1
|
Hoxha M, Spahiu E, Prendi E, Zappacosta B. A Systematic Review on the Role of Arachidonic Acid Pathway in Multiple Sclerosis. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 21:160-187. [PMID: 32842948 DOI: 10.2174/1871527319666200825164123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/28/2020] [Accepted: 07/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND & OBJECTIVE Multiple sclerosis (MS) is an inflammatory neurodegenerative disease characterized by destruction of oligodendrocytes, immune cell infiltration and demyelination. Inflammation plays a significant role in MS, and the inflammatory mediators such as eicosanoids, leukotrienes, superoxide radicals are involved in pro-inflammatory responses in MS. In this systematic review we tried to define and discuss all the findings of in vivo animal studies and human clinical trials on the potential association between arachidonic acid (AA) pathway and multiple sclerosis. METHODS A systematic literature search across Pubmed, Scopus, Embase and Cochrane database was conducted. This systematic review was performed according to PRISMA guidelines. RESULTS A total of 146 studies were included, of which 34 were conducted in animals, 58 in humans, and 60 studies reported the role of different compounds that target AA mediators or their corresponding enzymes/ receptors, and can have a therapeutic effect in MS. These results suggest that eicosanoids have significant roles in experimental autoimmune encephalomyelitis (EAE) and MS. The data from animal and human studies elucidated that PGI2, PGF2α, PGD2, isoprostanes, PGE2, PLA2, LTs are increased in MS. PLA2 inhibition modulates the progression of the disease. PGE1 analogues can be a useful option in the treatment of MS. CONCLUSIONS All studies reported the beneficial effects of COX and LOX inhibitors in MS. The hybrid compounds, such as COX-2 inhibitors/TP antagonists and 5-LOX inhibitors can be an innovative approach for multiple sclerosis treatment. Future work in MS should shed light in synthesizing new compounds targeting arachidonic acid pathway.
Collapse
Affiliation(s)
- Malvina Hoxha
- Department of Chemical-Toxicological and Pharmacological Evaluations of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, Rruga Dritan Hoxha, Tirana. Albania
| | | | - Emanuela Prendi
- Catholic University Our Lady of Good Counsel, Department of Biomedical Sciences, Rruga Dritan Hoxha, Tirana. Albania
| | - Bruno Zappacosta
- Department of Chemical-Toxicological and Pharmacological Evaluations of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, Rruga Dritan Hoxha, Tirana. Albania
| |
Collapse
|
2
|
Trotter A, Anstadt E, Clark RB, Nichols F, Dwivedi A, Aung K, Cervantes JL. The role of phospholipase A2 in multiple Sclerosis: A systematic review and meta-analysis. Mult Scler Relat Disord 2018; 27:206-213. [PMID: 30412818 DOI: 10.1016/j.msard.2018.10.115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/21/2018] [Accepted: 10/29/2018] [Indexed: 01/25/2023]
Abstract
Phospholipases A2 (PLA2) are a diverse group of enzymes that cleave the fatty acids of membrane phospholipids. They play critical roles in pathogenesis of neurodegenerative diseases such as multiple sclerosis by enhancing oxidative stress and initiating inflammation. The levels of PLA2 activity in MS patients compared to controls and role of inhibiting PLA2 activity on severity scores in different experimental models are not comprehensively assessed in the light of varying evidence from published studies. The objective of this systematic review is to determine the association between PLA2 activity and multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). We performed a systematic review of six studies that assessed PLA2 activity in MS patients compared to controls and nine studies that assessed PLA2 activity in EAE. sPLA2 nor Lp-PLA2 activity were not increased in MS compared to controls in five of those six studies. A difference in sPLA2 activity was only found in a study that measured the enzyme activity in urine. However, inhibiting cPLA2 or sPLA2 led to lower clinical severity or no signs of EAE in mice, and a lower incidence of EAE lesions compared to animals without cPLA2 inhibition. These findings indicate that PLA2 appears to play a role in the pathogenesis of EAE.
Collapse
Affiliation(s)
- Austin Trotter
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Emily Anstadt
- Department of Immunology, and Department of Medicine, Farmington, CT, USA
| | - Robert B Clark
- Department of Immunology, and Department of Medicine, Farmington, CT, USA; University of Connecticut School of Medicine, Farmington, CT, USA
| | - Frank Nichols
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, CT, USA
| | - Alok Dwivedi
- Department of Biomedical Sciences, Division of Biostatistics and Epidemiology, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Koko Aung
- Department of Internal Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Jorge L Cervantes
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA; Department of Medical Education, Texas Tech University Health Sciences Center, El Paso, TX, USA.
| |
Collapse
|
3
|
Floegel A, Kühn T, Sookthai D, Johnson T, Prehn C, Rolle-Kampczyk U, Otto W, Weikert C, Illig T, von Bergen M, Adamski J, Boeing H, Kaaks R, Pischon T. Serum metabolites and risk of myocardial infarction and ischemic stroke: a targeted metabolomic approach in two German prospective cohorts. Eur J Epidemiol 2017; 33:55-66. [PMID: 29181692 PMCID: PMC5803284 DOI: 10.1007/s10654-017-0333-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/20/2017] [Indexed: 11/24/2022]
Abstract
Metabolomic approaches in prospective cohorts may offer a unique snapshot into early metabolic perturbations that are associated with a higher risk of cardiovascular diseases (CVD) in healthy people. We investigated the association of 105 serum metabolites, including acylcarnitines, amino acids, phospholipids and hexose, with risk of myocardial infarction (MI) and ischemic stroke in the European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam (27,548 adults) and Heidelberg (25,540 adults) cohorts. Using case-cohort designs, we measured metabolites among individuals who were free of CVD and diabetes at blood draw but developed MI (n = 204 and n = 228) or stroke (n = 147 and n = 121) during follow-up (mean, 7.8 and 7.3 years) and among randomly drawn subcohorts (n = 2214 and n = 770). We used Cox regression analysis and combined results using meta-analysis. Independent of classical CVD risk factors, ten metabolites were associated with risk of MI in both cohorts, including sphingomyelins, diacyl-phosphatidylcholines and acyl-alkyl-phosphatidylcholines with pooled relative risks in the range of 1.21–1.40 per one standard deviation increase in metabolite concentrations. The metabolites showed positive correlations with total- and LDL-cholesterol (r ranged from 0.13 to 0.57). When additionally adjusting for total-, LDL- and HDL-cholesterol, triglycerides and C-reactive protein, acyl-alkyl-phosphatidylcholine C36:3 and diacyl-phosphatidylcholines C38:3 and C40:4 remained associated with risk of MI. When added to classical CVD risk models these metabolites further improved CVD prediction (c-statistics increased from 0.8365 to 0.8384 in EPIC-Potsdam and from 0.8344 to 0.8378 in EPIC-Heidelberg). None of the metabolites was consistently associated with stroke risk. Alterations in sphingomyelin and phosphatidylcholine metabolism, and particularly metabolites of the arachidonic acid pathway are independently associated with risk of MI in healthy adults.
Collapse
Affiliation(s)
- Anna Floegel
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany. .,Leibniz Institute for Prevention Research and Epidemiology - BIPS, Achterstraße 30, 28359, Bremen, Germany.
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Disorn Sookthai
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Theron Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cornelia Prehn
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Wolfgang Otto
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Cornelia Weikert
- Department of Food Safety, Federal Institute for Risk Assessment, Berlin, Germany.,Institute for Social Medicine, Epidemiology and Health Economics, Charité University Medical Center, Berlin, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany.,Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany.,University of Aalborg, Fredrik Bajers Vej 7H, 9220, Aalborg East, Denmark
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tobias Pischon
- Molecular Epidemiology Group, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.,Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
4
|
Rezapour-Firouzi S. Herbal Oil Supplement With Hot-Nature Diet for Multiple Sclerosis. NUTRITION AND LIFESTYLE IN NEUROLOGICAL AUTOIMMUNE DISEASES 2017:229-245. [DOI: 10.1016/b978-0-12-805298-3.00024-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
5
|
Ingber A, Cohen Y, Krimsky M, Yedgar S. A Novel Treatment of Contact Dermatitis by Topical Application of Phospholipase A2 Inhibitor: A Double-Blind Placebo-Controlled Pilot Study. Int J Immunopathol Pharmacol 2016; 20:191-5. [PMID: 17346444 DOI: 10.1177/039463200702000123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Phospholipase A2 hydrolyzes membrane phospholipids releasing arachidonic acid and lysophospholipids. These are key precursors of inflammatory mediators, such as prostaglandins, leukotrienes, thromboxanes and PAF, in numerous inflammatory/allergic diseases, including skin inflammation. Accordingly, inhibition of PLA2 has long been postulated as a potentially potent antiinflammatory therapy. In the present study we tested the effect of a novel PLA2 inhibitor on contact dermatitis in human subjects. A double-blind, placebo-controlled pilot study was conducted on contact dermatitis patients (n = 11) treated with the inhibitor-containing topical preparation (1% cream). Disease severity was assessed by physician's assessment before treatment (day 0) as well as after 14-days and 30-days. Patients treated with 1% PLA2 inhibitor-containing cream showed a 69.9% reduction in disease score while placebo-treated patients showed a reduction of 36.5% with p = 0.0024. The clear improvement in the disease score of inhibitor-treated patients supports the involvement of PLA2 activity in skin inflammation and the therapeutic prospective of its inhibition.
Collapse
|
6
|
Rezapour-Firouzi S, Arefhosseini SR, Ebrahimi-Mamaghani M, Baradaran B, Sadeghihokmabad E, Mostafaei S, Torbati M, Chehreh M. Alteration of delta-6-desaturase (FADS2), secretory phospholipase-A2 (sPLA2) enzymes by Hot-nature diet with co-supplemented hemp seed, evening primrose oils intervention in multiple sclerosis patients. Complement Ther Med 2015; 23:652-7. [DOI: 10.1016/j.ctim.2015.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 04/26/2015] [Accepted: 07/05/2015] [Indexed: 10/23/2022] Open
|
7
|
Jové M, Mauri-Capdevila G, Suárez I, Cambray S, Sanahuja J, Quílez A, Farré J, Benabdelhak I, Pamplona R, Portero-Otín M, Purroy F. Metabolomics predicts stroke recurrence after transient ischemic attack. Neurology 2014; 84:36-45. [PMID: 25471397 DOI: 10.1212/wnl.0000000000001093] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To discover, by using metabolomics, novel candidate biomarkers for stroke recurrence (SR) with a higher prediction power than present ones. METHODS Metabolomic analysis was performed by liquid chromatography coupled to mass spectrometry in plasma samples from an initial cohort of 131 TIA patients recruited <24 hours after the onset of symptoms. Pattern analysis and metabolomic profiling, performed by multivariate statistics, disclosed specific SR and large-artery atherosclerosis (LAA) biomarkers. The use of these methods in an independent cohort (162 subjects) confirmed the results obtained in the first cohort. RESULTS Metabolomics analyses could predict SR using pattern recognition methods. Low concentrations of a specific lysophosphatidylcholine (LysoPC[16:0]) were significantly associated with SR. Moreover, LysoPC(20:4) also arose as a potential SR biomarker, increasing the prediction power of age, blood pressure, clinical features, duration of symptoms, and diabetes scale (ABCD2) and LAA. Individuals who present early (<3 months) recurrence have a specific metabolomic pattern, differing from non-SR and late SR subjects. Finally, a potential LAA biomarker, LysoPC(22:6), was also described. CONCLUSIONS The use of metabolomics in SR biomarker research improves the predictive power of conventional predictors such as ABCD2 and LAA. Moreover, pattern recognition methods allow us to discriminate not only SR patients but also early and late SR cases.
Collapse
Affiliation(s)
- Mariona Jové
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Gerard Mauri-Capdevila
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Idalmis Suárez
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Serafi Cambray
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Jordi Sanahuja
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Alejandro Quílez
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Joan Farré
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Ikram Benabdelhak
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Reinald Pamplona
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Manuel Portero-Otín
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain
| | - Francisco Purroy
- From NUTREN-Nutrigenomics Center (M.J., M.P.-O.), Department of Experimental Medicine (R.P.), Parc Científic i Tecnològic Agroalimentari de Lleida-Universitat de Lleida-IRBLleida, Lleida; Stroke Unit (G.M.-C., I.S., S.C., J.S., A.Q., I.B., F.P.), Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova de Lleida, IRBLleida; and Laboratori Clinic (J.F.), Universitari Arnau de Vilanova de Lleida, IRBLleida, Spain.
| |
Collapse
|
8
|
The proteome of the differentiating mesencephalic progenitor cell line CSM14.1 in vitro. BIOMED RESEARCH INTERNATIONAL 2014; 2014:351821. [PMID: 24592386 PMCID: PMC3925624 DOI: 10.1155/2014/351821] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 12/16/2013] [Indexed: 11/23/2022]
Abstract
The treatment of Parkinson's disease by transplantation of dopaminergic (DA) neurons from human embryonic mesencephalic tissue is a promising approach. However, the origin of these cells causes major problems: availability and standardization of the graft. Therefore, the generation of unlimited numbers of DA neurons from various types of stem or progenitor cells has been brought into focus. A source for DA neurons might be conditionally immortalized progenitor cells. The temperature-sensitive immortalized cell line CSM14.1 derived from the mesencephalon of an embryonic rat has been used successfully for transplantation experiments. This cell line was analyzed by unbiased stereology of cell type specific marker proteins and 2D-gel electrophoresis followed by mass spectrometry to characterize the differentially expressed proteome. Undifferentiated CSM14.1 cells only expressed the stem cell marker nestin, whereas differentiated cells expressed GFAP or NeuN and tyrosine hydroxylase. An increase of the latter cells during differentiation could be shown. By using proteomics an explanation on the protein level was found for the observed changes in cell morphology during differentiation, when CSM14.1 cells possessed the morphology of multipolar neurons. The results obtained in this study confirm the suitability of CSM14.1 cells as an in vitro model for the study of neuronal and dopaminergic differentiation in rats.
Collapse
|
9
|
The role of secretory phospholipase A₂ in the central nervous system and neurological diseases. Mol Neurobiol 2013; 49:863-76. [PMID: 24113843 DOI: 10.1007/s12035-013-8565-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/25/2013] [Indexed: 12/31/2022]
Abstract
Secretory phospholipase A2 (sPLA2s) are small secreted proteins (14-18 kDa) and require submillimolar levels of Ca(2+) for liberating arachidonic acid from cell membrane lipids. In addition to the enzymatic function, sPLA2 can exert various biological responses by binding to specific receptors. Physiologically, sPLA2s play important roles on the neurotransmission in the central nervous system and the neuritogenesis in the peripheral nervous system. Pathologically, sPLA2s are involved in the neurodegenerative diseases (e.g., Alzheimer's disease) and cerebrovascular diseases (e.g., stoke). The common pathology (e.g., neuronal apoptosis) of Alzheimer's disease and stroke coexists in the mixed dementia, suggesting common pathogenic mechanisms of the two neurological diseases. Among mammalian sPLA2s, sPLA2-IB and sPLA2-IIA induce neuronal apoptosis in rat cortical neurons. The excess influx of calcium into neurons via L-type voltage-dependent Ca(2+) channels mediates the two sPLA2-induced apoptosis. The elevated concentration of intracellular calcium activates PKC, MAPK and cytosolic PLA2. Moreover, it is linked with the production of reactive oxygen species and apoptosis through activation of the superoxide producing enzyme NADPH oxidase. NADPH oxidase is involved in the neurotoxicity of amyloid β peptide, which impairs synaptic plasticity long before its deposition in the form of amyloid plaques of Alzheimer's disease. In turn, reactive oxygen species from NADPH oxidase can stimulate ERK1/2 phosphorylation and activation of cPLA2 and result in a release of arachidonic acid. sPLA2 is up-regulated in both Alzheimer's disease and cerebrovascular disease, suggesting the involvement of sPLA2 in the common pathogenic mechanisms of the two diseases. Thus, our review presents evidences for pathophysiological roles of sPLA2 in the central nervous system and neurological diseases.
Collapse
|
10
|
Siroos B, Balood M, Zahednasab H, Mesbah-Namin SA, Pourgholy F, Harirchian MH. Secretory Phospholipase A2 activity in serum and cerebrospinal fluid of patients with relapsing-remitting multiple sclerosis. J Neuroimmunol 2013; 262:125-7. [DOI: 10.1016/j.jneuroim.2013.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/17/2013] [Accepted: 06/19/2013] [Indexed: 02/01/2023]
|
11
|
Lysoglycerophospholipids in chronic inflammatory disorders: The PLA2/LPC and ATX/LPA axes. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:42-60. [DOI: 10.1016/j.bbalip.2012.07.019] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/20/2012] [Accepted: 07/24/2012] [Indexed: 02/08/2023]
|
12
|
Dan P, Rosenblat G, Yedgar S. Phospholipase A2 activities in skin physiology and pathology. Eur J Pharmacol 2012; 691:1-8. [DOI: 10.1016/j.ejphar.2012.07.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Revised: 06/21/2012] [Accepted: 07/02/2012] [Indexed: 01/22/2023]
|
13
|
Wang G, Qian P, Xu Z, Zhang J, Wang Y, Cheng S, Cai W, Qian G, Wang C, Decoster MA. Regulatory effects of the JAK3/STAT1 pathway on the release of secreted phospholipase A₂-IIA in microvascular endothelial cells of the injured brain. J Neuroinflammation 2012; 9:170. [PMID: 22788969 PMCID: PMC3409030 DOI: 10.1186/1742-2094-9-170] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Accepted: 07/12/2012] [Indexed: 02/07/2023] Open
Abstract
Background Secreted phospholipase A2-IIA (sPLA2-IIA) is an inducible enzyme released under several inflammatory conditions. It has been shown that sPLA2-IIA is released from rat brain astrocytes after inflammatory stimulus, and lipopolysaccharide (LPS) and nitric oxide (NO) have been implicated in regulation of this release. Here, brain microvascular endothelial cells (BMVECs) were treated with LPS to uncover whether sPLA2-IIA was released, whether nitric oxide regulated this release, and any related signal mechanisms. Methods Supernatants were collected from primary cultures of BMVECs. The release of sPLA2-IIA, and the expression of inducible nitric oxide synthase (iNOS), phospho-JAK3, phospho-STAT1, total JAK3 and STAT1, β-actin, and bovine serum albumin (BSA) were analyzed by Western blot or ELISA. NO production was calculated by the Griess reaction. sPLA2 enzyme activity was measured with a fluorometric assay. Specific inhibitors of NO (L-NAME and aminoguanidine, AG), JAK3 (WHI-P154,WHI), STAT1 (fludarabine, Flu), and STAT1 siRNA were used to determine the involvement of these molecules in the LPS-induced release of sPLA2-IIA from BMVECs. Nuclear STAT1 activation was tested with the EMSA method. The monolayer permeability of BMVECs was measured with a diffusion assay using biotinylated BSA. Results Treatment of BMVECs with LPS increased the release of sPLA2-IIA and nitrite into the cell culture medium up to 24 h. Pretreatment with an NO donor, sodium nitroprusside, decreased LPS-induced sPLA2-IIA release and sPLA2 enzyme activity, and enhanced the expression of iNOS and nitrite generation after LPS treatment. Pretreatment with L-NAME, AG, WHI-P154, or Flu notably reduced the expression of iNOS and nitrite, but increased sPLA2-IIA protein levels and sPLA2 enzyme activity. In addition, pretreatment of the cells with STAT1 siRNA inhibited the phosphorylation of STAT1, iNOS expression, and nitrite production, and enhanced the release of sPLA2-IIA. Pretreatment with the specific inhibitors of NOS, JAK2, and STAT3 decreased the permeability of BMVECs. In contrast, inhibition of sPLA2-IIA release increased cell permeability. These results suggest that sPLA2-IIA expression is regulated by the NO-JAK3-STAT1 pathway. Importantly, sPLA2-IIA augmentation could protect the LPS-induced permeability of BMVECs. Conclusion Our results demonstrate the important action of sPLA2-IIA in the permeability of microvascular endothelial cells during brain inflammatory events. The sPLA2 and NO pathways can be potential targets for the management of brain MVEC injuries and related inflammation.
Collapse
Affiliation(s)
- Guansong Wang
- Neuronscience Program, Institute of Respiratory Diseases in Xinqiao Hospital, Chongqing 400037, P.R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Chalbot S, Zetterberg H, Blennow K, Fladby T, Andreasen N, Grundke-Iqbal I, Iqbal K. Blood-cerebrospinal fluid barrier permeability in Alzheimer's disease. J Alzheimers Dis 2011; 25:505-15. [PMID: 21471645 DOI: 10.3233/jad-2011-101959] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The role of blood-cerebrospinal fluid barrier (BCB) dysfunction in Alzheimer's disease (AD) has been addressed but not yet established. We evaluated the BCB integrity in 179 samples of cerebrospinal fluid (CSF) retrospectively collected from AD patients and control cases using both CSF/serum albumin ratio (QAlb) and CSF secretory Ca2+-dependent phospholipase A2 (sPLA2) activity. These analyses were supplemented with the measurement of total tau, amyloid-β1-42 (Aβ1-42), and ubiquitin CSF levels. We found that due to its higher sensitivity, CSF sPLA2 activity could 1) discriminate AD from healthy controls and 2) showed BCB impairment in neurological control cases while QAlb could not. Moreover, the CSF sPLA2 activity measurement showed that around half of the AD patients were characterized by a BCB impairment. The BCB dysfunction observed in AD was independent from Mini-Mental State Examination score as well as CSF levels of total tau, Aβ1-42, and ubiquitin. Finally, the BCB dysfunction was not limited to any of the CSF biomarkers-based previously identified subgroups of AD. These results suggest that the BCB damage occurs independent of and probably precedes both Aβ and tau pathologies in a restricted subgroup of AD patients.
Collapse
Affiliation(s)
- Sonia Chalbot
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY 10314-6399, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Jeong EM, Ahn KH, Jeon HJ, Kim HD, Lee HS, Jung SY, Jung KM, Kim SK, Bonventre JV, Kim DK. Purification and characterization of a cytosolic Ca(2+)-independent phospholipase A(2) from bovine brain. Mol Cells 2011; 32:405-13. [PMID: 21874539 PMCID: PMC3887695 DOI: 10.1007/s10059-011-1058-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 06/13/2011] [Accepted: 07/01/2011] [Indexed: 02/02/2023] Open
Abstract
The Ca(2+)-independent phospholipase A(2) (iPLA(2)) subfamily of enzymes is associated with arachidonic acid (AA) release and the subsequent increase in fatty acid turnover. This phenomenon occurs not only during apoptosis but also during inflammation and lymphocyte proliferation. In this study, we purified and characterized a novel type of iPLA(2) from bovine brain. iPLA(2) was purified 4,174-fold from the bovine brain by a sequential process involving DEAE-cellulose anion exchange, phenyl-5PW hydrophobic interaction, heparin-Sepharose affinity, Sephacryl S-300 gel filtration, Mono S cation exchange, Mono Q anion exchange, and Superose 12 gel filtration. A single peak of iPLA(2) activity was eluted at an apparent molecular mass of 155 kDa during the final Superose 12 gel-filtration step. The purified enzyme had an isoelectric point of 5.3 on two-dimensional gel electrophoresis (2-DE) and was inhibited by arachidonyl trifluoromethyl ketone (AACOCF(3)), Triton X-100, iron, and Ca(2+). However, it was not inhibited by bromoenol lactone (BEL), an inhibitor of iPLA(2), and adenosine triphosphate (ATP). The spot with the iPLA(2) activity did not match with any known protein sequence, as determined by matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) analysis. Altogether, these data suggest that the purified enzyme is a novel form of cytosolic iPLA(2).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Joseph V. Bonventre
- Medical Services, Brigham and Women’s Hospital, Harvard Medical School, Boston, Longwood, MA, USA
| | | |
Collapse
|
16
|
Palumbo S, Toscano CD, Parente L, Weigert R, Bosetti F. Time-dependent changes in the brain arachidonic acid cascade during cuprizone-induced demyelination and remyelination. Prostaglandins Leukot Essent Fatty Acids 2011; 85:29-35. [PMID: 21530210 PMCID: PMC3109126 DOI: 10.1016/j.plefa.2011.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 03/30/2011] [Accepted: 04/04/2011] [Indexed: 12/21/2022]
Abstract
Phospholipases A(2) (PLA(2)) are the enzymatic keys for the activation of the arachidonic acid (AA) cascade and the subsequent synthesis of pro-inflammatory prostanoids (prostaglandins and tromboxanes). Prostanoids play critical roles in the initiation and modulation of inflammation and their levels have been reported increased in several neurological and neurodegenerative disorders, including multiple sclerosis (MS). Here, we aimed to determine whether brain expression PLA(2) enzymes and the terminal prostagland in levels are changed during cuprizone-induced demyelination and in the subsequent remyelination phase. Mice were given the neurotoxicant cuprizone through the diet for six weeks to induce brain demyelination. Then, cuprizone was withdrawn and mice were returned to a normal diet for 6 weeks to allow spontaneous remyelination. We found that after 4-6 weeks of cuprizone, sPLA(2)(V) and cPLA(2), but not iPLA(2)(VI), gene expression was upregulated in the cortex, concomitant with an increase in the expression of astrocyte and microglia markers. Cyclooxygenase (COX)-2 gene expression was consistently upregulated during all the demyelination period, whereas COX-1 sporadically increased only at week 5 of cuprizone exposure. However, we found that at the protein level only sPLA(2)(V) and COX-1 were elevated during demyelination, with COX-1 selectively expressed by activated and infiltrated microglia/macrophages and astrocytes. Levels of PGE(2), PGD(2), PGI(2) and TXB(2) were also increased during demyelination. During remyelination, none of the PLA(2) isoforms was significantly changed, whereas COX-1 and -2 were sporadically upregulated only at the gene expression level. PGE(2), PGI(2) and PGD(2) levels returned to normal, whereas TXB(2) was still upregulated after 3 weeks of cuprizone withdrawal. Our study characterizes for the first time time-dependent changes in the AA metabolic pathway during cuprizone-induced demyelination and the subsequent remyelination and suggests that sPLA(2)(V) is the major isoform contributing to AA release.
Collapse
Affiliation(s)
- S Palumbo
- National Institute on Aging, National Institutes of Health, Brain Physiology and Metabolism Section, Bethesda, MD 20892-0947, USA
| | | | | | | | | |
Collapse
|
17
|
Role of ethanolamine phosphate in the hippocampus of rats with acute experimental autoimmune encephalomyelitis. Neurochem Int 2011; 58:22-34. [DOI: 10.1016/j.neuint.2010.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 09/28/2010] [Accepted: 10/14/2010] [Indexed: 11/21/2022]
|
18
|
Chalbot S, Zetterberg H, Blennow K, Fladby T, Grundke-Iqbal I, Iqbal K. Cerebrospinal fluid secretory Ca2+-dependent phospholipase A2 activity: a biomarker of blood-cerebrospinal fluid barrier permeability. Neurosci Lett 2010; 478:179-83. [PMID: 20470866 PMCID: PMC2885518 DOI: 10.1016/j.neulet.2010.05.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 05/04/2010] [Indexed: 02/03/2023]
Abstract
The blood-brain barrier, the blood-cerebrospinal fluid barrier (BCB) and other specialized brain barriers are increasingly recognized as a major obstacle to the treatment of most brain disorders. The impairment of these barriers has been implicated in neuropathology of several diseases, such as autism, ischemia, multiple sclerosis and Alzheimer disease. This dual function of the blood-neural barriers points out the importance and need for the development of techniques that can evaluate the nature and level of their integrity. Here we report the discovery of CSF secretory Ca(2+)-dependent phospholipase A2 (sPLA2) activity as a measure of BCB permeability. Lumbar CSF from BCB impaired (n=26), multiple sclerosis (n=18) and healthy control (n=32) cases was analyzed using both a newly developed continuous fluorescence assay for CSF sPLA2 activity and CSF/Serum albumin ratio (Q(Alb)), the most common and established method to evaluate BCB permeability. While both measurements showed no significant differences between multiple sclerosis and age-matched normal healthy cases, they were highly correlated. Though the CSF sPLA2 activity and Q(Alb) had over 95% agreement, the former was found to be more sensitive than the latter in measuring low levels of BCB impairment.
Collapse
Affiliation(s)
- Sonia Chalbot
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Department of Neurology, Faculty Division Akershus University Hospital, University of Oslo, Oslo, Norway
| | - Inge Grundke-Iqbal
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Khalid Iqbal
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| |
Collapse
|
19
|
Goracci G, Ferrini M, Nardicchi V. Low Molecular Weight Phospholipases A2 in Mammalian Brain and Neural Cells: Roles in Functions and Dysfunctions. Mol Neurobiol 2010; 41:274-89. [DOI: 10.1007/s12035-010-8108-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 02/11/2010] [Indexed: 12/14/2022]
|
20
|
Abstract
Dynamic changes in cytoplasmic calcium concentration dictate the immunological fate and functions of lymphocytes. During the past few years, important details have been revealed about the mechanism of store-operated calcium entry in lymphocytes, including the molecular identity of calcium release-activated calcium (CRAC) channels and the endoplasmic reticulum (ER) calcium sensor (STIM1) responsible for CRAC channel activation following calcium depletion of stores. However, details of the potential fine regulation of CRAC channel activation that may be imposed on lymphocytes following physiologic stimulation within an inflammatory environment have not been fully addressed. In this review, we discuss several underexplored aspects of store-operated (CRAC-mediated) and store-independent calcium signaling in B lymphocytes. First, we discuss results suggesting that coupling between stores and CRAC channels may be regulated, allowing for fine tuning of CRAC channel activation following depletion of ER stores. Second, we discuss mechanisms that sustain the duration of calcium entry via CRAC channels. Finally, we discuss distinct calcium permeant non-selective cation channels (NSCCs) that are activated by innate stimuli in B cells, the potential means by which these innate calcium signaling pathways and CRAC channels crossregulate one another, and the mechanistic basis and physiologic consequences of innate calcium signaling.
Collapse
Affiliation(s)
- Leslie B King
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
21
|
Kabarowski JH. G2A and LPC: regulatory functions in immunity. Prostaglandins Other Lipid Mediat 2009; 89:73-81. [PMID: 19383550 PMCID: PMC2740801 DOI: 10.1016/j.prostaglandins.2009.04.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 04/10/2009] [Accepted: 04/13/2009] [Indexed: 02/07/2023]
Abstract
The G2A receptor was originally identified by virtue of its transcriptional induction in murine B lymphoid cells in response to oncogenic transformation and treatment with various DNA-damaging agents. While preliminary characterization of cellular responses to G2A overexpression in fibroblastic cell lines suggested that this receptor may negatively regulate cell growth under conditions of proliferative and genotoxic stress, subsequent studies driven by the discovery of lysophosphatidylcholine (LPC) as a regulator of G2A signaling in immunoregulatory cells point to an important role for this receptor in innate and adaptive immunity.
Collapse
Affiliation(s)
- Janusz H Kabarowski
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294-2170, USA.
| |
Collapse
|
22
|
Huang X, Reynolds AD, Mosley RL, Gendelman HE. CD 4+ T cells in the pathobiology of neurodegenerative disorders. J Neuroimmunol 2009; 211:3-15. [PMID: 19439368 PMCID: PMC2696588 DOI: 10.1016/j.jneuroim.2009.04.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Accepted: 04/03/2009] [Indexed: 12/21/2022]
Abstract
CD4+ T cells orchestrate innate and adaptive immunity. In the central nervous system they modulate immune responses including cell trafficking and glial neuroregulatory functions through an array of soluble molecules cell-cell interactions affecting tissue homeostasis. During disease their roles evolve to an auto-aggressive or, alternatively, protective phenotype. How such a balance is struck in the setting of neurodegenerative disorders may reflect a dichotomy between regulatory T cell, anti-inflammatory and neuroprotective activities versus effector T cell inflammation and neurodegeneration. Interestingly, such roles may show commonalities amongst neurodegenerative diseases. Herein we focus on strategies to modulate such CD4+ T cell responses for therapeutic gain.
Collapse
Affiliation(s)
- Xiuyan Huang
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Institute for Tissue Transplantation and Immunology, Jinan University, Guangzhou, Guangdong 510632 China
| | - Ashley D. Reynolds
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - R. Lee Mosley
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Howard E. Gendelman
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| |
Collapse
|
23
|
Osmers I, Smith SS, Parks BW, Yu S, Srivastava R, Wohler JE, Barnum SR, Kabarowski JHS. Deletion of the G2A receptor fails to attenuate experimental autoimmune encephalomyelitis. J Neuroimmunol 2009; 207:18-23. [PMID: 19135725 PMCID: PMC2692575 DOI: 10.1016/j.jneuroim.2008.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Accepted: 11/24/2008] [Indexed: 11/19/2022]
Abstract
Lysophosphatidylcholine (LPC) is a chemotactic lysolipid produced during inflammation by the hydrolytic action of phospholipase A(2) enzymes. LPC stimulates chemotaxis of T cells in vitro through activation of the G protein-coupled receptor, G2A. This has led to the proposition that G2A contributes to the recruitment of T cells to sites of inflammation and thus promotes chronic inflammatory autoimmune diseases associated with the generation and subsequent tissue infiltration of auto-antigen-specific effector T cells. However, one study suggests that G2A may negatively regulate T cell proliferative responses to antigen receptor engagement and thereby attenuates autoimmunity by reducing the generation of autoreactive T cells. To address the relative contribution of these G2A-mediated effects to the pathophysiology of T cell-mediated autoimmune disease, we examined the impact of G2A inactivation on the onset and severity of murine experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). Wild type (G2A(+/+)) and G2A-deficient (G2A(-/-)) C57BL/6J mice exhibited a similar incidence and onset of disease following immunization with MOG(35-55) peptide. Disease severity was only moderately reduced in G2A(-/-) mice. Similar numbers of MOG(35-55) specific T cells were generated in secondary lymphoid organs of MOG(35-55)-immunized G2A(+/+) and G2A(-/-) mice. Comparable numbers of T cells were detected in spinal cords of G2A(+/+) and G2A(-/-) mice. We conclude that the proposed anti-proliferative and chemotactic functions of G2A are not manifested in vivo and therefore therapeutic targeting of G2A is unlikely to be beneficial in the treatment of MS.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/physiology
- Cell Proliferation/drug effects
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Flow Cytometry/methods
- Gene Deletion
- Glycoproteins/adverse effects
- Interferon-gamma/metabolism
- Lymph Nodes/drug effects
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments/adverse effects
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/physiology
- Spleen/cytology
- Spleen/immunology
- Spleen/pathology
- T-Lymphocytes/immunology
- Time Factors
Collapse
Affiliation(s)
- Inga Osmers
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Kalyvas A, Baskakis C, Magrioti V, Constantinou-Kokotou V, Stephens D, López-Vales R, Lu JQ, Yong VW, Dennis EA, Kokotos G, David S. Differing roles for members of the phospholipase A2 superfamily in experimental autoimmune encephalomyelitis. ACTA ACUST UNITED AC 2009; 132:1221-35. [PMID: 19218359 DOI: 10.1093/brain/awp002] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The phospholipase A(2) (PLA(2)) superfamily hydrolyzes phospholipids to release free fatty acids and lysophospholipids, some of which can mediate inflammation and demyelination, hallmarks of the CNS autoimmune disease multiple sclerosis. The expression of two of the intracellular PLA(2)s (cPLA(2) GIVA and iPLA(2) GVIA) and two of the secreted PLA(2)s (sPLA(2) GIIA and sPLA(2) GV) are increased in different stages of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. We show using small molecule inhibitors, that cPLA(2) GIVA plays a role in the onset, and iPLA(2) GVIA in the onset and progression of EAE. We also show a potential role for sPLA(2) in the later remission phase. These studies demonstrate that selective inhibition of iPLA(2) can ameliorate disease progression when treatment is started before or after the onset of symptoms. The effects of these inhibitors on lesion burden, chemokine and cytokine expression as well as on the lipid profile provide insights into their potential modes of action. iPLA(2) is also expressed by macrophages and other immune cells in multiple sclerosis lesions. Our results therefore suggest that iPLA(2) might be an excellent target to block for the treatment of CNS autoimmune diseases, such as multiple sclerosis.
Collapse
Affiliation(s)
- Athena Kalyvas
- Center for Research in Neuroscience, McGill University Health Center Research Institute, Livingston Hall, Room L7-210, 1650 Cedar Ave., Montreal, Quebec, Canada H3G 1A4
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cunningham TJ, Yao L, Lucena A. Product inhibition of secreted phospholipase A2 may explain lysophosphatidylcholines' unexpected therapeutic properties. JOURNAL OF INFLAMMATION-LONDON 2008; 5:17. [PMID: 18945345 PMCID: PMC2580763 DOI: 10.1186/1476-9255-5-17] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Accepted: 10/22/2008] [Indexed: 11/24/2022]
Abstract
Background Lysophosphatidylcholines (lysoPCs) are products of phospholipase A2 (PLA2) enzyme activity, and like the enzyme, have a direct role in toxic inflammatory responses in variety of organ systems. Paradoxically, reduced plasma lysoPC levels have been noted in sepsis patients and systemic treatment with lysoPCs is therapeutic in rodent models of sepsis and ischemia. These observations suggest that elevation of plasma levels of these lipids can actually help to relieve serious inflammatory conditions. We demonstrate that specific lysoPCs act as uncompetitive product inhibitors of plasma secreted PLA2 enzymes (sPLA2s), especially under conditions of elevated enzyme activity, thus providing a feedback mechanism for the observed anti-inflammatory effects of these compounds. Methods Thin layer chromatography and mass spectroscopy were used to estimate total lysoPC concentration and the relative contributions of different lysoPC species in rat plasma samples. Kinetic studies of sPLA2 enzyme activity were conducted on these samples ex vivo and on purified group IA sPLA2 in vitro after addition of specific lysoPC species to the reaction mixture. Enzyme activity was also measured in plasma samples of rats injected with these same lysoPCs. Results Palmitoyl (16:0), stearoyl (18:0) are the most abundant lysoPCs in rat plasma consistent with other reports. Kinetic studies demonstrated that both were uncompetitive inhibitors of plasma sPLA2 enzyme activity. In vitro experiments with group IA sPLA2 confirmed the inhibition and the kinetic properties of these lysoPC species. Decanoyl lysoPC (10:0), which was not detected in plasma, did not inhibit enzyme activity in vitro. LysoPC injections into normal rats resulted in "buffering" of plasma sPLA2 activity in a narrow low range, consistent with the activity-dependent inhibition suggested by the ex vivo and in vitro experiments. Conclusion The results may explain the efficacy of lysoPC therapy during periods of elevated inflammatory activity and further highlight the utility uncompetitive enzyme inhibitors. In this case, the inhibitor is a product of the enzyme reaction, and therefore represents an example of activity-driven feedback inhibition.
Collapse
Affiliation(s)
- Timothy J Cunningham
- Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| | | | | |
Collapse
|
26
|
Titsworth WL, Liu NK, Xu XM. Role of secretory phospholipase a(2) in CNS inflammation: implications in traumatic spinal cord injury. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2008; 7:254-69. [PMID: 18673210 PMCID: PMC2800081 DOI: 10.2174/187152708784936671] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Secretory phospholipases A(2) (sPLA(2)s) are a subfamily of lipolytic enzymes which hydrolyze the acyl bond at the sn-2 position of glycerophospholipids to produce free fatty acids and lysophospholipids. These products are precursors of bioactive eicosanoids and platelet-activating factor (PAF). The hydrolysis of membrane phospholipids by PLA(2) is a rate-limiting step for generation of eicosanoids and PAF. To date, more than 10 isozymes of sPLA(2) have been found in the mammalian central nervous system (CNS). Under physiological conditions, sPLA(2)s are involved in diverse cellular responses, including host defense, phospholipid digestion and metabolism. However, under pathological situations, increased sPLA(2) activity and excessive production of free fatty acids and their metabolites may lead to inflammation, loss of membrane integrity, oxidative stress, and subsequent tissue injury. Emerging evidence suggests that sPLA(2) plays a role in the secondary injury process after traumatic or ischemic injuries in the brain and spinal cord. Importantly, sPLA(2) may act as a convergence molecule that mediates multiple key mechanisms involved in the secondary injury since it can be induced by multiple toxic factors such as inflammatory cytokines, free radicals, and excitatory amino acids, and its activation and metabolites can exacerbate the secondary injury. Blocking sPLA(2) action may represent a novel and efficient strategy to block multiple injury pathways associated with the CNS secondary injury. This review outlines the current knowledge of sPLA(2) in the CNS with emphasis placed on the possible roles of sPLA(2) in mediating CNS injuries, particularly the traumatic and ischemic injuries in the brain and spinal cord.
Collapse
Affiliation(s)
- W. Lee Titsworth
- Kentucky Spinal Cord Injury Research Center, Departments of Neurological Surgery, and Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Nai-Kui Liu
- Kentucky Spinal Cord Injury Research Center, Departments of Neurological Surgery, and Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute and Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiao-Ming Xu
- Kentucky Spinal Cord Injury Research Center, Departments of Neurological Surgery, and Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute and Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
27
|
Torregrosa G, Pérez-Asensio FJ, Burguete MC, Castelló-Ruiz M, Salom JB, Alborch E. Chronic intracerebroventricular delivery of the secretory phospholipase A2 inhibitor, 12-epi-scalaradial, does not improve outcome after focal cerebral ischemia-reperfusion in rats. Exp Brain Res 2008; 176:248-59. [PMID: 16874515 DOI: 10.1007/s00221-006-0611-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2005] [Accepted: 06/19/2006] [Indexed: 10/24/2022]
Abstract
Phospholipase A2s (PLA2s) seem to be involved in the pathophysiology of ischemic brain injury, but their specific role is far from being completely understood. The present study was carried out to ascertain how and to what extent secretory PLA2s (sPLA2s) activity influences outcome after cerebral ischemia-reperfusion, and to correlate this with the inflammatory response. To do this we used the potent and selective sPLA2 inhibitor, 12-epi-scalaradial. Male Wistar rats were separated into three groups: a control group receiving intracerebroventricular vehicle, and two groups receiving intracerebroventricular 0.005 or 0.5 microg/h 12-epi-scalaradial. Every animal was subjected to middle cerebral artery (MCA) occlusion (90 min, intraluminal thread technique) under continuous moni-torization of cerebrocortical perfusion (CP, laser-Doppler flowmetry), followed by reperfusion (3 days). Neurological status, infarct volume, and myeloperoxidase (MPO) activity were the main end points. Three days after the 90-min ischemia period, neurological examination did not reveal significant differences between the three groups of rats. Control rats showed a mean infarct volume of 145.9 +/- 24.7 mm3 (21 +/- 4.1% of the ipsilateral hemisphere volume), while mean infarct volume in rats treated with 0.005 or 0.5 microg/h 12-epi-scalaradial increased to 164.8 +/- 86.8 mm3 (22.0 +/- 10.9%) and 211.5 +/- 12.2 mm3 (28 +/- 3%, P < 0.05), respectively. Treatment with the highest dose of 12-epi-scalaradial (0.5 microg/h) increased MPO activity in the ipsilateral hemisphere by about 140% (from 0.59 +/- 0.59 to 1.42 +/- 1.03 units of activity/g of tissue in comparison with the control ischemic hemisphere, P < 0.05). Overall, our results point to a positive rather than a negative influence of sPLA2 activity during ischemia. This, along with its inability to decrease the inflammatory response, does not allow to propose the use of 12-epi-scalardial as a potential drug for stroke therapy.
Collapse
Affiliation(s)
- Germán Torregrosa
- Centro de Investigación, Hospital Universitario La Fe, Ave. Campanar 21, 46009 Valencia, Spain.
| | | | | | | | | | | |
Collapse
|
28
|
Nizri E, Irony-Tur-Sinai M, Lavon I, Meshulam H, Amitai G, Brenner T. IBU-octyl-cytisine, a novel bifunctional compound eliciting anti-inflammatory and cholinergic activity, ameliorates CNS inflammation by inhibition of T-cell activity. Int Immunopharmacol 2007; 7:1129-39. [PMID: 17630191 DOI: 10.1016/j.intimp.2007.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 03/22/2007] [Accepted: 03/26/2007] [Indexed: 11/25/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a central nervous system (CNS) inflammatory model in which MOG-specific T-cells initiate an autoimmune attack leading to demyelinization and consequently, neurological damage and morbidity. As EAE pathogenesis results from the involvement of immune cells, CNS resident-cells and inflammatory mediators, our treatment strategy was to use a bifunctional compound with dual anti-inflammatory properties: a non-steroidal anti-inflammatory moiety and a nicotinic agonist moiety, intended to interact with the alpha7 nicotinic receptor present on immune cells. We used IBU-Octyl-Cytisine, with an ibuprofen (IBU) moiety and Cytisine, as the nicotinic agonist. The two moieties are attached by an eight carbon (octyl) spacer. Treatment of EAE with IBU-Octyl-Cytisine (2.5 mg/kg/day, i.p.) reduced significantly (by 70%) disease severity and inflammatory infiltrates in the spinal cord. An equivalent dose of IBU was ineffective, whereas Cytisine was significantly toxic. Treatment with IBU-Octyl-Cytisine inhibited the T-cell response toward the encephalitogenic epitope of myelin oligodendrocyte glycoprotein (MOG). In addition, expression of CCR5 by CD4(+)T-cells was lower, indicating a reduced migratory capacity following treatment. IBU-Octyl-Cytisine reduced Th(1) but not Th(2) cytokine production. This reduction was accompanied by a drop in the level of T-bet mRNA, a transcription factor pivotal to Th(1) lineage differentiation. Thus, IBU-Octyl-Cytisine is an effective treatment for EAE, influencing T-cell responses in several stages of disease pathogenesis. This bifunctional compound was more efficient than IBU or Cytisine separately, as well as than both moieties unconjugated. Thus, it seems that this strategy may be applicable in wider context.
Collapse
Affiliation(s)
- Eran Nizri
- Laboratory of Neuroimmunology, Department of Neurology and the Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
29
|
Cunningham TJ, Maciejewski J, Yao L. Inhibition of secreted phospholipase A2 by neuron survival and anti-inflammatory peptide CHEC-9. J Neuroinflammation 2006; 3:25. [PMID: 16965626 PMCID: PMC1624820 DOI: 10.1186/1742-2094-3-25] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2006] [Accepted: 09/11/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The nonapeptide CHEC-9 (CHEASAAQC), a putative inhibitor of secreted phospholipase A2 (sPLA2), has been shown previously to inhibit neuron death and aspects of the inflammatory response following systemic treatment of rats with cerebral cortex lesions. In this study, the properties of CHEC-9 inhibition of sPLA2 enzymes were investigated, using a venom-derived sPLA2 group I and the plasma of rats and humans as the sources of enzyme activity. The results highlight the advantages of inhibitors with uncompetitive properties for inflammatory disorders including those resulting in degeneration of neurons. METHODS Samples of enzyme and plasma were reacted with 1-Palmitoyl-2-Pyrenedecanoyl Phosphatidylcholine, a sPLA2 substrate that forms phospholipid vesicles in aqueous solutions. Some of the plasma samples were collected from restrained peptide-treated rats in order to confirm the validity of the in vitro assays for extrapolation to in vivo effects of the peptide. The enzyme reactions were analyzed in terms of well-studied relationships between the degree of inhibition and the concentrations of different reactants. We also examined interactions between different components of the reaction mixture on native polyacrylamide gels. RESULTS In all cases, the peptide showed the properties of an uncompetitive (or anti-competitive) enzyme inhibitor with Ki values less than 100 nanomolar. The electrophoresis experiments suggested CHEC-9 modifies the binding properties of the enzyme only in the presence of substrate, consistent with its classification as an uncompetitive inhibitor. Both the in vitro observations and the analysis of plasma samples from restrained rats injected with peptide suggest the efficacy of the peptide increases under conditions of high enzyme activity. CONCLUSION Modeling studies by others have shown that uncompetitive inhibitors may be optimal for enzyme inhibition therapy because, unlike competitive inhibitors, they are not rendered ineffective by the accumulation of unmodified substrate. Such conditions likely apply to several instances of neuroinflammation where there are cascading increases in sPLA2s and their substrates, both systemically and in the CNS. Thus, the present results may explain the efficacy of CHEC-9 in vivo.
Collapse
Affiliation(s)
- Timothy J Cunningham
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Jaquie Maciejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Lihua Yao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| |
Collapse
|
30
|
Cunningham TJ, Yao L, Oetinger M, Cort L, Blankenhorn EP, Greenstein JI. Secreted phospholipase A2 activity in experimental autoimmune encephalomyelitis and multiple sclerosis. J Neuroinflammation 2006; 3:26. [PMID: 16965627 PMCID: PMC1592473 DOI: 10.1186/1742-2094-3-26] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2006] [Accepted: 09/11/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There is increased interest in the contribution of the innate immune system to multiple sclerosis (MS), including the activity of acute inflammatory mediators. The purpose of this study was to test the involvement of systemic secreted phospholipase A2 (sPLA2) enzymes in experimental autoimmune encephalomyelitis (EAE), an MS model, and to determine if enzyme activity is elevated in MS patients. METHODS A non-invasive urinary assay was developed in order to monitor enzymatically active sPLA2 levels in Dark Agouti rats after induction of EAE. Some Rats were treated with nonapeptide CHEC-9, an uncompetitive sPLA2 enzyme inhibitor, during the initial rise in urinary enzyme levels. Body weight and clinical EAE score were measured for 18 days post immunization (PI), after which the rats were sacrificed for H&E and myelin staining, and for ED-1 immunocytochemistry, the latter to quantify macrophages and activated microglia. The urinary sPLA2 assay was also applied to un-timed samples collected from a cross section of 44 MS patients and 14 healthy controls. RESULTS Mean levels of enzymatically active sPLA2 in the urine increased following immunization and peaked between days 8-10 PI which was just prior to the onset of EAE symptoms. At this time, a transient attenuation of activity was detected in the urine of CHEC-9 treated rats consistent with the activity-dependent properties of the inhibitor. The peptide also reduced or abolished EAE symptoms compared to vehicle-injected controls. Histopathological changes in the spinal cords of the EAE rats correlated generally with clinical score including a significant reduction in ED-1+ cells after peptide treatment. Multiple Sclerosis patients also showed elevations in sPLA2 enzyme activity. Mean levels of sPLA2 were increased 6-fold in the urine of patients with active disease and 4-fold for patients in remission, regardless of immunomodulating therapy. CONCLUSION The results suggest that sPLA2 enzymes, traditionally thought to be part the acute phase inflammatory response, are therapeutic targets for MS.
Collapse
Affiliation(s)
- Timothy J Cunningham
- Departments of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Lihua Yao
- Departments of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Michelle Oetinger
- Departments of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Laura Cort
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Elizabeth P Blankenhorn
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | | |
Collapse
|
31
|
Farooqui AA, Ong WY, Horrocks LA. Inhibitors of brain phospholipase A2 activity: their neuropharmacological effects and therapeutic importance for the treatment of neurologic disorders. Pharmacol Rev 2006; 58:591-620. [PMID: 16968951 DOI: 10.1124/pr.58.3.7] [Citation(s) in RCA: 247] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The phospholipase A(2) family includes secretory phospholipase A(2), cytosolic phospholipase A(2), plasmalogen-selective phospholipase A(2), and calcium-independent phospholipase A(2). It is generally thought that the release of arachidonic acid by cytosolic phospholipase A(2) is the rate-limiting step in the generation of eicosanoids and platelet activating factor. These lipid mediators play critical roles in the initiation and modulation of inflammation and oxidative stress. Neurological disorders, such as ischemia, spinal cord injury, Alzheimer's disease, multiple sclerosis, prion diseases, and epilepsy are characterized by inflammatory reactions, oxidative stress, altered phospholipid metabolism, accumulation of lipid peroxides, and increased phospholipase A(2) activity. Increased activities of phospholipases A(2) and generation of lipid mediators may be involved in oxidative stress and neuroinflammation associated with the above neurological disorders. Several phospholipase A(2) inhibitors have been recently discovered and used for the treatment of ischemia and other neurological diseases in cell culture and animal models. At this time very little is known about in vivo neurochemical effects, mechanism of action, or toxicity of phospholipase A(2) inhibitors in human or animal models of neurological disorders. In kainic acid-mediated neurotoxicity, the activities of phospholipase A(2) isoforms and their immunoreactivities are markedly increased and phospholipase A(2) inhibitors, quinacrine and chloroquine, arachidonyl trifluoromethyl ketone, bromoenol lactone, cytidine 5-diphosphoamines, and vitamin E, not only inhibit phospholipase A(2) activity and immunoreactivity but also prevent neurodegeneration, suggesting that phospholipase A(2) is involved in the neurodegenerative process. This also suggests that phospholipase A(2) inhibitors can be used as neuroprotectants and anti-inflammatory agents against neurodegenerative processes in neurodegenerative diseases.
Collapse
Affiliation(s)
- Akhlaq A Farooqui
- Department of Molecular and Cellular Biochemistry, The Ohio State University, 1645 Neil Avenue, Columbus, OH 43210-1218, USA
| | | | | |
Collapse
|
32
|
Yedgar S, Cohen Y, Shoseyov D. Control of phospholipase A2 activities for the treatment of inflammatory conditions. Biochim Biophys Acta Mol Cell Biol Lipids 2006; 1761:1373-82. [PMID: 16978919 DOI: 10.1016/j.bbalip.2006.08.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Accepted: 08/01/2006] [Indexed: 01/10/2023]
Abstract
Phospholipase-A2 (PLA2) enzymes hydrolyze cell membrane phospholipids to produce arachidonic acid (AA) and lyso-phospholipids (LysoPL), playing a key role in the production of inflammatory lipid mediators, mainly eicosanoids. They are therefore considered pro-inflammatory enzymes and their inhibition has long been recognized as a desirable therapeutic target. However, attempts to develop suitable PLA2 inhibitors for the treatment of inflammatory diseases have yet to succeed. This is due to their functional and structural diversity, and their homeostatic and even anti-inflammatory roles in certain circumstances. In the present review we outline the diversity and functions of PLA2 isoforms, and their interplay in the induction and inhibition of inflammatory processes, with emphasis on discussing approaches for therapeutic manipulation of PLA2 activities.
Collapse
Affiliation(s)
- Saul Yedgar
- Department of Biochemistry, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | | | | |
Collapse
|
33
|
Zhu P, Liu X, Labelle EF, Freedman BD. Mechanisms of hypotonicity-induced calcium signaling and integrin activation by arachidonic acid-derived inflammatory mediators in B cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:4981-9. [PMID: 16210600 DOI: 10.4049/jimmunol.175.8.4981] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously characterized the initial steps in the activation of novel (calcium-permeant) nonselective cation channels (NSCCs) and calcium release-activated calcium channels in primary murine B lymphocytes. Phospholipase C products, namely diacylglycerol and d-myo-inositol 1,4,5-trisphosphate, were identified as proximal intracellular agonists of these respective channels following mechanical stimulation of B cells. However, neither the distal steps in NSCC activation nor the contribution of these channels to sustained mechanical signaling were defined in these previous studies. In this study, single cell measurements of intracellular Ca(2+) were used to define the mechanisms of NSCC activation and demonstrate a requirement for arachidonic acid liberated from diacylglycerol. Several arachidonic acid-derived derivatives were identified that trigger Ca(2+) entry into B cells, including the lipoxygenase product 5-hydroperoxyeicosatetranenoic acid and the cytochrome P450 hydroxylase product 20-hydroxyeicosatetraenoic; however, the cytochrome P450 epoxygenase product 5,6-epoxyeicosatrienoic acid is primarily responsible for hypotonicity-induced responses. In addition to regulating calcium entry, our data suggest that eicosanoid-activated NSCCs have a separate and direct role in regulating the avidity of integrins on B cells for extracellular matrix proteins, including ICAM-1 and VCAM-1. Thus, in addition to defining a novel osmotically activated signal transduction pathway in B cells, our results have broad implications for understanding how inflammatory mediators dynamically and rapidly regulate B cell adhesion and trafficking.
Collapse
Affiliation(s)
- Peimin Zhu
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
34
|
Wang G, Daniel BM, DeCoster MA. Role of nitric oxide in regulating secreted phospholipase A2 release from astrocytes. Neuroreport 2005; 16:1345-50. [PMID: 16056137 DOI: 10.1097/01.wnr.0000174403.79020.65] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Inflammatory stimuli such as lipopolysaccharide increase nitric oxide and secreted phospholipase A2 release from glial cells. However, the signaling mechanism(s) regulating secreted phospholipase A2 in glial cells is not known. Here, rat brain astrocytes treated with lipopolysaccharide generated nitrite and released secreted phospholipase A2, while microglia generated nitrite without releasing secreted phospholipase A2. Unexpectedly, attenuation of nitrite production by pretreatment with the nitric oxide synthase inhibitor N-Omega-nitro-L-arginine methyl ester greatly enhanced lipopolysaccharide-stimulated secreted phospholipase A2 release from astrocytes; postreatment with N-Omega-nitro-L-arginine methyl ester did not potentiate secreted phospholipase A2 release, and addition of an nitric oxide donor attenuated the secreted phospholipase A2 release. The regulation of secreted phospholipase A2 may act via the transcription factor nuclear factor-kappaB, as a nuclear factor-kappaB inhibitor attenuated lipopolysaccharide-stimulated secreted phospholipase A2 release. These results demonstrate the role of basal nitric oxide levels as a regulator of inflammatory secreted phospholipase A2 release from glial cells of the brain.
Collapse
Affiliation(s)
- Guansong Wang
- Neuroscience Center, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
35
|
Shoseyov D, Bibi H, Offer S, Schwob O, Krimsky M, Kleiman M, Yedgar S. Treatment of ovalbumin-induced experimental allergic bronchitis in rats by inhaled inhibitor of secretory phospholipase A(2). Thorax 2005; 60:747-53. [PMID: 15994250 PMCID: PMC1747528 DOI: 10.1136/thx.2005.043695] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND The pathophysiology of asthma involves the action of inflammatory/allergic lipid mediators formed following membrane phospholipid hydrolysis by phospholipase A(2) (PLA(2)). Cysteinyl leukotrienes are considered potent inducers of bronchoconstriction and airway remodelling. Ovalbumin (OVA) induced bronchoconstriction in rats is associated with increased secretory PLA(2) (sPLA(2)) activation and cysteinyl leukotriene production, together with suppression of cytosolic PLA(2) and prostaglandin E(2). These processes are reversed when the animals are pretreated systemically with an extracellular cell impermeable sPLA(2) inhibitor which also suppresses the early allergic reaction to OVA challenge. In this study we examine the capacity of the sPLA(2) inhibitor to ameliorate inflammatory and allergic manifestations (early and late bronchoconstriction) of OVA induced allergic bronchitis in rats when the inhibitor was administered by inhalation to confine it to the airways. METHODS Rats sensitised with OVA were treated with the sPLA(2) inhibitor hyaluronic acid-linked phosphatidyl ethanolamine (HyPE). The rats were divided into four groups (n = 10 per group): (1) naïve controls (no sensitisation/no treatment); (2) positive controls (sensitisation + challenge with OVA inhalation and subcutaneous injection of 1 ml saline before each challenge; (3) sensitisation + challenge with OVA and HyPE inhalation before every challenge; and (4) sensitisation + challenge with OVA and treatment with subcutaneous dexamethasone (300 mug) before each challenge as a conventional reference. Another group received no treatment with HyPE during the sensitisation process but only before or after challenge of already sensitised rats. Pulmonary function was assessed and changes in the histology of the airways, levels of cysteinyl leukotrienes in BAL fluid, and the production of nitric oxide (No) and tumour necrosis factor alpha (TNFalpha) by BAL macrophages were determined. RESULTS Inhalation of HyPE markedly suppressed OVA induced early and late asthmatic reactions as expressed by bronchoconstriction, airway remodelling (histology), cysteinyl leukotriene level in BAL fluid, and production of TNFalpha and NO by BAL macrophages. OVA induced bronchoconstriction in sensitised non-pretreated rats was also inhibited by inhalation of HyPE either before or after the challenge. CONCLUSIONS These findings confirm the pivotal role of sPLA(2) in the pathophysiology of both the immediate allergic response and the inflammatory asthmatic process. Control of airway sPLA(2) may be a new therapeutic approach to the treatment of asthma.
Collapse
Affiliation(s)
- D Shoseyov
- Department of Biochemistry, Hebrew University-Hadassah Medical School, Jerusalem, Israel 91120
| | | | | | | | | | | | | |
Collapse
|
36
|
Nizri E, Adani R, Meshulam H, Amitai G, Brenner T. Bifunctional compounds eliciting both anti-inflammatory and cholinergic activity as potential drugs for neuroinflammatory impairments. Neurosci Lett 2005; 376:46-50. [PMID: 15694272 DOI: 10.1016/j.neulet.2004.11.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Revised: 11/10/2004] [Accepted: 11/11/2004] [Indexed: 10/26/2022]
Abstract
We tested two novel bifunctional compounds: ibuprofen-N-octyl-pyridostigmine bromide (IBU-PO) and ibuprofen-N-decyl-pyridostigmine bromide (IBU-PD). They both contain a non-steroidal anti-inflammatory drug (NSAID), ibuprofen (IBU) and pyridostigmine (PO), a cholinesterase inhibitor that acts as a cholinergic up-regulator (CURE). The two moieties are conjugated by a hydrocarbon spacer consisting of 8 (octyl) and 10 (decyl) carbons, respectively. The compounds were tested for their efficiency in reducing the neurological symptoms observed in experimental autoimmune encephalomyelitis induced in mice by myelin oligodendrocyte glycoprotein (MOG). IBU-PO and IBU-PD significantly ameliorated the clinical score (a 40-50% reduction in disease severity) over a period of 30 days, following daily administration of 1 and 0.1mg/kg, i.p., respectively. Clinical improvement was accompanied by reduced responsiveness of MOG-specific T-cells. In addition, IBU-PO and IBU-PD down-regulated the production of nitric oxide (NO) and prostaglandin E2 (PGE2) in cultured astrocytes. To determine which moiety was responsible for these effects, we tested each of the two components, IBU and PO. Our findings indicate that combining NSAID with cholinergic intervention contributes an added therapeutic value for each distinct entity and that these bifunctional compounds act both on the peripheral immunological system and on the central nervous system (CNS) inflammatory pathways.
Collapse
Affiliation(s)
- Eran Nizri
- Laboratory of Neuroimmunology, Department of Neurology and the Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, P.O. Box 12000, Jerusalem 91120, Israel
| | | | | | | | | |
Collapse
|
37
|
Offer S, Yedgar S, Schwob O, Krimsky M, Bibi H, Eliraz A, Madar Z, Shoseyov D. Negative feedback between secretory and cytosolic phospholipase A2 and their opposing roles in ovalbumin-induced bronchoconstriction in rats. Am J Physiol Lung Cell Mol Physiol 2004; 288:L523-9. [PMID: 15557087 DOI: 10.1152/ajplung.00199.2004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Phospholipase A2 (PLA2) hydrolyzes cell membrane phospholipids (PL) to produce arachidonic acid and lyso-PL. The PLA2 enzymes include the secretory (sPLA2) and cytosolic (cPLA2) isoforms, which are assumed to act synergistically in production of eicosanoids that are involved in inflammatory processes. However, growing evidence raises the possibility that in airways and asthma-related inflammatory cells (eosinophils, basophils), the production of the bronchoconstrictor cysteinyl leukotrienes (CysLT) is linked exclusively to sPLA2, whereas the bronchodilator prostaglandin PGE2 is produced by cPLA2. It has been further reported that the capacity of airway epithelial cells to produce CysLT is inversely proportional to PGE2 production. This seems to suggest that sPLA2 and cPLA2 play opposing roles in asthma pathophysiology and the possibility of a negative feedback between the two isoenzymes. To test this hypothesis, we examined the effect of a cell-impermeable extracellular sPLA2 inhibitor on bronchoconstriction and PLA2 expression in rats with ovalbumin (OVA)-induced asthma. It was found that OVA-induced bronchoconstriction was associated with elevation of lung sPLA2 expression and CysLT production, concomitantly with suppression of cPLA2 expression and PGE2 production. These were reversed by treatment with the sPLA2 inhibitor, resulting in amelioration of bronchoconstriction and reduced CysLT production and sPLA2 expression, concomitantly with enhanced PGE2 production and cPLA2 expression. This study demonstrates, for the first time in vivo, a negative feedback between sPLA2 and cPLA2 and assigns opposing roles for these enzymes in asthma pathophysiology: sPLA2 activation induces production of the bronchoconstrictor CysLT and suppresses cPLA2 expression and the subsequent production of the bronchodilator PGE2.
Collapse
Affiliation(s)
- Sarit Offer
- Institute of Biochemistry, Faculty of Agriculture, The Hebrew University, Rehovat, Jerusalem
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Yeo JF, Ong WY, Ling SF, Farooqui AA. Intracerebroventricular injection of phospholipases A2 inhibitors modulates allodynia after facial carrageenan injection in mice. Pain 2004; 112:148-55. [PMID: 15494195 DOI: 10.1016/j.pain.2004.08.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Revised: 07/07/2004] [Accepted: 08/03/2004] [Indexed: 11/26/2022]
Abstract
The present study was carried out, using inhibitors to secretory phospholipase A2 (sPLA2, 12-epi-scalaradial), cytosolic phospholipase A2 (cPLA2, AACOCF3), or calcium-independent phospholipase A2 (iPLA2, bromoenol lactone), to compare possible contributions of central nervous PLA2 isoforms to the development of allodynia after facial carrageenan injection in mice. C57BL/6J (B6) mice showed increased responses to facial stimulation using a von Frey hair (1 g force), at 8 h, 1 day, and 3 days after facial carrageenan injection. On the other hand, BALB/c mice did not show increased responses at any of the time points. In both B6 and BALB/c mice, intracerebroventricular injection of inhibitors to each of the three PLA2 isoforms significantly reduced responses to von Frey hair stimulation at 8 h and 1 day after facial carrageenan injection, but at 3 days after injection, only the sPLA2 inhibitor had an effect. Since BALB/c mice did not show increased responses after facial carrageenan injection, the reduction in responses actually indicates that there is loss of normal sensitivity to von Frey hair stimulation after intracerebroventricular injection of each of these inhibitors, in this strain of mice. The effects of PLA2 inhibitors are unlikely to be due simply to inhibition of arachidonic acid generation, since intracerebroventricular injection of arachidonic acid also had an anti-nociceptive effect. The above results support an important role of central nervous PLA2s in neurotransmission and pain transmission.
Collapse
Affiliation(s)
- Jin-Fei Yeo
- Department of Oral and Maxillofacial Surgery, National University of Singapore, Lower Kent Ridge Road, Singapore, Singapore 119260
| | | | | | | |
Collapse
|
39
|
Darville T, Yedgar S, Krimsky M, Andrews CW, Jungas T, Ojcius DM. Protection against Chlamydia trachomatis infection in vitro and modulation of inflammatory response in vivo by membrane-bound glycosaminoglycans. Microbes Infect 2004; 6:369-76. [PMID: 15050964 DOI: 10.1016/j.micinf.2003.12.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2003] [Accepted: 12/24/2003] [Indexed: 11/28/2022]
Abstract
Glycosaminoglycans (GAG) efficiently inhibit adherence of several strains of Chlamydia trachomatis to cell lines in vitro, but none of the GAG have been able to inhibit infections in vivo. One possible cause for failure of GAG inhibition in vivo is the inability to deliver a sustained concentration of GAG at the mucosal surface. We tested the possibility of enhancing cell protection by increasing the cell-surface concentration of GAG using membrane-anchored GAG (MAG), composed of phosphatidylethanolamine (PE)-linked GAG. These lipid conjugates were originally designed as extracellular phospholipase A2 (PLA2) inhibitors and exhibit a dual effect: the lipid moiety incorporates into the cell membrane, interfering with the action of PLA2 on cell membranes, and the anchored GAG protects the cell membrane from exogenous inflammatory mediators. We tested the ability of MAG to block chlamydia infection in vitro and in vivo. The MAG blocked infection of epithelial cells in vitro when added to the cells at the same time or before infection, but not if added after the bacteria had already invaded the host cells. One of the MAG led to the production of aberrant Chlamydia vacuoles, suggesting it may inhibit intracellular PLA2 associated with development of the vacuole. Although the MAG did not inhibit vaginal infection of mice, they decreased significantly the level of secretion of the inflammatory cytokines TNF-alpha and IFN-gamma but had no effect on secretion of the neutrophil chemokine, macrophage inflammatory protein-2 (MIP-2). Acute and chronic inflammatory cell infiltrates were not altered by MAG treatment. These findings suggest that lipid conjugation of GAG could be used as a novel approach for increasing cell-surface concentrations of GAG. The inconclusive in vivo results might be due to the physical properties of the tested MAG or an insufficient application protocol, and their improvement might provide the desired inhibitory effects.
Collapse
Affiliation(s)
- Toni Darville
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR 72205, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Kolko M, Christoffersen NR, Barreiro SG, Bazan NG. Expression and location of mRNAs encoding multiple forms of secretory phospholipase A2 in the rat retina. J Neurosci Res 2004; 77:517-24. [PMID: 15264221 DOI: 10.1002/jnr.20187] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Low-molecular-weight secretory phospholipases A(2) (sPLA(2)s) are a subgroup of PLA(2)s, which are secreted, bind to receptors, and may act as intercellular signaling modulators. At least 10 different groups have been characterized in mammals, and there is expanding evidence of the significance of sPLA(2)s in neuronal signaling and survival [Kolko et al. (1996) J. Biol. Chem. 271: 32722-32728]. To date, no retinal sPLA(2)s have been cloned or characterized. We evaluated the existence and abundance of sPLA(2) subtypes in rat retina and explored their possible involvement in light-induced retinal damage. We designed primers to identify the sPLA(2)s in rat retina, based on known sequences of sPLA(2)-specific mRNAs in other tissues. RNA was isolated from rat retina, and cDNA was produced and used for PCR cloning to identify the novel subtypes of sPLA(2). Our study revealed the presence of mRNAs encoding sPLA(2)-IB, -X, -V, -IIE, -IIA, and -IIF in the retina, and quantification by real-time PCR revealed different abundances of the sPLA(2)s. We showed a time-dependent gene induction of sPLA(2)-X, -IB, and -V in light-induced retinal damage. We further explored the location of sPLA(2)-IB by in situ hybridization and immunohistochemistry. This study is the first to reveal the presence, abundance, and induction of mRNAs encoding sPLA(2)s in rat retina. We suggest that these enzymes are themselves intercellular signaling modulators of retinal cell function and perhaps also of retinal degeneration.
Collapse
Affiliation(s)
- Miriam Kolko
- Neuroscience Center of Excellence and Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | |
Collapse
|