hee Park
- Department of Biomedical Science, CHA University, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Miji Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Gyeong Ryeong Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Seung Chel Yang
- Department of Biomedical Science, CHA University, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Yeon Sun Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Hyunjung J Lim
- Department of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hye-Ryun Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea.
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea.
| |
Collapse
|
14
|
Varricchi G, Loffredo S, Bencivenga L, Ferrara AL, Gambino G, Ferrara N, de Paulis A, Marone G, Rengo G. Angiopoietins, Vascular Endothelial Growth Factors and Secretory Phospholipase A 2 in Ischemic and Non-Ischemic Heart Failure. J Clin Med 2020; 9:jcm9061928. [PMID: 32575548 PMCID: PMC7356305 DOI: 10.3390/jcm9061928] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a growing public health burden, with high prevalence and mortality rates. In contrast to ischemic heart failure (IHF), the diagnosis of non-ischemic heart failure (NIHF) is established in the absence of coronary artery disease. Angiopoietins (ANGPTs), vascular endothelial growth factors (VEGFs) and secretory phospholipases A2 (sPLA2s) are proinflammatory mediators and key regulators of endothelial cells. In the present manuscript, we analyze the plasma concentrations of angiogenic (ANGPT1, ANGPT2, VEGF-A) and lymphangiogenic (VEGF-C, VEGF-D) factors and the plasma activity of sPLA2 in patients with IHF and NIHF compared to healthy controls. The concentrations of ANGPT1, ANGPT2 and their ratio significantly differed between HF patients and healthy controls. Similarly, plasma levels of VEGF-D and sPLA2 activity were higher in HF as compared to controls. Concentrations of ANGPT2 and the ANGPT2/ANGPT1 ratio (an index of vascular permeability) were increased in NIHF patients. VEGF-A and VEGF-C concentrations did not differ among the three examined groups. Interestingly, VEGF-D was selectively increased in IFH patients compared to controls. Plasma activity of sPLA2 was increased in IHF and NIHF patients compared to controls. Our results indicate that several regulators of vascular permeability and smoldering inflammation are specifically altered in IHF and NIHF patients. Studies involving larger cohorts of these patients will be necessary to demonstrate the clinical implications of our findings.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80100 Naples, Italy
- Correspondence: (G.V.); (S.L.)
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80100 Naples, Italy
- Correspondence: (G.V.); (S.L.)
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80100 Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
| | - Giuseppina Gambino
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80100 Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Istituti Clinici Scientifici Maugeri SpA Società Benefit, Via Bagni Vecchi, 1, 82037 Telese BN, Italy
| |
Collapse
|
15
|
Generation of a conditional transgenic mouse model expressing human Phospholipase A2 Receptor 1. Sci Rep 2020; 10:8190. [PMID: 32424163 PMCID: PMC7235081 DOI: 10.1038/s41598-020-64863-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 04/22/2020] [Indexed: 11/09/2022] Open
Abstract
The Phospholipase A2 Receptor 1 (PLA2R1) was first identified for its ability to bind some secreted PLA2s (sPLA2s). It belongs to the C-type lectin superfamily and it binds different types of proteins. It is likely a multifunctional protein that plays a role i) in inflammation and inflammatory diseases, ii) in cellular senescence, a mechanism participating in aging and age-related diseases including cancer, and iii) in membranous nephropathy (MN), a rare autoimmune kidney disease where PLA2R1 is the major autoantigen. To help study the role of PLA2R1 in these pathophysiological conditions, we have generated a versatile NeoR-hPLA2R1 conditional transgenic mice which will allow the specific expression of human PLA2R1 (hPLA2R1) in relevant organs and cells following Cre recombinase-driven excision of the NeoR-stop cassette flanked by LoxP sites. Proof-of-concept breeding of NeoR-hPLA2R1 mice with the ubiquitous adenoviral EIIa promoter-driven Cre mouse line resulted in the expected excision of the NeoR-stop cassette and the expression of hPLA2R1 in all tested tissues. These Tg-hPLA2R1 animals breed normally, with no reproduction or apparent growth defect. These models, especially the NeoR-hPLA2R1 conditional transgenic mouse line, will facilitate the future investigation of PLA2R1 functions in relevant pathophysiological contexts, including inflammatory diseases, age-related diseases and MN.
Collapse
|
16
|
Abstract
Lipid droplets have a unique structure among organelles consisting of a dense hydrophobic core of neutral lipids surrounded by a single layer of phospholipids decorated with various proteins. Often labeled merely as passive fat storage repositories, they in fact have a remarkably dynamic life cycle. Being formed within the endoplasmic reticulum membrane, lipid droplets rapidly grow, shrink, traverse the cytosol, and engage in contacts with other organelles to exchange proteins and lipids. Their lipid and protein composition changes dynamically in response to cellular states and nutrient availability. Remarkably, their biogenesis is induced when cells experience various forms of nutrient, energy, and redox imbalances, including lipid excess and complete nutrient deprivation. Cancer cells are continuously exposed to nutrient and oxygen fluctuations and have the capacity to switch between alternative nutrient acquisition and metabolic pathways in order to strive even during severe stress. Their supply of lipids is ensured by a series of nutrient uptake and scavenging mechanisms, upregulation of de novo lipid synthesis, repurposing of their structural lipids via enzymatic remodeling, or lipid recycling through autophagy. Importantly, most of these pathways of lipid acquisition converge at lipid droplets, which combine different lipid fluxes and control their usage based on specific cellular needs. It is thus not surprising that lipid droplet breakdown is an elaborately regulated process that occurs via a complex interplay of neutral lipases and autophagic degradation. Cancer cells employ lipid droplets to ensure energy production and redox balance, modulate autophagy, drive membrane synthesis, and control its composition, thereby minimizing stress and fostering tumor progression. As regulators of (poly)unsaturated fatty acid trafficking, lipid droplets are also emerging as modulators of lipid peroxidation and sensitivity to ferroptosis. Clearly, dysregulated lipid droplet turnover may also be detrimental to cancer cells, which should provide potential therapeutic opportunities in the future. In this review, we explore how lipid droplets consolidate lipid acquisition and trafficking pathways in order to match lipid supply with the requirements for cancer cell survival, growth, and metastasis.
Collapse
|
17
|
Wu C, Su J, Wang X, Wang J, Xiao K, Li Y, Xiao Q, Ling M, Xiao Y, Qin C, Long W, Zhang F, Pan Y, Xiang F, Liu Q. Overexpression of the phospholipase A2 group V gene in glioma tumors is associated with poor patient prognosis. Cancer Manag Res 2019; 11:3139-3152. [PMID: 31114356 PMCID: PMC6489671 DOI: 10.2147/cmar.s199207] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/17/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose: Gliomas are the most common primary malignant neoplasms of the central nervous system. Secreted phospholipases A2 (sPLA2s) are known to play an important role in various physiological processes, including bioactive lipid production, defense mechanisms, and cell signaling. However, their roles and clinical importance in gliomas remain unclear. Patients and methods: In this study, we analyzed the association between the expression of various sPLA2-encoding genes and the clinicopathology of gliomas, using the data of 1047 patients obtained from a public database. Immunohistochemical analysis of 82 glioma tissues was also carried out to assess the relationship between phospholipase A2 group V (PLA2G5) protein expression and the World Health Organization (WHO) glioma grades. Results: We found that high PLA2G5 gene expression was associated with unfavorable prognosis in both low-grade and high-grade gliomas. The immunohistochemistry of the 82 glioma tissues further confirmed that PLA2G5 protein expression was dependent on the WHO glioma grade. In addition, we found a correlation between PLA2G5 gene expression and both epithelial-mesenchymal transition and the isocitrate dehydrogenase 1 mutation status in these tumors. Conclusion: Our results indicate that PLA2G5 could be a potential biomarker for predicting poor prognosis in patients with gliomas.
Collapse
Affiliation(s)
- Changwu Wu
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Jun Su
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Xiangyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Junquan Wang
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Kai Xiao
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Yang Li
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Qun Xiao
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Min Ling
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Yao Xiao
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Chaoying Qin
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Wenyong Long
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China.,Institute of Skull Base Surgery and Neuro-Oncology at Hunan Neurosurgery Institute of Central South University, Changsha, Hunan, People's Republic of China
| | - Fengqi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Yimin Pan
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Feng Xiang
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China.,Institute of Skull Base Surgery and Neuro-Oncology at Hunan Neurosurgery Institute of Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
18
|
Martinez G, Hograindleur JP, Jeammet L, Le Blévec E, Coutton C, Mermillod P, Lambeau G, Schmitt E, Ray PF, Arnoult C. Enzymatic activity of mouse group X-sPLA2 improves in vitro production of preimplantation bovine embryos. Theriogenology 2019; 131:113-122. [PMID: 30959437 DOI: 10.1016/j.theriogenology.2019.03.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/27/2019] [Accepted: 03/30/2019] [Indexed: 10/27/2022]
Abstract
Assisted reproductive technologies (ART) are widely used for both humans and domestic animals. In bovine species, in vitro embryo production is increasingly used and significant efforts are being made to optimize media and culture conditions. Phospholipase A2 (PLA2) are lipolytic enzymes that hydrolyze glycerophospholipids to produce free fatty acids and lysophospholipids that have been found to be critical for many biological processes. Mouse group X secreted PLA2 (mGX) is abundant in the male reproductive tract and its use during sperm capacitation has been shown to improve in vitro production of viable embryos in a mouse model. Here, we examined its effect in the bovine species, testing the impact of mGX on the three steps involved in vitro production of preimplantation embryos: oocyte maturation, fertilization and preimplantation development. We found that incubating cumulus oocyte complexes (COC) or gametes with mGX resulted in increased blastocyst hatching and blastocyst production, respectively. The increases of embryo production induced by the phospholipase mGX were not observed for the catalytically inactive mutant H48Q-mGX, suggesting that these effects require the enzymatic activity of mGX. We also tested bGIB, a bovine homolog of mGX. bGIB failed to improve blastocyst production, underlining the high specificity of mGX. In conclusion, the results presented show that the effects of mGX are not restricted to the mouse model and that it is potent in the bovine species as well. This result strengthens the potential of mGX as a "pro-fertility drug" for mammalian reproduction.
Collapse
Affiliation(s)
- Guillaume Martinez
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France; IMV Technologies, ZI N 1 Est, F-61300, L'Aigle, France
| | - Jean-Pascal Hograindleur
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France
| | - Louise Jeammet
- Université Côte d'Azur, CNRS, IPMC, F-06560, Valbonne, Sophia Antipolis, France
| | - Emilie Le Blévec
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France
| | - Charles Coutton
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France
| | - Pascal Mermillod
- Laboratoire Physiologie de la Reproduction et des Comportements, INRA UMR 7247, Inra-Cnrs-Université de Tours-Haras Nationaux, F-37380, Nouzilly, France
| | - Gérard Lambeau
- Université Côte d'Azur, CNRS, IPMC, F-06560, Valbonne, Sophia Antipolis, France
| | - Eric Schmitt
- IMV Technologies, ZI N 1 Est, F-61300, L'Aigle, France
| | - Pierre F Ray
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France; CHU de Grenoble, UM GI-DPI, Grenoble, F-38000, France
| | - Christophe Arnoult
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France.
| |
Collapse
|
19
|
van Hensbergen VP, Movert E, de Maat V, Lüchtenborg C, Le Breton Y, Lambeau G, Payré C, Henningham A, Nizet V, van Strijp JAG, Brügger B, Carlsson F, McIver KS, van Sorge NM. Streptococcal Lancefield polysaccharides are critical cell wall determinants for human Group IIA secreted phospholipase A2 to exert its bactericidal effects. PLoS Pathog 2018; 14:e1007348. [PMID: 30321240 PMCID: PMC6201954 DOI: 10.1371/journal.ppat.1007348] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 10/25/2018] [Accepted: 09/20/2018] [Indexed: 12/21/2022] Open
Abstract
Human Group IIA secreted phospholipase A2 (hGIIA) is an acute phase protein with bactericidal activity against Gram-positive bacteria. Infection models in hGIIA transgenic mice have suggested the importance of hGIIA as an innate defense mechanism against the human pathogens Group A Streptococcus (GAS) and Group B Streptococcus (GBS). Compared to other Gram-positive bacteria, GAS is remarkably resistant to hGIIA activity. To identify GAS resistance mechanisms, we exposed a highly saturated GAS M1 transposon library to recombinant hGIIA and compared relative mutant abundance with library input through transposon-sequencing (Tn-seq). Based on transposon prevalence in the output library, we identified nine genes, including dltA and lytR, conferring increased hGIIA susceptibility. In addition, seven genes conferred increased hGIIA resistance, which included two genes, gacH and gacI that are located within the Group A Carbohydrate (GAC) gene cluster. Using GAS 5448 wild-type and the isogenic gacI mutant and gacI-complemented strains, we demonstrate that loss of the GAC N-acetylglucosamine (GlcNAc) side chain in the ΔgacI mutant increases hGIIA resistance approximately 10-fold, a phenotype that is conserved across different GAS serotypes. Increased resistance is associated with delayed penetration of hGIIA through the cell wall. Correspondingly, loss of the Lancefield Group B Carbohydrate (GBC) rendered GBS significantly more resistant to hGIIA-mediated killing. This suggests that the streptococcal Lancefield antigens, which are critical determinants for streptococcal physiology and virulence, are required for the bactericidal enzyme hGIIA to exert its bactericidal function.
Collapse
Affiliation(s)
- Vincent P. van Hensbergen
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elin Movert
- Department of Experimental Medical Science, Section for Immunology, Lund University, Lund, Sweden
| | - Vincent de Maat
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Yoann Le Breton
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, MD, United States of America
| | - Gérard Lambeau
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Department of Biochemistry, Valbonne, France
| | - Christine Payré
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Department of Biochemistry, Valbonne, France
| | - Anna Henningham
- Department of Pediatrics and Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Jos A. G. van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Britta Brügger
- Heidelberg University, Biochemistry Center (BZH), Heidelberg, Germany
| | - Fredric Carlsson
- Department of Experimental Medical Science, Section for Immunology, Lund University, Lund, Sweden
- Department of Biology, Section for Molecular Cell Biology, Lund University, Lund, Sweden
| | - Kevin S. McIver
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, MD, United States of America
| | - Nina M. van Sorge
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
20
|
Dore E, Boilard E. Roles of secreted phospholipase A 2 group IIA in inflammation and host defense. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:789-802. [PMID: 30905346 DOI: 10.1016/j.bbalip.2018.08.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 01/08/2023]
Abstract
Among all members of the secreted phospholipase A2 (sPLA2) family, group IIA sPLA2 (sPLA2-IIA) is possibly the most studied enzyme. Since its discovery, many names have been associated with sPLA2-IIA, such as "non-pancreatic", "synovial", "platelet-type", "inflammatory", and "bactericidal" sPLA2. Whereas the different designations indicate comprehensive functions or sources proposed for this enzyme, the identification of the precise roles of sPLA2-IIA has remained a challenge. This can be attributed to: the expression of the enzyme by various cells of different lineages, its limited activity towards the membranes of immune cells despite its expression following common inflammatory stimuli, its ability to interact with certain proteins independently of its catalytic activity, and its absence from multiple commonly used mouse models. Nevertheless, elevated levels of the enzyme during inflammatory processes and associated consistent release of arachidonic acid from the membrane of extracellular vesicles suggest that sPLA2-IIA may contribute to inflammation by using endogenous substrates in the extracellular milieu. Moreover, the remarkable potency of sPLA2-IIA towards bacterial membranes and its induced expression during the course of infections point to a role for this enzyme in the defense of the host against invading pathogens. In this review, we present current knowledge related to mammalian sPLA2-IIA and its roles in sterile inflammation and host defense.
Collapse
Affiliation(s)
- Etienne Dore
- Centre de Recherche du CHU de Québec, Université Laval, Department of Infectious Diseases and Immunity, Québec City, QC, Canada
| | - Eric Boilard
- Centre de Recherche du CHU de Québec, Université Laval, Department of Infectious Diseases and Immunity, Québec City, QC, Canada; Canadian National Transplantation Research Program, Edmonton, AB, Canada.
| |
Collapse
|
21
|
Loffredo S, Ferrara AL, Bova M, Borriello F, Suffritti C, Veszeli N, Petraroli A, Galdiero MR, Varricchi G, Granata F, Zanichelli A, Farkas H, Cicardi M, Lambeau G, Marone G. Secreted Phospholipases A 2 in Hereditary Angioedema With C1-Inhibitor Deficiency. Front Immunol 2018; 9:1721. [PMID: 30083168 PMCID: PMC6064723 DOI: 10.3389/fimmu.2018.01721] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/12/2018] [Indexed: 12/21/2022] Open
Abstract
Background Hereditary angioedema (HAE) caused by deficiency (type I) or dysfunction (type II) of the C1 inhibitor protein (C1-INH-HAE) is a disabling, potentially fatal condition characterized by recurrent episodes of swelling. We have recently found that patients with C1-INH-HAE have increased plasma levels of vascular endothelial growth factors and angiopoietins (Angs), which have been associated with vascular permeability in several diseases. Among these and other factors, blood endothelial cells and vascular permeability can be modulated by extracellular or secreted phospholipases A2 (sPLA2s). Objective We sought to investigate the enzymatic activity and biological functions of sPLA2 in patients with C1-INH-HAE. Methods sPLA2s enzymatic activity was evaluated in the plasma from 109 adult patients with C1-INH-HAE and 68 healthy donors in symptom-free period and attacks. Plasma level of group IIA sPLA2 (hGIIA) protein was measured in selected samples. The effect of C1-INH-HAE plasma on endothelial permeability was examined in vitro using a vascular permeability assay. The role of hGIIA was determined using highly specific sPLA2 indole inhibitors. The effect of recombinant hGIIA on C1-INH activity was examined in vitro by functional assay. Results Plasma sPLA2 activity and hGIIA levels are increased in symptom-free C1-INH-HAE patients compared with controls. sPLA2 activity negatively correlates with C1-INH protein level and function. C1-INH-HAE plasma increases endothelial permeability in vitro, and this effect is partially reverted by a specific hGIIA enzymatic inhibitor. Finally, recombinant hGIIA inhibits C1-INH activity in vitro. Conclusion sPLA2 enzymatic activity (likely attributable to hGIIA), which is increased in C1-INH-HAE patients, can promote vascular permeability and impairs C1-INH activity. Our results may pave the way for investigating the functions of sPLA2s (in particular, hGIIA) in the pathophysiology of C1-INH-HAE and may inform the development of new therapeutic targets.
Collapse
Affiliation(s)
- Stefania Loffredo
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, Naples, Italy
| | - Maria Bova
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, Naples, Italy
| | - Francesco Borriello
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, Naples, Italy.,Division of Gastroenterology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Chiara Suffritti
- Department of Biomedical and Clinical Sciences, University of Milan, Luigi Sacco Hospital Milan, Milan, Italy
| | - Nóra Veszeli
- Hungarian Angioedema Center, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Angelica Petraroli
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, Naples, Italy
| | - Francescopaolo Granata
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, Naples, Italy
| | - Andrea Zanichelli
- Department of Biomedical and Clinical Sciences, University of Milan, Luigi Sacco Hospital Milan, Milan, Italy
| | - Henriette Farkas
- Hungarian Angioedema Center, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Marco Cicardi
- Department of Biomedical and Clinical Sciences, University of Milan, Luigi Sacco Hospital Milan, Milan, Italy
| | - Gérard Lambeau
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne Sophia Antipolis, France
| | - Gianni Marone
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| |
Collapse
|
22
|
Prebiotic Oligosaccharides Potentiate Host Protective Responses against L. Monocytogenes Infection. Pathogens 2017; 6:pathogens6040068. [PMID: 29257110 PMCID: PMC5750592 DOI: 10.3390/pathogens6040068] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/05/2017] [Accepted: 12/15/2017] [Indexed: 12/22/2022] Open
Abstract
Prebiotic oligosaccharides are used to modulate enteric pathogens and reduce pathogen shedding. The interactions with prebiotics that alter Listeria monocytogenes infection are not yet clearly delineated. L. monocytogenes cellular invasion requires a concerted manipulation of host epithelial cell membrane receptors to initiate internalization and infection often via receptor glycosylation. Bacterial interactions with host glycans are intimately involved in modulating cellular responses through signaling cascades at the membrane and in intracellular compartments. Characterizing the mechanisms underpinning these modulations is essential for predictive use of dietary prebiotics to diminish pathogen association. We demonstrated that human milk oligosaccharide (HMO) pretreatment of colonic epithelial cells (Caco-2) led to a 50% decrease in Listeria association, while Biomos pretreatment increased host association by 150%. L. monocytogenes-induced gene expression changes due to oligosaccharide pretreatment revealed global alterations in host signaling pathways that resulted in differential subcellular localization of L. monocytogenes during early infection. Ultimately, HMO pretreatment led to bacterial clearance in Caco-2 cells via induction of the unfolded protein response and eIF2 signaling, while Biomos pretreatment resulted in the induction of host autophagy and L. monocytogenes vacuolar escape earlier in the infection progression. This study demonstrates the capacity of prebiotic oligosaccharides to minimize infection through induction of host-intrinsic protective responses.
Collapse
|
23
|
Structure of Human M-type Phospholipase A2 Receptor Revealed by Cryo-Electron Microscopy. J Mol Biol 2017; 429:3825-3835. [DOI: 10.1016/j.jmb.2017.10.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/17/2017] [Accepted: 10/17/2017] [Indexed: 01/29/2023]
|
24
|
Vulfius CA, Kasheverov IE, Kryukova EV, Spirova EN, Shelukhina IV, Starkov VG, Andreeva TV, Faure G, Zouridakis M, Tsetlin VI, Utkin YN. Pancreatic and snake venom presynaptically active phospholipases A2 inhibit nicotinic acetylcholine receptors. PLoS One 2017; 12:e0186206. [PMID: 29023569 PMCID: PMC5638340 DOI: 10.1371/journal.pone.0186206] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/27/2017] [Indexed: 01/08/2023] Open
Abstract
Phospholipases A2 (PLA2s) are enzymes found throughout the animal kingdom. They hydrolyze phospholipids in the sn-2 position producing lysophospholipids and unsaturated fatty acids, agents that can damage membranes. PLA2s from snake venoms have numerous toxic effects, not all of which can be explained by phospholipid hydrolysis, and each enzyme has a specific effect. We have earlier demonstrated the capability of several snake venom PLA2s with different enzymatic, cytotoxic, anticoagulant and antiproliferative properties, to decrease acetylcholine-induced currents in Lymnaea stagnalis neurons, and to compete with α-bungarotoxin for binding to nicotinic acetylcholine receptors (nAChRs) and acetylcholine binding protein. Since nAChRs are implicated in postsynaptic and presynaptic activities, in this work we probe those PLA2s known to have strong presynaptic effects, namely β-bungarotoxin from Bungarus multicinctus and crotoxin from Crotalus durissus terrificus. We also wished to explore whether mammalian PLA2s interact with nAChRs, and have examined non-toxic PLA2 from porcine pancreas. It was found that porcine pancreatic PLA2 and presynaptic β-bungarotoxin blocked currents mediated by nAChRs in Lymnaea neurons with IC50s of 2.5 and 4.8 μM, respectively. Crotoxin competed with radioactive α-bungarotoxin for binding to Torpedo and human α7 nAChRs and to the acetylcholine binding protein. Pancreatic PLA2 interacted similarly with these targets; moreover, it inhibited radioactive α-bungarotoxin binding to the water-soluble extracellular domain of human α9 nAChR, and blocked acetylcholine induced currents in human α9α10 nAChRs heterologously expressed in Xenopus oocytes. These and our earlier results show that all snake PLA2s, including presynaptically active crotoxin and β-bungarotoxin, as well as mammalian pancreatic PLA2, interact with nAChRs. The data obtained suggest that this interaction may be a general property of all PLA2s, which should be proved by further experiments.
Collapse
Affiliation(s)
- Catherine A. Vulfius
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Igor E. Kasheverov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Elena V. Kryukova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina N. Spirova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Irina V. Shelukhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vladislav G. Starkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Tatyana V. Andreeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Grazyna Faure
- Unité Récepteurs-Canaux, Institut Pasteur, Paris, France
| | | | - Victor I. Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Yuri N. Utkin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- * E-mail:
| |
Collapse
|
25
|
|
26
|
Loffredo S, Borriello F, Iannone R, Ferrara AL, Galdiero MR, Gigantino V, Esposito P, Varricchi G, Lambeau G, Cassatella MA, Granata F, Marone G. Group V Secreted Phospholipase A 2 Induces the Release of Proangiogenic and Antiangiogenic Factors by Human Neutrophils. Front Immunol 2017; 8:443. [PMID: 28458672 PMCID: PMC5394767 DOI: 10.3389/fimmu.2017.00443] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/30/2017] [Indexed: 02/05/2023] Open
Abstract
Secreted phospholipases A2 (sPLA2s) are extracellular enzymes that catalyze the release of free fatty acids and lysophospholipids from membrane phospholipids and also bind to different receptors (e.g., PLA2R1 or integrins). To date, 12 mammalian sPLA2s have been identified, which play a critical role in pathophysiological processes including inflammation and cancer. sPLA2s activate immune cells such as human neutrophils (PMNs) by enzymatic activity- or receptor-mediated mechanisms. In addition, human PMNs synthesize and store human group V (hGV) and human group X (hGX) sPLA2s in their granules, but only the former is released upon cellular activation. We investigated the effects of sPLA2s on the release of proangiogenic and antiangiogenic factors by PMNs. We found that exogenous hGV and hGX sPLA2s induce the release of vascular endothelial growth factor (VEGF)-A, angiopoietin 1 (Ang1), and CXCL8/IL-8. Only hGV induces the secretion of the antiangiogenic isoform of VEGF-A, namely, VEGF-A165b. While the release of VEGF-A, Ang1, and CXCL8/IL-8 was likely mediated by hGV enzymatic activity and/or binding to PLA2R1 and heparan sulfate proteoglycans, the release of VEGF-A165b requires the interaction with αVβ3 and α4β1 integrins. We also provide evidence that endogenous hGV released by N-formyl-met-leu-phe (fMLF)-activated PMNs is involved in the release of angiogenic factors. The translational relevance of these data is supported by our findings that hGV expression is increased in human samples of lung cancer which are infiltrated by PMNs. Overall, our results suggest that the hGV-neutrophil axis may play a relevant role in the modulation of cancer-related inflammation and angiogenesis.
Collapse
Affiliation(s)
- Stefania Loffredo
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- *Correspondence: Stefania Loffredo, ; Gianni Marone,
| | - Francesco Borriello
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- Division of Infectious Diseases, Department of Medicine, Boston Children’s Hospital, Boston, MA, USA
| | - Raffaella Iannone
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Anne L. Ferrara
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Maria R. Galdiero
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”, Naples, Italy
| | - Pasquale Esposito
- U.O.C. Immunohematology and Transfusion Medicine, University of Naples Federico II, Naples, Italy
| | - Gilda Varricchi
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Gerard Lambeau
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne Sophia Antipolis, France
| | - Marco A. Cassatella
- Department of Medicine, Division of General Pathology, University of Verona, Verona, Italy
| | - Francescopaolo Granata
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Gianni Marone
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- CNR Institute of Experimental Endocrinology and Oncology “G. Salvatore”, Naples, Italy
- *Correspondence: Stefania Loffredo, ; Gianni Marone,
| |
Collapse
|
27
|
Inhibition of Human Group IIA-Secreted Phospholipase A2 and THP-1 Monocyte Recruitment by Maslinic Acid. Lipids 2016; 51:1153-1159. [PMID: 27540737 DOI: 10.1007/s11745-016-4186-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/08/2016] [Indexed: 10/21/2022]
Abstract
Maslinic acid is a natural pentacyclic triterpenoid which has anti-inflammatory properties. A recent study showed that secretory phospholipase A2 (sPLA2) may be a potential binding target of maslinic acid. The human group IIA (hGIIA)-sPLA2 is found in human sera and their levels are correlated with severity of inflammation. This study aims to determine whether maslinic acid interacts with hGIIA-sPLA2 and inhibits inflammatory response induced by this enzyme. It is shown that maslinic acid enhanced intrinsic fluorescence of hGIIA-sPLA2 and inhibited its enzyme activity in a concentration-dependent manner. Molecular docking revealed that maslinic acid binds to calcium binding and interfacial phospholipid binding site, suggesting that it inhibit access of catalytic calcium ion for enzymatic reaction and block binding of the enzyme to membrane phospholipid. The hGIIA-sPLA2 enzyme is also responsible in mediating monocyte recruitment and differentiation. Results showed that maslinic acid inhibit hGIIA-sPLA2-induced THP-1 cell differentiation and migration, and the effect observed is specific to hGIIA-sPLA2 as cells treated with maslinic acid alone did not significantly affect the number of adherent and migrated cells. Considering that hGIIA-sPLA2 enzyme is known to hydrolyze glyceroacylphospholipids present in lipoproteins and cell membranes, maslinic acid may bind and inhibit hGIIA-sPLA2 enzymatic activity, thereby reduces the release of fatty acids and lysophospholipids which stimulates monocyte migration and differentiation. This study is the first to report on the molecular interaction between maslinic acid and inflammatory target hGIIA-sPLA2 as well as its effect towards hGIIA-sPLA2-induced THP-1 monocyte adhesive and migratory capabilities, an important immune-inflammation process in atherosclerosis.
Collapse
|
28
|
Almeida J, Resende L, Silva A, Ribeiro R, Stábeli R, Soares A, Calderon L, Marangoni S, Da Silva S. Biochemical and functional studies of ColTx-I, a new myotoxic phospholipase A2 isolated from Crotalus oreganus lutosus (Great Basin rattlesnake) snake venom. Toxicon 2016; 117:1-12. [DOI: 10.1016/j.toxicon.2016.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/05/2016] [Accepted: 03/15/2016] [Indexed: 01/13/2023]
|
29
|
Murakami M, Yamamoto K, Miki Y, Murase R, Sato H, Taketomi Y. The Roles of the Secreted Phospholipase A 2 Gene Family in Immunology. Adv Immunol 2016; 132:91-134. [PMID: 27769509 PMCID: PMC7112020 DOI: 10.1016/bs.ai.2016.05.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Within the phospholipase A2 (PLA2) family that hydrolyzes phospholipids to yield fatty acids and lysophospholipids, secreted PLA2 (sPLA2) enzymes comprise the largest group containing 11 isoforms in mammals. Individual sPLA2s exhibit unique tissue or cellular distributions and enzymatic properties, suggesting their distinct biological roles. Although PLA2 enzymes, particularly cytosolic PLA2 (cPLA2α), have long been implicated in inflammation by driving arachidonic acid metabolism, the precise biological roles of sPLA2s have remained a mystery over the last few decades. Recent studies employing mice gene-manipulated for individual sPLA2s, in combination with mass spectrometric lipidomics to identify their target substrates and products in vivo, have revealed their roles in diverse biological events, including immunity and associated disorders, through lipid mediator-dependent or -independent processes in given microenvironments. In this review, we summarize our current knowledge of the roles of sPLA2s in various immune responses and associated diseases.
Collapse
Affiliation(s)
- M Murakami
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan.
| | - K Yamamoto
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Y Miki
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - R Murase
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - H Sato
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Y Taketomi
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
30
|
Vasilakaki S, Barbayianni E, Magrioti V, Pastukhov O, Constantinou-Kokotou V, Huwiler A, Kokotos G. Inhibitors of secreted phospholipase A2 suppress the release of PGE2 in renal mesangial cells. Bioorg Med Chem 2016; 24:3029-3034. [PMID: 27234891 DOI: 10.1016/j.bmc.2016.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/11/2016] [Indexed: 01/15/2023]
Abstract
The upregulation of PGE2 by mesangial cells has been observed under chronic inflammation condition. In the present work, renal mesangial cells were stimulated to trigger a huge increase of PGE2 synthesis and were treated in the absence or presence of known PLA2 inhibitors. A variety of synthetic inhibitors, mainly developed in our labs, which are known to selectively inhibit each of GIVA cPLA2, GVIA iPLA2, and GIIA/GV sPLA2, were used as tools in this study. Synthetic sPLA2 inhibitors, such as GK115 (an amide derivative based on the non-natural amino acid (R)-γ-norleucine) as well as GK126 and GK241 (2-oxoamides based on the natural (S)-α-amino acid leucine and valine, respectively) presented an interesting effect on the suppression of PGE2 formation.
Collapse
Affiliation(s)
- Sofia Vasilakaki
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Efrosini Barbayianni
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Victoria Magrioti
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | | | | | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Bern 3010, Switzerland
| | - George Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| |
Collapse
|
31
|
Vasilakaki S, Barbayianni E, Leonis G, Papadopoulos MG, Mavromoustakos T, Gelb MH, Kokotos G. Development of a potent 2-oxoamide inhibitor of secreted phospholipase A2 guided by molecular docking calculations and molecular dynamics simulations. Bioorg Med Chem 2016; 24:1683-95. [PMID: 26970660 DOI: 10.1016/j.bmc.2016.02.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 02/24/2016] [Accepted: 02/27/2016] [Indexed: 10/22/2022]
Abstract
Inhibition of group IIA secreted phospholipase A2 (GIIA sPLA2) has been an important objective for medicinal chemists. We have previously shown that inhibitors incorporating the 2-oxoamide functionality may inhibit human and mouse GIIA sPLA2s. Herein, the development of new potent inhibitors by molecular docking calculations using the structure of the known inhibitor 7 as scaffold, are described. Synthesis and biological evaluation of the new compounds revealed that the long chain 2-oxoamide based on (S)-valine GK241 led to improved activity (IC50=143 nM and 68 nM against human and mouse GIIA sPLA2, respectively). In addition, molecular dynamics simulations were employed to shed light on GK241 potent and selective inhibitory activity.
Collapse
Affiliation(s)
- Sofia Vasilakaki
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Efrosini Barbayianni
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Georgios Leonis
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vas. Constantinou Avenue, Athens 11635, Greece
| | - Manthos G Papadopoulos
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vas. Constantinou Avenue, Athens 11635, Greece
| | - Thomas Mavromoustakos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Michael H Gelb
- Department of Chemistry and Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - George Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| |
Collapse
|
32
|
Yamaguchi M, Zacharia J, Laidlaw TM, Balestrieri B. PLA2G5 regulates transglutaminase activity of human IL-4-activated M2 macrophages through PGE2 generation. J Leukoc Biol 2016; 100:131-41. [PMID: 26936936 DOI: 10.1189/jlb.3a0815-372r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 02/12/2016] [Indexed: 12/22/2022] Open
Abstract
Phospholipases A2 are enzymes that liberate membrane-bound lipids in a tissue and cell-specific fashion. Group V secretory phospholipase A2 is necessary for the development of M2 macrophages and their effector functions in a mouse model of the T-helper-2 allergic airway inflammation. However, the function of group V phospholipase A2 in human M2 activation and T-helper-2 inflammation is ill-defined. Transglutaminase-2, a protein cross-linking enzyme, is a newly identified marker of both human and mouse interleukin-4-activated M2 macrophages and is also found in the lungs of patients with asthma. We report that group V phospholipase A2 and transglutaminase-2 colocalized in macrophages of human nasal polyp tissue obtained from patients with T-helper-2 eosinophilic inflammation, and their coexpression positively correlated with the number of eosinophils in each tissue specimen. We demonstrate that in human monocyte-derived macrophages activated by interleukin-4, group V phospholipase A2 translocated and colocalized with transglutaminase-2 in the cytoplasm and on the membrane of macrophages. Moreover, knocking down group V phospholipase A2 with small interfering ribonucleic acid reduced macrophage transglutaminase activity, whereas mass spectrometry analysis of lipids also showed reduced prostaglandin E2 production. Finally, exogenous prostaglandin E2 restored transglutaminase activity of group V phospholipase A2-small interfering ribonucleic acid-treated macrophages. Thus, our study shows a novel function of group V phospholipase A2 in regulating the transglutaminase activity of human interleukin-4-activated M2 macrophages through prostaglandin E2 generation and suggests that group V phospholipase A2 is a functionally relevant enzyme that may have therapeutic value for the treatment of human T-helper-2 inflammatory disorders.
Collapse
Affiliation(s)
- Munehiro Yamaguchi
- Department of Medicine, Harvard Medical School, Boston Massachusetts, USA; and the Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jennifer Zacharia
- Department of Medicine, Harvard Medical School, Boston Massachusetts, USA; and the Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Tanya M Laidlaw
- Department of Medicine, Harvard Medical School, Boston Massachusetts, USA; and the Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Barbara Balestrieri
- Department of Medicine, Harvard Medical School, Boston Massachusetts, USA; and the Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Menschikowski M, Hagelgans A, Nacke B, Jandeck C, Mareninova OA, Asatryan L, Siegert G. Epigenetic control of group V phospholipase A2 expression in human malignant cells. Tumour Biol 2015; 37:8097-105. [PMID: 26715269 DOI: 10.1007/s13277-015-4670-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/16/2015] [Indexed: 12/27/2022] Open
Abstract
Secreted phospholipases A2 (sPLA2) are suggested to play an important role in inflammation and tumorigenesis. Different mechanisms of epigenetic regulation are involved in the control of group IIA, III and X sPLA2s expression in cancer cells, but group V sPLA2 (GV-PLA2) in this respect has not been studied. Here, we demonstrate the role of epigenetic mechanisms in regulation of GV-PLA2 expression in different cell lines originating from leukaemia and solid cancers. In blood leukocytes from leukaemic patients, levels of GV-PLA2 transcripts were significantly lower in comparison to those from healthy individuals. Similarly, in DU-145 and PC-3 prostate and CAL-51 and MCF-7 mammary cancer cell lines, levels of GV-PLA2 transcripts were significantly lower in relation to those found in normal epithelial cells of prostate or mammary. By sequencing and methylation-specific high-resolution melting (MS-HRM) analyses of bisulphite-modified DNA, distinct CpG sites in the GV-PLA2 promoter region were identified that were differentially methylated in cancer cells in comparison to normal epithelial and endothelial cells. Spearman rank order analysis revealed a significant negative correlation between the methylation degree and the cellular expression of GV-PLA2 (r = -0.697; p = 0.01). The effects of demethylating agent (5-aza-2'-deoxycytidine) and histone deacetylase inhibitor (trichostatin A) on GV-PLA2 transcription in the analysed cells confirmed the importance of DNA methylation and histone modification in the regulation of the GV-PLA2 gene expression in leukaemic, prostate and mammary cancer cell lines. The exposure of tumour cells to human recombinant GV-PLA2 resulted in a reduced colony forming activity of MCF-7, HepG2 and PC-3 cells, but not of DU-145 cells suggesting a cell-type-dependent effect of GV-PLA2 on cell growth. In conclusion, our results suggest that epigenetic mechanisms such as DNA methylation and histone modification play an important role in downregulation of GV-PLA2 expression in cancer cells.
Collapse
Affiliation(s)
- Mario Menschikowski
- Institute of Clinical Chemistry and Laboratory Medicine, Carl Gustav Carus University Hospital, Technical University of Dresden, Fetscherstr. 74, D-01307, Dresden, Germany.
| | - Albert Hagelgans
- Institute of Clinical Chemistry and Laboratory Medicine, Carl Gustav Carus University Hospital, Technical University of Dresden, Fetscherstr. 74, D-01307, Dresden, Germany
| | - Brit Nacke
- Institute of Clinical Chemistry and Laboratory Medicine, Carl Gustav Carus University Hospital, Technical University of Dresden, Fetscherstr. 74, D-01307, Dresden, Germany
| | - Carsten Jandeck
- Institute of Clinical Chemistry and Laboratory Medicine, Carl Gustav Carus University Hospital, Technical University of Dresden, Fetscherstr. 74, D-01307, Dresden, Germany
| | - Olga A Mareninova
- Veterans Affairs Greater Los Angeles Healthcare System and University of California at Los Angeles, Los Angeles, CA, USA
| | - Liana Asatryan
- Titus Family Department of Clinical Pharmacy, USC School of Pharmacy, Los Angeles, CA, USA
| | - Gabriele Siegert
- Institute of Clinical Chemistry and Laboratory Medicine, Carl Gustav Carus University Hospital, Technical University of Dresden, Fetscherstr. 74, D-01307, Dresden, Germany
| |
Collapse
|
34
|
Abi Nahed R, Martinez G, Escoffier J, Yassine S, Karaouzène T, Hograindleur JP, Turk J, Kokotos G, Ray PF, Bottari S, Lambeau G, Hennebicq S, Arnoult C. Progesterone-induced Acrosome Exocytosis Requires Sequential Involvement of Calcium-independent Phospholipase A2β (iPLA2β) and Group X Secreted Phospholipase A2 (sPLA2). J Biol Chem 2015; 291:3076-89. [PMID: 26655718 DOI: 10.1074/jbc.m115.677799] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Indexed: 11/06/2022] Open
Abstract
Phospholipase A2 (PLA2) activity has been shown to be involved in the sperm acrosome reaction (AR), but the molecular identity of PLA2 isoforms has remained elusive. Here, we have tested the role of two intracellular (iPLA2β and cytosolic PLA2α) and one secreted (group X) PLA2s in spontaneous and progesterone (P4)-induced AR by using a set of specific inhibitors and knock-out mice. iPLA2β is critical for spontaneous AR, whereas both iPLA2β and group X secreted PLA2 are involved in P4-induced AR. Cytosolic PLA2α is dispensable in both types of AR. P4-induced AR spreads over 30 min in the mouse, and kinetic analyses suggest the presence of different sperm subpopulations, using distinct PLA2 pathways to achieve AR. At low P4 concentration (2 μm), sperm undergoing early AR (0-5 min post-P4) rely on iPLA2β, whereas sperm undergoing late AR (20-30 min post-P4) rely on group X secreted PLA2. Moreover, the role of PLA2s in AR depends on P4 concentration, with the PLA2s being key actors at low physiological P4 concentrations (≤2 μm) but not at higher P4 concentrations (~10 μm).
Collapse
Affiliation(s)
- Roland Abi Nahed
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - Guillaume Martinez
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - Jessica Escoffier
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - Sandra Yassine
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - Thomas Karaouzène
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - Jean-Pascal Hograindleur
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - John Turk
- the Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri 63110
| | - George Kokotos
- the Department of Chemistry, University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Pierre F Ray
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France, the Centre Hospitalier Universitaire de Grenoble, Unité Fonctionnelle de Biochimie et Génétique Moléculaire, Grenoble F-38000, France
| | - Serge Bottari
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France, the Centre Hospitalier Universitaire de Grenoble, Plate-forme de Radioanalyse, IBP, CS10217, Grenoble F-38000, France
| | - Gérard Lambeau
- the Université de Nice-Sophia Antipolis, Valbonne 06560, France, the Centre Hospitalier Universitaire de Grenoble, Centre d'AMP-CECOS, CS1021, Grenoble F-38000, France
| | - Sylviane Hennebicq
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France, the Centre Hospitalier Universitaire de Grenoble, Centre d'AMP-CECOS, CS1021, Grenoble F-38000, France
| | - Christophe Arnoult
- From the Université Grenoble Alpes, F-38000 Grenoble, France, the Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France,
| |
Collapse
|
35
|
Abstract
PURPOSE To evaluate the association of phospholipase A2, Group V (PLA2G5), with benign familial fleck retina in a consanguineous family with triplets. METHODS Clinical eye examination, including fundus examination and spectral domain optical coherence tomography, was performed for all the family members. After blood sample collection and DNA extraction, polymerase chain reaction was performed to amplify regions spanning Exons 2, 3, 4, and 5 of PLA2G5. The amplified products were sequenced to observe the presence of any mutations. RESULTS Fundus examination in two of the triplets revealed discrete yellow-white flecks and both had good vision and absence of night blindness, consistent with benign familial fleck retina. The flecks were hyperautofluorescent. Furthermore, spectral domain optical coherence tomography showed focal thickening of the retinal pigment epithelium because of the presence of these flecks. Molecular investigations showed that PLA2G5 Exons 2, 4, and 5 harbored no misalignments among all family members. However, PLA2G5 Exon 3 showed a p.Gly45Cys mutation for the father and the third triplet who was affected. CONCLUSION The clinical findings in this family suggest a diagnosis of benign familial fleck retina with excellent prognosis, in which the PLA2G5 gene may play a role.
Collapse
|
36
|
Tanabe T, Shimokawaji T, Kanoh S, Rubin BK. Secretory phospholipases A2 are secreted from ciliated cells and increase mucin and eicosanoid secretion from goblet cells. Chest 2015; 147:1599-1609. [PMID: 25429648 DOI: 10.1378/chest.14-0258] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Secretory phospholipases A2 (sPLA2) initiate the biosynthesis of eicosanoids, are increased in the airways of people with severe asthma, and induce mucin hypersecretion. We used IL-13-transformed, highly enriched goblet cells and differentiated (ciliary cell-enriched) human bronchial epithelial cell culture to evaluate the relative contribution of ciliated and goblet cells to airway sPLA2 generation and response. We wished to determine the primary source(s) of sPLA2 and leukotrienes in human airway epithelial cells. METHODS Human bronchial epithelial cells from subjects without lung disease were differentiated to a ciliated-enriched or goblet-enriched cell phenotype. Synthesis of sPLA2, cysteinyl leukotrienes (cysLTs), and airway mucin messenger RNA and protein was measured by real-time-polymerase chain reaction and an enzyme-linked immunosorbent assay, and the localization of mucin and sPLA2 to specific cells types was confirmed by confocal microscopy. RESULTS sPLA2 group IIa, V, and X messenger RNA expression was increased in ciliated-enriched cells (P < .001) but not in goblet-enriched cells. sPLA2 were secreted from the apical (air) side of ciliated-enriched cells but not goblet-enriched cells (P < .001). Immunostaining of sPLA2 V was strongly positive in ciliated-enriched cells but not in goblet-enriched cells. sPLA2 released cysLTs from goblet-enriched cells but not from ciliated-enriched cells, and this result was greatest with sPLA2 V (P < .05). sPLA2 V increased goblet-enriched cell mucin secretion, which was inhibited by inhibitors of lipoxygenase or cyclooxygenase (P < .02). CONCLUSIONS sPLA2 are secreted from ciliated cells and appear to induce mucin and cysLT secretion from goblet cells, strongly suggesting that airway goblet cells are proinflammatory effector cells.
Collapse
Affiliation(s)
- Tsuyoshi Tanabe
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA.
| | - Tadasuke Shimokawaji
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Soichiro Kanoh
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Bruce K Rubin
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA
| |
Collapse
|
37
|
Nakamura H, Wakita S, Yasufuku K, Makiyama T, Waraya M, Hashimoto N, Murayama T. Sphingomyelin Regulates the Activity of Secretory Phospholipase A2in the Plasma Membrane. J Cell Biochem 2015; 116:1898-907. [DOI: 10.1002/jcb.25145] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 02/20/2015] [Indexed: 12/26/2022]
Affiliation(s)
- Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology; Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| | - Shigeo Wakita
- Laboratory of Chemical Pharmacology; Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| | - Kana Yasufuku
- Laboratory of Chemical Pharmacology; Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| | - Tomohiko Makiyama
- Laboratory of Chemical Pharmacology; Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| | - Misa Waraya
- Laboratory of Chemical Pharmacology; Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| | - Naohiro Hashimoto
- Laboratory of Chemical Pharmacology; Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology; Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| |
Collapse
|
38
|
Xue YL, Miyakawa T, Nakamura A, Hatano KI, Sawano Y, Tanokura M. Yam Tuber Storage Protein Reduces Plant Oxidants Using the Coupled Reactions as Carbonic Anhydrase and Dehydroascorbate Reductase. MOLECULAR PLANT 2015; 8:1115-8. [PMID: 25747844 DOI: 10.1016/j.molp.2015.02.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 02/13/2015] [Accepted: 02/27/2015] [Indexed: 05/13/2023]
Affiliation(s)
- You-Lin Xue
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; College of Light Industry, Liaoning University, 66 Chongshan Middle Road, Shenyang 110036, China
| | - Takuya Miyakawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Akira Nakamura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ken-Ichi Hatano
- Division of Molecular and Science, Faculty of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Yoriko Sawano
- Laboratory of Chemistry, College of Liberal Arts and Sciences, Tokyo Medical and Dental University, 2-8-30 Kounodai, Ichikawa-shi, Chiba 272-0827, Japan
| | - Masaru Tanokura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| |
Collapse
|
39
|
Lipidome and transcriptome profiling of pneumolysin intoxication identifies networks involved in statin-conferred protection of airway epithelial cells. Sci Rep 2015; 5:10624. [PMID: 26023727 PMCID: PMC4448502 DOI: 10.1038/srep10624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/22/2015] [Indexed: 12/12/2022] Open
Abstract
Pneumonia remains one of the leading causes of death in both adults and children worldwide. Despite the adoption of a wide variety of therapeutics, the mortality from community-acquired pneumonia has remained relatively constant. Although viral and fungal acute airway infections can result in pneumonia, bacteria are the most common cause of community-acquired pneumonia, with Streptococcus pneumoniae isolated in nearly 50% of cases. Pneumolysin is a cholesterol-dependent cytolysin or pore-forming toxin produced by Streptococcus pneumonia and has been shown to play a critical role in bacterial pathogenesis. Airway epithelium is the initial site of many bacterial contacts and its barrier and mucosal immunity functions are central to infectious lung diseases. In our studies, we have shown that the prior exposure to statins confers significant resistance of airway epithelial cells to the cytotoxicity of pneumolysin. We decided to take this study one step further, assessing changes in both the transcriptome and lipidome of human airway epithelial cells exposed to toxin, statin or both. Our current work provides the first global view in human airway epithelial cells of both the transcriptome and the lipid interactions that result in cellular protection from pneumolysin.
Collapse
|
40
|
Such GK, Yan Y, Johnston APR, Gunawan ST, Caruso F. Interfacing materials science and biology for drug carrier design. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:2278-2297. [PMID: 25728711 DOI: 10.1002/adma.201405084] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/11/2014] [Indexed: 06/04/2023]
Abstract
Over the last ten years, there has been considerable research interest in the development of polymeric carriers for biomedicine. Such delivery systems have the potential to significantly reduce side effects and increase the bioavailability of poorly soluble therapeutics. The design of carriers has relied on harnessing specific variations in biological conditions, such as pH or redox potential, and more recently, by incorporating specific peptide cleavage sites for enzymatic hydrolysis. Although much progress has been made in this field, the specificity of polymeric carriers is still limited when compared with their biological counterparts. To synthesize the next generation of carriers, it is important to consider the biological rationale for materials design. This requires a detailed understanding of the cellular microenvironments and how these can be harnessed for specific applications. In this review, several important physiological cues in the cellular microenvironments are outlined, with a focus on changes in pH, redox potential, and the types of enzymes present in specific regions. Furthermore, recent studies that use such biologically inspired triggers to design polymeric carriers are highlighted, focusing on applications in the field of therapeutic delivery.
Collapse
Affiliation(s)
- Georgina K Such
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | | | | | | |
Collapse
|
41
|
Oberčkal J, Kovačič L, Šribar J, Leonardi A, Dolinar K, Pucer Janež A, Križaj I. On the role of protein disulfide isomerase in the retrograde cell transport of secreted phospholipases A2. PLoS One 2015; 10:e0120692. [PMID: 25763817 PMCID: PMC4357439 DOI: 10.1371/journal.pone.0120692] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/25/2015] [Indexed: 11/29/2022] Open
Abstract
Following the finding that ammodytoxin (Atx), a neurotoxic secreted phospholipase A2 (sPLA2) in snake venom, binds specifically to protein disulfide isomerase (PDI) in vitro we show that these proteins also interact in living rat PC12 cells that are able to internalize this group IIA (GIIA) sPLA2. Atx and PDI co-localize in both differentiated and non-differentiated PC12 cells, as shown by fluorescence microscopy. Based on a model of the complex between Atx and yeast PDI (yPDI), a three-dimensional model of the complex between Atx and human PDI (hPDI) was constructed. The Atx binding site on hPDI is situated between domains b and b’. Atx interacts hPDI with an extensive area on its interfacial binding surface. The mammalian GIB, GIIA, GV and GX sPLA2s have the same fold as Atx. The first three sPLA2s have been detected intracellularly but not the last one. The models of their complexes with hPDI were constructed by replacement of Atx with the respective mammalian sPLA2 in the Atx—hPDI complex and molecular docking of the structures. According to the generated models, mammalian GIB, GIIA and GV sPLA2s form complexes with hPDI very similar to that with Atx. The contact area between GX sPLA2 and hPDI is however different from that of the other sPLA2s. Heterologous competition of Atx binding to hPDI with GV and GX sPLA2s confirmed the model-based expectation that GV sPLA2 was a more effective inhibitor than GX sPLA2, thus validating our model. The results suggest a role of hPDI in the (patho)physiology of some snake venom and mammalian sPLA2s by assisting the retrograde transport of these molecules from the cell surface. The sPLA2–hPDI model constitutes a valuable tool to facilitate further insights into this process and into the (patho)physiology of sPLA2s in relation to their action intracellularly.
Collapse
Affiliation(s)
- Jernej Oberčkal
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Lidija Kovačič
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Jernej Šribar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Adrijana Leonardi
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Klemen Dolinar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Anja Pucer Janež
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
42
|
Xu J, Bourgeois H, Vandermeulen E, Vlaeminck B, Meyer E, Demeyere K, Hesta M. Secreted phospholipase A2 inhibitor modulates fatty acid composition and reduces obesity-induced inflammation in Beagle dogs. Vet J 2015; 204:214-9. [PMID: 25920769 DOI: 10.1016/j.tvjl.2015.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 02/07/2023]
Abstract
Secreted phospholipase A2 inhibitor (sPLA2i) has been reported to have an anti-inflammatory function by blocking the production of inflammatory mediators. Obesity is characterized by low-grade inflammation and oxidative stress. The aim of this study was to investigate the effects of dietary supplementation of sPLA2i on inflammation, oxidative stress and serum fatty acid profile in dogs. Seven obese and seven lean Beagle dogs were used in a 28-day double blind cross-over design. Dogs were fed a control diet without supplemental sPLA2i or an sPLA2i supplemented diet. The sPLA2i diet decreased plasma fibrinogen levels and increased the protein:fibrinogen ratio in obese dogs to levels similar to those of lean dogs fed the same diet. Obese dogs had a higher plasma concentration of the lipophilic vitamin A with potential antioxidative capacity and a lower ratio of retinol binding protein 4:vitamin A compared to lean dogs, independent of the diets. A higher proportion of myristic acid (C14:0) and a lower proportion of linoleic acid (C18:2n-6) were observed in the dogs fed with the sPLA2i diet compared to dogs fed with the control diet. Furthermore, a higher ratio of n-6 to n-3, a lower proportion of n-3 polyunsaturated fatty acids and lower omega-3 index were observed in obese compared to lean dogs. The results indicate that obese dogs are characterized by a more 'proinflammatory' serum fatty acid profile and that diet inclusion of sPLA2i may reduce inflammation and alter fatty acid profile.
Collapse
Affiliation(s)
- J Xu
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium
| | - H Bourgeois
- Diana Pet Food, ZA du Gohélis, 56250 Elven, France
| | - E Vandermeulen
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - B Vlaeminck
- Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Proefhoevestraat 10, 9090 Melle, Belgium
| | - E Meyer
- Department of Pharmacology, Biochemistry and Toxicology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - K Demeyere
- Department of Pharmacology, Biochemistry and Toxicology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - M Hesta
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium.
| |
Collapse
|
43
|
Kocbek V, Bersinger NA, Brglez V, Mueller MD, Petan T, Rižner TL. Phospholipase A2 group IIA is elevated in endometriomas but not in peritoneal fluid and serum of ovarian endometriosis patients. Gynecol Endocrinol 2015; 31:214-8. [PMID: 25366587 DOI: 10.3109/09513590.2014.975690] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Our previous gene expression analysis identified phospholipase A2 group IIA (PLA2G2A) as a potential biomarker of ovarian endometriosis. The aim of this study was to evaluate PLA2G2A mRNA and protein levels in tissue samples (endometriomas and normal endometrium) and in serum and peritoneal fluid of ovarian endometriosis patients and control women. One-hundred and sixteen women were included in this study: the case group included 70 ovarian endometriosis patients, and the control group included 38 healthy women and 8 patients with benign ovarian cysts. We observed 41.6-fold greater PLA2G2A mRNA levels in endometrioma tissue, compared to normal endometrium tissue. Using Western blotting, PLA2G2A was detected in all samples of endometriomas, but not in normal endometrium, and immunohistochemistry showed PLA2G2A-specific staining in epithelial cells of endometrioma paraffin sections. However, there were no significant differences in PLA2G2A levels between cases and controls according to ELISA of peritoneal fluid (6.0 ± 4.4 ng/ml, 6.6 ± 4.3 ng/ml; p = 0.5240) and serum (2.9 ± 2.1 ng/ml, 3.1 ± 2.2 ng/ml; p = 0.7989). Our data indicate that PLA2G2A is implicated in the pathophysiology of ovarian endometriosis, but that it cannot be used as a diagnostic biomarker.
Collapse
Affiliation(s)
- Vida Kocbek
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana , Ljubljana , Slovenia
| | | | | | | | | | | |
Collapse
|
44
|
Anti-inflammatory Activity of Magnesium Isoglycyrrhizinate Through Inhibition of Phospholipase A2/Arachidonic Acid Pathway. Inflammation 2015; 38:1639-48. [DOI: 10.1007/s10753-015-0140-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
45
|
Bourgeois EA, Subramaniam S, Cheng TY, De Jong A, Layre E, Ly D, Salimi M, Legaspi A, Modlin RL, Salio M, Cerundolo V, Moody DB, Ogg G. Bee venom processes human skin lipids for presentation by CD1a. J Exp Med 2015; 212:149-63. [PMID: 25584012 PMCID: PMC4322046 DOI: 10.1084/jem.20141505] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/11/2014] [Indexed: 12/31/2022] Open
Abstract
Venoms frequently co-opt host immune responses, so study of their mode of action can provide insight into novel inflammatory pathways. Using bee and wasp venom responses as a model system, we investigated whether venoms contain CD1-presented antigens. Here, we show that venoms activate human T cells via CD1a proteins. Whereas CD1 proteins typically present lipids, chromatographic separation of venoms unexpectedly showed that stimulatory factors partition into protein-containing fractions. This finding was explained by demonstrating that bee venom-derived phospholipase A2 (PLA2) activates T cells through generation of small neoantigens, such as free fatty acids and lysophospholipids, from common phosphodiacylglycerides. Patient studies showed that injected PLA2 generates lysophospholipids within human skin in vivo, and polyclonal T cell responses are dependent on CD1a protein and PLA2. These findings support a previously unknown skin immune response based on T cell recognition of CD1a proteins and lipid neoantigen generated in vivo by phospholipases. The findings have implications for skin barrier sensing by T cells and mechanisms underlying phospholipase-dependent inflammatory skin disease.
Collapse
Affiliation(s)
- Elvire A Bourgeois
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Sumithra Subramaniam
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK
| | - Tan-Yun Cheng
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Annemieke De Jong
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Emilie Layre
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Dalam Ly
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Maryam Salimi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK
| | - Annaliza Legaspi
- Division of Dermatology, David Geffen School of Medicine, Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095 Division of Dermatology, David Geffen School of Medicine, Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095
| | - Robert L Modlin
- Division of Dermatology, David Geffen School of Medicine, Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095 Division of Dermatology, David Geffen School of Medicine, Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095
| | - Mariolina Salio
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK
| | - D Branch Moody
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Graham Ogg
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and University of Oxford NIHR Biomedical Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK
| |
Collapse
|
46
|
Kabashima K, Izuhara K. Lipid mediators in allergy: Link between human and animal models. Allergol Int 2015; 64:2-3. [PMID: 25572552 DOI: 10.1016/j.alit.2014.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Indexed: 10/24/2022] Open
Affiliation(s)
- Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan.
| |
Collapse
|
47
|
Thamphiwatana S, Gao W, Pornpattananangkul D, Zhang Q, Fu V, Li J, Li J, Obonyo M, Zhang L. Phospholipase A2-responsive antibiotic delivery via nanoparticle-stabilized liposomes for the treatment of bacterial infection. J Mater Chem B 2014; 2:8201-8207. [PMID: 25544886 PMCID: PMC4276341 DOI: 10.1039/c4tb01110d] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Adsorbing small charged nanoparticles onto liposome surfaces to stabilize them against fusion and payload leakage has resulted in a new class of liposomes capable of environment-responsive drug delivery. Herein, we engineered a liposome formulation with a lipid composition sensitive to bacterium-secreted phospholipase A2 (PLA2) and adsorbed chitosan-modified gold nanoparticles (AuChi) onto the liposome surface. The resulting AuChi-stabilized liposomes (AuChi-liposomes) showed prohibited fusion activity and negligible drug leakage. However, upon exposure to either purified PLA2 enzyme or PLA2 secreted by Helicobacter pylori (H. pylori) bacteria in culture, AuChi-liposomes rapidly released the encapsulated payloads and such responsive release was retarded by adding quinacrine dihydrochloride, a PLA2 inhibitor. When loaded with doxycycline, AuChi-liposomes effectively inhibited H. pylori growth. Overall, the AuChi-liposomes allowed for smart "on-demand" antibitoic delivery: the more enzymes or bacteria present at the infection site, the more drug will be released to treat the infection. Given the strong association of PLA2 with a diverse range of diseases, the present liposomal delivery technique holds broad application potential for tissue microenvironment-responsive drug delivery.
Collapse
Affiliation(s)
- Soracha Thamphiwatana
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Weiwei Gao
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Dissaya Pornpattananangkul
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Qiangzhe Zhang
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Victoria Fu
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Jiayang Li
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Jieming Li
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Marygorret Obonyo
- Department of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Liangfang Zhang
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
48
|
Brglez V, Lambeau G, Petan T. Secreted phospholipases A2 in cancer: Diverse mechanisms of action. Biochimie 2014; 107 Pt A:114-23. [DOI: 10.1016/j.biochi.2014.09.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 09/25/2014] [Indexed: 12/24/2022]
|
49
|
Neutrophil effector responses are suppressed by secretory phospholipase A2 modified HDL. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:184-93. [PMID: 25463476 DOI: 10.1016/j.bbalip.2014.11.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/20/2014] [Accepted: 11/17/2014] [Indexed: 01/04/2023]
Abstract
Secretory phospholipase A2 (sPLA2) generates bioactive lysophospholipids implicated in acute and chronic inflammation, but the pathophysiologic role of sPLA2 is poorly understood. Given that high-density lipoprotein (HDL) is the major substrate for sPLA2 in plasma, we investigated the effects of sPLA2-mediated modification of HDL (sPLA2-HDL) on neutrophil function, an essential arm of the innate immune response and atherosclerosis. Treatment of neutrophils with sPLA2-HDL rapidly prevented agonist-induced neutrophil activation, including shape change, neutrophil extracellular trap formation, CD11b activation, adhesion under flow and migration of neutrophils. The cholesterol-mobilizing activity of sPLA2-HDL was markedly increased when compared to native HDL, promoting a significant reduction of cholesterol-rich signaling microdomains integral to cellular signaling pathways. Moreover, sPLA2-HDL effectively suppressed agonist-induced rise in intracellular Ca²⁺ levels. Native HDL showed no significant effects and removing lysophospholipids from sPLA2-HDL abolished all anti-inflammatory activities. Overall, our studies suggest that the increased cholesterol-mobilizing activity of sPLA2-HDL and suppression of rise in intracellular Ca²⁺ levels are likely mechanism that counteracts agonist-induced activation of neutrophils. These counterintuitive findings imply that neutrophil trafficking and effector responses are altered by sPLA2-HDL during inflammatory conditions.
Collapse
|
50
|
Ubiquitin activates patatin-like phospholipases from multiple bacterial species. J Bacteriol 2014; 197:529-41. [PMID: 25404699 DOI: 10.1128/jb.02402-14] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Phospholipase A2 enzymes are ubiquitously distributed throughout the prokaryotic and eukaryotic kingdoms and are utilized in a wide array of cellular processes and physiological and immunological responses. Several patatin-like phospholipase homologs of ExoU from Pseudomonas aeruginosa were selected on the premise that ubiquitin activation of this class of bacterial enzymes was a conserved process. We found that ubiquitin activated all phospholipases tested in both in vitro and in vivo assays via a conserved serine-aspartate catalytic dyad. Ubiquitin chains versus monomeric ubiquitin were superior in inducing catalysis, and ubiquitin-like proteins failed to activate phospholipase activity. Toxicity studies in a prokaryotic dual-expression system grouped the enzymes into high- and low-toxicity classes. Toxicity measured in eukaryotic cells also suggested a two-tiered classification but was not predictive of the severity of cellular damage, suggesting that each enzyme may correspond to unique properties perhaps based on its specific biological function. Additional studies on lipid binding preference suggest that some enzymes in this family may be differentially sensitive to phosphatidyl-4,5-bisphosphate in terms of catalytic activation enhancement and binding affinity. Further analysis of the function and amino acid sequences of this enzyme family may lead to a useful approach to formulating a unifying model of how these phospholipases behave after delivery into the cytoplasmic compartment.
Collapse
|