1
|
Dalmaso B, Liber AMP, Ventura DF, Jancar S, Del Debbio CB. Platelet-activating factor receptor (PAFR) regulates neuronal maturation and synaptic transmission during postnatal retinal development. Front Cell Neurosci 2024; 18:1343745. [PMID: 38572071 PMCID: PMC10988781 DOI: 10.3389/fncel.2024.1343745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/19/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction Platelet-activating factor (PAF), PAF receptor (PAFR), and PAF- synthesis/degradation systems are involved in essential CNS processes such as neuroblast proliferation, differentiation, migration, and synaptic modulation. The retina is an important central nervous system (CNS) tissue for visual information processing. During retinal development, the balance between Retinal Progenitor Cell (RPC) proliferation and differentiation is crucial for proper cell determination and retinogenesis. Despite its importance in retinal development, the effects of PAFR deletion on RPC dynamics are still unknown. Methods We compared PAFR knockout mice (PAFR-/-) retinal postnatal development proliferation and differentiation aspects with control animals. Electrophysiological responses were analyzed by electroretinography (ERG). Results and discussion In this study, we demonstrate that PAFR-/- mice increased proliferation during postnatal retinogenesis and altered the expression of specific differentiation markers. The retinas of postnatal PAFR-/- animals decreased neuronal differentiation and synaptic transmission markers, leading to differential responses to light stimuli measured by ERG. Our findings suggest that PAFR signaling plays a critical role in regulating postnatal RPC cell differentiation dynamics during retinal development, cell organization, and neuronal circuitry formation.
Collapse
Affiliation(s)
- Barbara Dalmaso
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo (ICB-USP), São Paulo, Brazil
| | - Andre Mauricio Passos Liber
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, France
- Department of Experimental Psychology, Institute of Psychology, University of São Paulo (IP-USP), São Paulo, Brazil
| | - Dora Fix Ventura
- Department of Experimental Psychology, Institute of Psychology, University of São Paulo (IP-USP), São Paulo, Brazil
| | - Sonia Jancar
- Department of Immunology, Biomedical Sciences Institute, University of São Paulo (ICB-USP), São Paulo, Brazil
| | - Carolina Beltrame Del Debbio
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo (ICB-USP), São Paulo, Brazil
| |
Collapse
|
2
|
Tremblay MÈ, Almsherqi ZA, Deng Y. Plasmalogens and platelet-activating factor roles in chronic inflammatory diseases. Biofactors 2022; 48:1203-1216. [PMID: 36370412 DOI: 10.1002/biof.1916] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022]
Abstract
Fatty acids and phospholipid molecules are essential for determining the structure and function of cell membranes, and they hence participate in many biological processes. Platelet activating factor (PAF) and its precursor plasmalogen, which represent two subclasses of ether phospholipids, have attracted increasing research attention recently due to their association with multiple chronic inflammatory, neurodegenerative, and metabolic disorders. These pathophysiological conditions commonly involve inflammatory processes linked to an excess presence of PAF and/or decreased levels of plasmalogens. However, the molecular mechanisms underlying the roles of plasmalogens in inflammation have remained largely elusive. While anti-inflammatory responses most likely involve the plasmalogen signal pathway; pro-inflammatory responses recruit arachidonic acid, a precursor of pro-inflammatory lipid mediators which is released from membrane phospholipids, notably derived from the hydrolysis of plasmalogens. Plasmalogens per se are vital membrane phospholipids in humans. Changes in their homeostatic levels may alter cell membrane properties, thus affecting key signaling pathways that mediate inflammatory cascades and immune responses. The plasmalogen analogs of PAF are also potentially important, considering that anti-PAF activity has strong anti-inflammatory effects. Plasmalogen replacement therapy was further identified as a promising anti-inflammatory strategy allowing for the relief of pathological hallmarks in patients affected by chronic diseases with an inflammatory component. The aim of this Short Review is to highlight the emerging roles and implications of plasmalogens in chronic inflammatory disorders, along with the promising outcomes of plasmalogen replacement therapy for the treatment of various PAF-related chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, Canada
- Department of Molecular Medicine, Université de Laval, Québec City, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
| | - Zakaria A Almsherqi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| |
Collapse
|
3
|
Gao Y, Hai L, Kang Y, Qin W, Liu F, Cai R, Yang X, Qi Y. Compound Kushen Injection Induces Immediate Hypersensitivity Reaction Through Promoting the Production of Platelet-Activating Factor via de Novo Pathway. Front Pharmacol 2021; 12:768643. [PMID: 34690789 PMCID: PMC8531113 DOI: 10.3389/fphar.2021.768643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
Compound Kushen Injection (CKI) is a bis-herbal formulation extracted from Kushen (Radix Sophorae Flavescentis) and Baituling (Rhizoma Heterosmilacis Yunnanensis). Clinically, it is used as the adjuvant treatment of cancer. However, with the increased application, the cases of immediate hypersensitivity reactions (IHRs) also gradually rise. In this study, we investigated the underlying mechanism(s) and active constituent(s) for CKI-induced IHRs in experimental models. The obtained results showed that CKI did not elevate serum total IgE (tIgE) and mouse mast cell protease 1 (MMCP1) after consecutive immunization for 5 weeks, but could induce Evans blue extravasation (local) and cause obvious hypothermia (systemic) after a single injection. Further study showed that alkaloids in Kushen, especially matrine, were responsible for CKI-induced IHRs. Mechanism study showed that various platelet-activating factor (PAF) receptor antagonists could significantly counter CKI-induced IHRs locally or systemically. In cell system, CKI was able to promote PAF production in a non-cell-selective manner. In cell lysate, the effect of CKI on PAF production became stronger and could be abolished by blocking de novo pathway. In conclusion, our study identifies, for the first time, that CKI is a PAF inducer. It causes non-immunologic IHRs, rather than IgE-dependent IHRs, by promoting PAF production through de novo pathway. Alkaloids in Kushen, especially matrine, are the prime culprits for IHRs. Our findings may provide a potential approach for preventing and treating CKI-induced IHRs.
Collapse
Affiliation(s)
- Yuan Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lina Hai
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.,Beijing Zhendong Guangming Pharmaceutical Research Institute, Beijing, China
| | - Yuan Kang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenjie Qin
- Beijing Zhendong Guangming Pharmaceutical Research Institute, Beijing, China
| | - Fang Liu
- Beijing Zhendong Guangming Pharmaceutical Research Institute, Beijing, China
| | - Runlan Cai
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiuwei Yang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.,State Key Laboratory of Natural and Biomimetic Drugs, Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yun Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Cai J, Li L, Song Y, Xu L, Mao Y, Wang H. Neuroprotective Effect of Brimonidine against Facial Nerve Crush Injury in Rats via Suppressing GFAP/PAF Activation and Neuroinflammation. ORL J Otorhinolaryngol Relat Spec 2021; 83:449-456. [PMID: 33965946 DOI: 10.1159/000514994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/02/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE This study aimed to investigate the potential neuroprotective action of brimonidine against facial nerve crush injury in rats and the possible underlying mechanisms. METHODS Sixty Wistar adult rats were randomly and equally divided into 3 groups: 40 rats underwent unilateral facial nerve crush injury and were administered with either saline (intraperitoneal, n = 20) or brimonidine 1 mg/kg/day (intraperitoneal, n = 20) for 5 consecutive days. Functional and electromyographic recovery was recorded postoperatively. The facial nucleus of 5 mice in each group was analyzed for mRNA expression levels of GFAP, PAF, NT-4, P75NTR, NF-κB, TNF-α, IL-6, and α2-ARs by qRT-PCR. RESULTS Brimonidine promoted the recovery of vibrissae movement, eyelid closure, and electrophysiological function in a rat model of nerve crush injury. Hematoxylin and eosin staining and electron microscopy showed significant recovery of Schwann cells and axons in the brimonidine group. Brimonidine attenuated the crush-induced upregulation in GFAP and PAF mRNA (p < 0.05), as well as enhanced the mRNA levels of NT-4 and P75NTR (p < 0.05), while decreased the expression of NF-κB, TNF-α and IL-6 (p < 0.05). CONCLUSIONS Brimonidine could promote the recovery of facial nerve crush injury in rats via suppressing of GFAP/PAF activation and neuroinflammation and increasing neurotrophic factors. Brimonidine may be apromising candidate agent for the treatment of facial nerve injury.
Collapse
Affiliation(s)
- Jing Cai
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liheng Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Otolaryngology, The First Affiliated Hospital of Huzhou University, Zhejiang, China
| | - Yongdong Song
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanyan Mao
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haibo Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
5
|
Dalmaso B, da Silva-Junior IA, Fragel-Madeira L, Jancar S, Del Debbio CB. Platelet activating factor in the eye: Physiological roles, diseases and future perspectives. Prostaglandins Other Lipid Mediat 2021; 153:106522. [PMID: 33358892 DOI: 10.1016/j.prostaglandins.2020.106522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/03/2020] [Accepted: 12/18/2020] [Indexed: 01/09/2023]
Abstract
Platelet Activating Factor (PAF) is a known phospholipid mediator of inflammation. Since its first description in 1972, it has emerged as a key regulator of vital cellular signaling functions, as proliferation, cell adhesion, and apoptosis. Evidence suggests that interactions between PAF and its receptor (PAFR) play a critical role in nervous system tissues, including the retina. The retina is a very important constituent of the visual system, along with the cornea, sclera, choroid, iris, and ciliary body, that acts synergistically to provide vision and to maintain optical homeostasis. There is evidence that PAF may regulate a wide range of physiological functions in the visual system tissues, such as eye development, inflammation, epithelial wound healing, and synapsis. Due to their multiple functions, PAF and PAFR also have important pathological and clinical implications in ocular disorders such as Choroidal Neovascularization (CNV), Age Macular Degeneration, (AMD), Diabetic Retinopathy (DR), transplant responses, and pharmacological interactions. Studies with PAFR antagonists have shown promising results such as inhibition of neovascularization and chloroquine-induced retinopathies, as well as reducing inflammation and retinal cell death. Due to the importance of PAFR signaling in the visual system and ophthalmology research, this review aims to provide a general overview of current and future perspectives about PAF in eye biology.
Collapse
Affiliation(s)
- Barbara Dalmaso
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of Sao Paulo, São Paulo, Brazil
| | | | - Lucianne Fragel-Madeira
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Sonia Jancar
- Department of Immunology, Biomedical Sciences Institute, University of Sao Paulo, São Paulo, Brazil
| | - Carolina Beltrame Del Debbio
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of Sao Paulo, São Paulo, Brazil.
| |
Collapse
|
6
|
Travers JB, Rohan JG, Sahu RP. New Insights Into the Pathologic Roles of the Platelet-Activating Factor System. Front Endocrinol (Lausanne) 2021; 12:624132. [PMID: 33796070 PMCID: PMC8008455 DOI: 10.3389/fendo.2021.624132] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022] Open
Abstract
Described almost 50 years ago, the glycerophosphocholine lipid mediator Platelet-activating factor (PAF) has been implicated in many pathologic processes. Indeed, elevated levels of PAF can be measured in response to almost every type of pathology involving inflammation and cell damage/death. In this review, we provide evidence for PAF involvement in pathologic processes, with focus on cancer, the nervous system, and in photobiology. Importantly, recent insights into how PAF can generate and travel via bioactive extracellular vesicles such as microvesicle particles (MVP) are presented. What appears to be emerging from diverse pathologies in different organ systems is a common theme where pro-oxidative stressors generate oxidized glycerophosphocholines with PAF agonistic effects, which then trigger more enzymatic PAF synthesis via the PAF receptor. A downstream consequence of PAF receptor activation is the generation and release of MVP which provide a mechanism to transmit PAF as well as other bioactive agents. The knowledge gaps which when addressed could result in novel therapeutic strategies are also discussed. Taken together, an enhanced understanding of the PAF family of lipid mediators is essential in our improved comprehension of the relationship amongst the diverse cutaneous, cancerous, neurologic and systemic pathologic processes.
Collapse
Affiliation(s)
- Jeffrey B. Travers
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
- Department of Dermatology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
- Dayton Veterans Administration Medical Center, Dayton, OH, United States
- *Correspondence: Jeffrey B. Travers, ; orcid.org/0000-0001-7232-1039
| | - Joyce G. Rohan
- Naval Medical Research Unit Dayton, Environmental Health Effects Directorate, Wright Patterson Air Force Base, OH, United States
| | - Ravi P. Sahu
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
| |
Collapse
|
7
|
Tian Z, Chu T, Shields LBE, Zhu Q, Zhang YP, Kong M, Barnes GN, Wang Y, Shields CB, Cai J. Platelet-Activating Factor Deteriorates Lysophosphatidylcholine-Induced Demyelination Via Its Receptor-Dependent and -Independent Effects. Mol Neurobiol 2020; 57:4069-4081. [PMID: 32661728 DOI: 10.1007/s12035-020-02003-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 06/26/2020] [Indexed: 11/30/2022]
Abstract
Accumulating evidence suggests that platelet-activating factor (PAF) increases the inflammatory response in demyelinating diseases such as multiple sclerosis. However, PAF receptor (PAFR) antagonists do not show therapeutic efficacy for MS, and its underlying mechanisms remain poorly understood. In the present study, we investigated the effects of PAF on an ex vivo demyelination cerebellar model following lysophosphatidylcholine (LPC, 0.5 mg/mL) application using wild-type and PAFR conventional knockout (PAFR-KO) mice. Demyelination was induced in cerebellar slices that were cultured with LPC for 18 h. Exogenous PAF (1 μM) acting on cerebellar slices alone did not cause demyelination but increased the severity of LPC-induced demyelination in both wild-type and PAFR-KO mice. LPC inhibited the expression of PAF-AH, MBP, TNF-α, and TGF-β1 but facilitated the expression of IL-1β and IL-6 in wild-type preparations. Of note, exogenous PAF stimulated microglial activation in both wild-type and PAFR-KO mice. The subsequent inflammatory cytokines TNFα, IL-1β, and IL-6 as well as the anti-inflammatory cytokine TGF-β1 demonstrated a diverse transcriptional profile with or without LPC treatment. PAF promoted TNF-α expression and suppressed TGF-β1 expression indiscriminately in wild-type and knockout slices; however, transcription of IL-1β and IL-6 was not significantly affected in both slices. The syntheses of IL-1β and IL-6 were significantly increased in LPC-induced demyelination preparations without PAF but showed a redundancy in PAF-treated wild-type and knockout slices. These data suggest that PAF can play a detrimental role in LPC-induced demyelination probably due to a redundant response of PAFR-dependent and PAFR-independent effects on inflammatory cytokines.
Collapse
Affiliation(s)
- Zhisen Tian
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Tianci Chu
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Lisa B E Shields
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, 40202, USA
| | - Qingsan Zhu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China.
| | - Yi Ping Zhang
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, 40202, USA
| | - Maiying Kong
- Department of Bioinformatics and Biostatistics, University of Louisville School of Public Health & Information Sciences, Louisville, KY, 40202, USA
| | - Gregory N Barnes
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, 40202, USA.,Department of Neurology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Yuanyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China.
| | - Christopher B Shields
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, 40202, USA.,Department of Neurosurgery, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Jun Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, 40202, USA. .,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
8
|
Joensuu M, Wallis TP, Saber SH, Meunier FA. Phospholipases in neuronal function: A role in learning and memory? J Neurochem 2020; 153:300-333. [PMID: 31745996 DOI: 10.1111/jnc.14918] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/29/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022]
Abstract
Despite the human brain being made of nearly 60% fat, the vast majority of studies on the mechanisms of neuronal communication which underpin cognition, memory and learning, primarily focus on proteins and/or (epi)genetic mechanisms. Phospholipids are the main component of all cellular membranes and function as substrates for numerous phospholipid-modifying enzymes, including phospholipases, which release free fatty acids (FFAs) and other lipid metabolites that can alter the intrinsic properties of the membranes, recruit and activate critical proteins, and act as lipid signalling molecules. Here, we will review brain specific phospholipases, their roles in membrane remodelling, neuronal function, learning and memory, as well as their disease implications. In particular, we will highlight key roles of unsaturated FFAs, particularly arachidonic acid, in neurotransmitter release, neuroinflammation and memory. In light of recent findings, we will also discuss the emerging role of phospholipase A1 and the creation of saturated FFAs in the brain.
Collapse
Affiliation(s)
- Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia.,Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Tristan P Wallis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Saber H Saber
- Laboratory of Molecular Cell Biology, Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
9
|
Forty Years Since the Structural Elucidation of Platelet-Activating Factor (PAF): Historical, Current, and Future Research Perspectives. Molecules 2019; 24:molecules24234414. [PMID: 31816871 PMCID: PMC6930554 DOI: 10.3390/molecules24234414] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
In the late 1960s, Barbaro and Zvaifler described a substance that caused antigen induced histamine release from rabbit platelets producing antibodies in passive cutaneous anaphylaxis. Henson described a ‘soluble factor’ released from leukocytes that induced vasoactive amine release in platelets. Later observations by Siraganuan and Osler observed the existence of a diluted substance that had the capacity to cause platelet activation. In 1972, the term platelet-activating factor (PAF) was coined by Benveniste, Henson, and Cochrane. The structure of PAF was later elucidated by Demopoulos, Pinckard, and Hanahan in 1979. These studies introduced the research world to PAF, which is now recognised as a potent phospholipid mediator. Since its introduction to the literature, research on PAF has grown due to interest in its vital cell signalling functions and more sinisterly its role as a pro-inflammatory molecule in several chronic diseases including cardiovascular disease and cancer. As it is forty years since the structural elucidation of PAF, the aim of this review is to provide a historical account of the discovery of PAF and to provide a general overview of current and future perspectives on PAF research in physiology and pathophysiology.
Collapse
|
10
|
Hyland IK, O'Toole RF, Smith JA, Bissember AC. Progress in the Development of Platelet-Activating Factor Receptor (PAFr) Antagonists and Applications in the Treatment of Inflammatory Diseases. ChemMedChem 2018; 13:1873-1884. [DOI: 10.1002/cmdc.201800401] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/08/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Isabel K. Hyland
- School of Natural Sciences Chemistry; University of Tasmania; Hobart Australia
| | | | - Jason A. Smith
- School of Natural Sciences Chemistry; University of Tasmania; Hobart Australia
| | - Alex C. Bissember
- School of Natural Sciences Chemistry; University of Tasmania; Hobart Australia
| |
Collapse
|
11
|
Tsoupras A, Lordan R, Zabetakis I. Inflammation, not Cholesterol, Is a Cause of Chronic Disease. Nutrients 2018; 10:E604. [PMID: 29757226 PMCID: PMC5986484 DOI: 10.3390/nu10050604] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 12/17/2022] Open
Abstract
Since the Seven Countries Study, dietary cholesterol and the levels of serum cholesterol in relation to the development of chronic diseases have been somewhat demonised. However, the principles of the Mediterranean diet and relevant data linked to the examples of people living in the five blue zones demonstrate that the key to longevity and the prevention of chronic disease development is not the reduction of dietary or serum cholesterol but the control of systemic inflammation. In this review, we present all the relevant data that supports the view that it is inflammation induced by several factors, such as platelet-activating factor (PAF), that leads to the onset of cardiovascular diseases (CVD) rather than serum cholesterol. The key to reducing the incidence of CVD is to control the activities of PAF and other inflammatory mediators via diet, exercise, and healthy lifestyle choices. The relevant studies and data supporting these views are discussed in this review.
Collapse
Affiliation(s)
- Alexandros Tsoupras
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland.
| | - Ronan Lordan
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland.
| | - Ioannis Zabetakis
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland.
| |
Collapse
|
12
|
Brailoiu E, Barlow CL, Ramirez SH, Abood ME, Brailoiu GC. Effects of Platelet-Activating Factor on Brain Microvascular Endothelial Cells. Neuroscience 2018. [PMID: 29522856 DOI: 10.1016/j.neuroscience.2018.02.039] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Platelet-activating factor (PAF) is a potent phospholipid mediator that exerts various pathophysiological effects by interacting with a G protein-coupled receptor. PAF has been reported to increase the permeability of the blood-brain barrier (BBB) via incompletely characterized mechanisms. We investigated the effect of PAF on rat brain microvascular endothelial cells (RBMVEC), a critical component of the BBB. PAF produced a dose-dependent increase in cytosolic Ca2+ concentration; the effect was prevented by the PAF receptor antagonist, WEB2086. The effect of PAF on cytosolic Ca2+ was abolished in Ca2+-free saline or in the presence of L-type voltage-gated Ca2+ channel inhibitor, nifedipine, indicating that Ca2+ influx is critical for PAF-induced increase in cytosolic Ca2+. PAF produced RBMVEC depolarization; the effect was inhibited by WEB2086. In cells loaded with [(4-amino-5-methylamino-2',7'-difluoro-fluorescein)diacetate] (DAF-FM), a nitric oxide (NO)-sensitive fluorescent dye, PAF increased the NO level; the effect was prevented by WEB2086, nifedipine or by l-NAME, an inhibitor of NO synthase. Immunocytochemistry studies indicate that PAF reduced the immunostaining of ZO-1, a tight junction-associated protein, increased F-actin fibers, and produced intercellular gaps. PAF produced a decrease in RBMVEC monolayer electrical resistance assessed with Electric Cell-Substrate Impedance Sensing (ECIS), indicative of a disruption of endothelial barrier function. In vivo studies indicate that PAF increased the BBB permeability, assessed with sodium fluorescein and Evans Blue methods, via PAF receptor-dependent mechanisms, consequent to Ca2+ influx and increased NO levels. Our studies reveal that PAF alters the BBB permeability by multiple mechanisms, which may be relevant for central nervous system (CNS) inflammatory disorders.
Collapse
Affiliation(s)
- Eugen Brailoiu
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Philadelphia, PA 19140, United States
| | - Christine L Barlow
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Philadelphia, PA 19107, United States
| | - Servio H Ramirez
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Philadelphia, PA 19140, United States; Department of Pathology & Laboratory Medicine, Lewis Katz School of Medicine, Philadelphia, PA 19140, United States
| | - Mary E Abood
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Philadelphia, PA 19140, United States
| | - G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Philadelphia, PA 19107, United States.
| |
Collapse
|
13
|
Li ZY, Chung YH, Shin EJ, Dang DK, Jeong JH, Ko SK, Nah SY, Baik TG, Jhoo JH, Ong WY, Nabeshima T, Kim HC. YY-1224, a terpene trilactone-strengthened Ginkgo biloba, attenuates neurodegenerative changes induced by β-amyloid (1-42) or double transgenic overexpression of APP and PS1 via inhibition of cyclooxygenase-2. J Neuroinflammation 2017; 14:94. [PMID: 28449688 PMCID: PMC5408406 DOI: 10.1186/s12974-017-0866-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/18/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Ginkgo biloba has been reported to possess free radical-scavenging antioxidant activity and anti-inflammatory properties. In our pilot study, YY-1224, a terpene trilactone-strengthened extract of G. biloba, showed anti-inflammatory, neurotrophic, and antioxidant effects. RESULTS We investigated the pharmacological potential of YY-1224 in β-amyloid (Aβ) (1-42)-induced memory impairment using cyclooxygenase-2 (COX-2) knockout (-/-) and APPswe/PS1dE9 transgenic (APP/PS1 Tg) mice. Repeated treatment with YY-1224 significantly attenuated Aβ (1-42)-induced memory impairment in COX-2 (+/+) mice, but not in COX-2 (-/-) mice. YY-1224 significantly attenuated Aβ (1-42)-induced upregulation of platelet-activating factor (PAF) receptor gene expression, reactive oxygen species, and pro-inflammatory factors. In addition, YY-1224 significantly inhibited Aβ (1-42)-induced downregulation of PAF-acetylhydrolase-1 (PAF-AH-1) and peroxisome proliferator-activated receptor γ (PPARγ) gene expression. These changes were more pronounced in COX-2 (+/+) mice than in COX-2 (-/-) mice. YY-1224 significantly attenuated learning impairment, Aβ deposition, and pro-inflammatory microglial activation in APP/PS1 Tg mice, whereas it significantly enhanced PAF-AH and PPARγ expression. A preferential COX-2 inhibitor, meloxicam, did not affect the pharmacological activity by YY-1224, suggesting that the COX-2 gene is a critical mediator of the neuroprotective effects of YY-1224. The protective activity of YY-1224 appeared to be more efficacious than a standard G. biloba extract (Gb) against Aβ insult. CONCLUSIONS Our results suggest that the protective effects of YY-1224 against Aβ toxicity may be associated with its PAF antagonistic- and PPARγ agonistic-potential as well as inhibition of the Aβ-mediated pro-inflammatory switch of microglia phenotypes through suppression of COX-2 expression.
Collapse
Affiliation(s)
- Zheng-Yi Li
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341 Republic of Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, 06974 Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341 Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341 Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, 06974 Republic of Korea
| | - Sung Kwon Ko
- Department of Oriental Medical Food and Nutrition, Semyung University, Jecheon, 27136 Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, 05029 Republic of Korea
| | - Tae Gon Baik
- R&D Center, Yuyu Pharma, Seoul, 04598 Republic of Korea
| | - Jin Hyeong Jhoo
- Department of Psychiatry, Medical School, Kangwon National University, Chunchon, 24341 Republic of Korea
| | - Wei-Yi Ong
- Department of Anatomy, National University of Singapore, Singapore, 119260 Singapore
| | - Toshitaka Nabeshima
- Nabeshima Laboratory, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, 468-8503 Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341 Republic of Korea
| |
Collapse
|
14
|
Liu Y, Shields LBE, Gao Z, Wang Y, Zhang YP, Chu T, Zhu Q, Shields CB, Cai J. Current Understanding of Platelet-Activating Factor Signaling in Central Nervous System Diseases. Mol Neurobiol 2016; 54:5563-5572. [PMID: 27613281 DOI: 10.1007/s12035-016-0062-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/17/2016] [Indexed: 12/13/2022]
Abstract
Platelet-activating factor (PAF) is a bioactive lipid mediator which serves as a reciprocal messenger between the immune and nervous systems. PAF, a pluripotent inflammatory mediator, is extensively expressed in many cells and tissues and has either beneficial or detrimental effects on the progress of inflammation-related neuropathology. Its wide distribution and various biological functions initiate a cascade of physiological or pathophysiological responses during development or diseases. Current evidence indicates that excess PAF accumulation in CNS diseases exacerbates the inflammatory response and pathological consequences, while application of PAF inhibitors or PAFR antagonists by blocking this signaling pathway significantly reduces inflammation, protects cells, and improves the recovery of neural functions. In this review, we integrate the current findings of PAF signaling in CNS diseases and elucidate topics less appreciated but important on the role of PAF signaling in neurological diseases. We propose that the precise use of PAF inhibitors or PAFR antagonists that target the specific neural cells during the appropriate temporal window may constitute a potential therapy for CNS diseases.
Collapse
Affiliation(s)
- Yulong Liu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA
| | - Lisa B E Shields
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, 40202, USA
| | - Zhongwen Gao
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA
| | - Yuanyi Wang
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA
- Department of Spine Surgery, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Yi Ping Zhang
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, 40202, USA
| | - Tianci Chu
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA
| | - Qingsan Zhu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China.
| | | | - Jun Cai
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA.
| |
Collapse
|
15
|
Toscano ECDB, Silva BC, Victoria ECG, Cardoso ACDS, Miranda ASD, Sugimoto MA, Sousa LP, Carvalho BAD, Kangussu LM, Silva DGD, Rodrigues FG, Barcelos LDS, Vasconcelos AC, Amaral FA, Teixeira MM, Teixeira AL, Rachid MA. Platelet-activating factor receptor (PAFR) plays a crucial role in experimental global cerebral ischemia and reperfusion. Brain Res Bull 2016; 124:55-61. [PMID: 27040712 DOI: 10.1016/j.brainresbull.2016.03.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 11/19/2022]
Abstract
Stroke is one of the most frequent causes of death and disability worldwide leading to a significant clinical and socioeconomic burden. Although different mechanisms are involved in the pathogenesis of stroke, inflammatory response occurs after ischemia and contributes to the expansion of brain injury. Platelet-activating factor receptor (PAF) plays crucial roles in both physiological and pathological conditions in the brain. PAF receptor (PAFR) may be expressed on cellular and nuclear membranes of various cell types, especially leukocytes, platelets, endothelial cells, neuronal cells and microglia. Herein, using mice lacking the PAFR receptor (PAFR(-/-)), we investigate a potential role for this receptor during experimental transient global cerebral ischemia and reperfusion (BCCAo). In PAFR deficiency, we observed a significant improvement in the neurological deficits, which were associated with a reduction of brain infarcted area as evaluated by triphenyltetrazolium chloride (TTC). Moreover, a decrease in the percentage of necrotic cavities areas and in the frequency of ischemic neurons was also found by employing histometric analysis. In addition, in PAFR(-/-) mice there was prevention of caspase-3 activation and decreased vascular permeability and brain edema. Decreased brain levels of the cytokines tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta) and the chemokine (C-X-C motif) ligand 1 (CXCL1) by ELISA were also detected in PAFR(-/-) BCCAo animals. Taken together, our results suggest that PAFR activation might be crucial for the global brain ischemia and reperfusion injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Flávia Guimarães Rodrigues
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | | | | | - Flávio Almeida Amaral
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, School of Medicine, University of Texas Health Science Center at Houston, TX, United States
| | | |
Collapse
|
16
|
Reiner B, Wang W, Liu J, Xiong H. Platelet-activating factor attenuation of long-term potentiation in rat hippocampal slices via protein tyrosine kinase signaling. Neurosci Lett 2016; 615:83-7. [PMID: 26808643 DOI: 10.1016/j.neulet.2016.01.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 02/08/2023]
Abstract
It is well established that HIV-1-infected mononuclear phagocytes release platelet activating factor (PAF) and elevated levels of PAF have been detected in blood and in the cerebrospinal fluid (CSF) of acquired immunodeficiency syndrome (AIDS) patients with HIV-associated neurocognitive disorders (HAND). It is our hypothesis that the elevated levels of PAF alter long-term potentiation (LTP) in the hippocampus, leading to neurocognitive dysfunction. To test this hypothesis, we studied the effects of PAF on LTP in the CA1 region of rat hippocampal slices. Our results showed incubation of hippocampal slices with PAF attenuated LTP. The PAF-mediated attenuation was blocked by ginkgolide B, a PAF receptor antagonist, suggesting PAF attenuation of LTP via PAF receptors. Application of lyso-PAF, an inactive PAF analog, had no apparent effect on LTP. Further investigation revealed an involvement of tyrosine kinase in PAF attenuation of LTP, which was demonstrated by lavendustin A (a specific protein tyrosine kinase inhibitor) blockage of PAF attenuation of LTP. As LTP is widely considered as the cellular and synaptic basis for learning and memory, the attenuation of LTP by PAF may contribute at least in part to the HAND pathogenesis.
Collapse
Affiliation(s)
- Benjamin Reiner
- The Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Biology, West Chester University of Pennsylvania, West Chester, PA 19383, USA
| | - Wenwei Wang
- The Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Physiology, Fudan University School of Medicine, Shanghai, China
| | - Jianuo Liu
- The Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Huangui Xiong
- The Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| |
Collapse
|
17
|
Hammond JW, Lu SM, Gelbard HA. Platelet Activating Factor Enhances Synaptic Vesicle Exocytosis Via PKC, Elevated Intracellular Calcium, and Modulation of Synapsin 1 Dynamics and Phosphorylation. Front Cell Neurosci 2016; 9:505. [PMID: 26778968 PMCID: PMC4705275 DOI: 10.3389/fncel.2015.00505] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/15/2015] [Indexed: 11/25/2022] Open
Abstract
Platelet activating factor (PAF) is an inflammatory phospholipid signaling molecule implicated in synaptic plasticity, learning and memory and neurotoxicity during neuroinflammation. However, little is known about the intracellular mechanisms mediating PAF’s physiological or pathological effects on synaptic facilitation. We show here that PAF receptors are localized at the synapse. Using fluorescent reporters of presynaptic activity we show that a non-hydrolysable analog of PAF (cPAF) enhances synaptic vesicle release from individual presynaptic boutons by increasing the size or release of the readily releasable pool and the exocytosis rate of the total recycling pool. cPAF also activates previously silent boutons resulting in vesicle release from a larger number of terminals. The underlying mechanism involves elevated calcium within presynaptic boutons and protein kinase C activation. Furthermore, cPAF increases synapsin I phosphorylation at sites 1 and 3, and increases dispersion of synapsin I from the presynaptic compartment during stimulation, freeing synaptic vesicles for subsequent release. These findings provide a conceptual framework for how PAF, regardless of its cellular origin, can modulate synapses during normal and pathologic synaptic activity.
Collapse
Affiliation(s)
- Jennetta W Hammond
- Center for Neural Development and Disease, University of Rochester Rochester, NY, USA
| | - Shao-Ming Lu
- Center for Neural Development and Disease, University of Rochester Rochester, NY, USA
| | - Harris A Gelbard
- Center for Neural Development and Disease, University of Rochester Rochester, NY, USA
| |
Collapse
|
18
|
Wang Y, Gao Z, Zhang Y, Feng SQ, Liu Y, Shields LBE, Zhao YZ, Zhu Q, Gozal D, Shields CB, Cai J. Attenuated Reactive Gliosis and Enhanced Functional Recovery Following Spinal Cord Injury in Null Mutant Mice of Platelet-Activating Factor Receptor. Mol Neurobiol 2015; 53:3448-3461. [PMID: 26084439 DOI: 10.1007/s12035-015-9263-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/28/2015] [Indexed: 12/11/2022]
Abstract
Platelet-activating factor (PAF) is a unique phosphoglycerine that mediates the biological functions of both immune and nervous systems. Excessive PAF plays an important role in neural injury via its specific receptor (PAFR). In this study, we hypothesized that PAF signaling activates reactive gliosis after spinal cord injury (SCI), and blocking the PAF pathway would modify the glia scar formation and promote functional recovery. PAF microinjected into the normal wild-type spinal cord induced a dose-dependent activation of microglia and astrocytes. In the SCI mice, PAFR null mutant mice showed a better functional recovery in grip and rotarod performances than wild-type mice. Although both microglia and astrocytes were activated after SCI in wild-type and PAFR null mutant mice, expressions of IL-6, vimentin, nestin, and GFAP were not significantly elevated in PAFR null mutants. Disruption of PAF signaling inhibited the expressions of proteoglycan CS56 and neurocan (CSPG3). Intriguingly, compared to the wild-type SCI mice, less axonal retraction/dieback at 7 dpi but more NFH-labeled axons at 28 dpi was found in the area adjacent to the epicenter in PAFR null mutant SCI mice. Moreover, treatment with PAFR antagonist Ginkgolide B (GB) at the chronic phase rather than acute phase enhanced the functional recovery in the wild-type SCI mice. These findings suggest that PAF signaling participates in reactive gliosis after SCI, and blocking of this signaling enhances functional recovery and to some extent may promote axon regrowth.
Collapse
Affiliation(s)
- Yuanyi Wang
- Department of Spine Surgery, First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China.,Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA
| | - Zhongwen Gao
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA.,Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Yiping Zhang
- Norton Healthcare, Norton Neuroscience Institute, Louisville, KY, 40202, USA
| | - Shi-Qing Feng
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA.,Department of Orthopedics, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Yulong Liu
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA.,Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Lisa B E Shields
- Norton Healthcare, Norton Neuroscience Institute, Louisville, KY, 40202, USA
| | - Ying-Zheng Zhao
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA.,Pharmacy School, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qingsan Zhu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - David Gozal
- Comer Children's Hospital, Department of Pediatrics, University of Chicago, Chicago, IL, 60637, USA
| | - Christopher B Shields
- Norton Healthcare, Norton Neuroscience Institute, Louisville, KY, 40202, USA.,Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Jun Cai
- Department of Pediatrics, University of Louisville School of Medicine, 570 S. Preston Street, Donald Baxter Building, Suite 321B, Louisville, KY, 40202, USA. .,Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
19
|
Singh P, Singh IN, Mondal SC, Singh L, Garg VK. Platelet-activating factor (PAF)-antagonists of natural origin. Fitoterapia 2012; 84:180-201. [PMID: 23160091 DOI: 10.1016/j.fitote.2012.11.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 10/30/2012] [Accepted: 11/04/2012] [Indexed: 01/06/2023]
Abstract
Presently herbal medicines are being used by about 80% of the world population for primary health care as they stood the test of time for their safety, efficacy, cultural acceptability and lesser side effects. The discovery of platelet activating factor antagonists (PAF antagonists) during these decades are going on with different framework, but the researchers led their efficiency in studying in vitro test models. Since it is assumed that PAF play a central role in etiology of many diseases in humans such as asthma, neuronal damage, migraine, cardiac diseases, inflammatory, headache etc. Present days instinctively occurring PAF antagonist exists as a specific grade of therapeutic agents for the humans against these and different diseases either laid hold of immunological or non-immunological types. Ginkgolide, cedrol and many other natural PAF antagonists such as andrographolide, α-bulnesene, cinchonine, piperine, kadsurenone, different Piper species' natural products and marine origin plants extracts or even crude drugs having PAF antagonist properties are being used currently against different inflammatory pathologies. This review is an attempt to summarize the data on PAF and action of natural PAF antagonists on it, which were evaluated by in vivo and in vitro assays.
Collapse
Affiliation(s)
- Preeti Singh
- Department of Pharmacology, R.V.N.I. Dadri, Greater Noida, 201301, Uttar Pradesh, India.
| | | | | | | | | |
Collapse
|
20
|
Ha SW, Lee CS. Differential effect of platelet activating factor on 1-methyl-4-phenylpyridinium-induced cell death through regulation of apoptosis-related protein activation. Neurochem Int 2010; 56:819-28. [DOI: 10.1016/j.neuint.2010.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 03/10/2010] [Accepted: 03/12/2010] [Indexed: 11/27/2022]
|
21
|
Umemura K, Kato I, Hirashima Y, Ishii Y, Inoue T, Aoki J, Kono N, Oya T, Hayashi N, Hamada H, Endo S, Oda M, Arai H, Kinouchi H, Hiraga K. Neuroprotective Role of Transgenic PAF-Acetylhydrolase II in Mouse Models of Focal Cerebral Ischemia. Stroke 2007; 38:1063-8. [PMID: 17272759 DOI: 10.1161/01.str.0000257981.09329.d2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose—
Platelet-activating factor (PAF) and oxidized unsaturated free fatty acids have been postulated to aggravate neuronal damage in the postischemic brain. Type II PAF-acetylhydrolase (PAF-AH II) not only terminates signals by PAF by its PAF-hydrolyzing activity but also protects cells against oxidative stress. We examined whether PAF-AH II can rescue cerebral neurons against ischemic insults.
Methods—
Transgenic mice overexpressing human PAF-AH II in neurons were generated and enzyme expressions were examined biochemically and histochemically. The mice were subjected to 60 minutes of transient middle cerebral artery occlusion followed by reperfusion for 24 hours. The infarction and apoptosis were estimated by TTC staining and fluorescence TUNEL staining, respectively.
Results—
Overexpression of PAF-AH II was found in brains of transgenic mice by Western blot and enzymatic activity analyses. In immunohistochemistry, human PAF-AH II expression was found throughout the central nervous system, especially in neurons of neocortex, hippocampus, and basal ganglia. The neurological deficit scores, cerebral edema index, and relative infarction volume were all significantly (
P
<0.05) lower in transgenic mice (1.30±0.72, 1.12±0.04, and 14.0±7.7%, respectively) than in wild-type mice (2.56±0.93, 1.23±0.12, and 31.9±9.7%, respectively). Percentages of apoptotic cells were also significantly (
P
<0.001) lower in transgenic mice (cortex, 5.2±3.3%; hippocampus, 3.4±7.0%) than in wild-type mice (cortex, 41.1±16.9%; hippocampus, 58.9±15.3%).
Conclusions—
These results indicate that PAF-AH II exerts strong neuroprotective effects against ischemic injury and suggest a possibility for clinical use of this enzyme in cerebral ischemia.
Collapse
Affiliation(s)
- Kimiko Umemura
- Department of Neurosurgery, University of Toyama Faculty of Medicine, Toyama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Neuroinflammation is a host defense mechanism associated with neutralization of an insult and restoration of normal structure and function of brain. Neuroinflammation is a hallmark of all major CNS diseases. The main mediators of neuroinflammation are microglial cells. These cells are activated during a CNS injury. Microglial cells initiate a rapid response that involves cell migration, proliferation, release of cytokines/chemokines and trophic and/or toxic effects. Cytokines/chemokines stimulate phospholipases A2 and cyclooxygenases. This results in breakdown of membrane glycerophospholipids with the release of arachidonic acid (AA) and docosahexaenoic acid (DHA). Oxidation of AA produces pro-inflammatory prostaglandins, leukotrienes, and thromboxanes. One of the lyso-glycerophospholipids, the other products of reactions catalyzed by phospholipase A2, is used for the synthesis of pro-inflammatory platelet-activating factor. These pro-inflammatory mediators intensify neuroinflammation. Lipoxin, an oxidized product of AA through 5-lipoxygenase, is involved in the resolution of inflammation and is anti-inflammatory. Docosahexaenoic acid is metabolized to resolvins and neuroprotectins. These lipid mediators inhibit the generation of prostaglandins, leukotrienes, and thromboxanes. Levels of prostaglandins, leukotrienes, and thromboxanes are markedly increased in acute neural trauma and neurodegenerative diseases. Docosahexaenoic acid and its lipid mediators prevent neuroinflammation by inhibiting transcription factor NFkappaB, preventing cytokine secretion, blocking the synthesis of prostaglandins, leukotrienes, and thromboxanes, and modulating leukocyte trafficking. Depending on its timing and magnitude in brain tissue, inflammation serves multiple purposes. It is involved in the protection of uninjured neurons and removal of degenerating neuronal debris and also in assisting repair and recovery processes. The dietary ratio of AA to DHA may affect neurodegeneration associated with acute neural trauma and neurodegenerative diseases. The dietary intake of docosahexaenoic acid offers the possibility of counter-balancing the harmful effects of high levels of AA-derived pro-inflammatory lipid mediators.
Collapse
Affiliation(s)
- Akhlaq A Farooqui
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | |
Collapse
|
23
|
Heusler P, Boehmer G. Platelet-activating factor contributes to the induction of long-term potentiation in the rat somatosensory cortex in vitro. Brain Res 2007; 1135:85-91. [PMID: 17196945 DOI: 10.1016/j.brainres.2006.12.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Revised: 12/04/2006] [Accepted: 12/07/2006] [Indexed: 11/25/2022]
Abstract
The contribution of platelet-activating factor (PAF) to the induction of neocortical LTP was examined in rat brain slices containing the primary somatosensory cortex (SI). Field potentials evoked by single pulse stimulation in cortical layer IV were recorded from layer II/III. In control experiments, tetanic high frequency stimulation (HFS) resulted in input-specific, NMDA receptor-dependent LTP (21.1+/-3.2%; mean+/-SEM; n=15; P<0.001). BN-52021 (5 microM), an antagonist at the extracellular PAF receptor, weakened the HFS-induced LTP to 12.4+/-2.7% (n=11; P<0.05). In contrast, HFS-induced LTP was significantly enhanced to 29.4+/-2.3% (n=11; P<0.05) when brain slices were superfused with ACSF containing the PAF receptor-agonist C-PAF (1.5 microM). The difference between LTP weakened by BN-52021 and LTP enhanced by C-PAF was highly significant (P<0.0005). These results suggest a physiological contribution of PAF to the induction of LTP in neocortical area SI. This contribution of PAF is mediated by an action at extracellular receptor sites.
Collapse
Affiliation(s)
- Peter Heusler
- Institute of Physiology and Pathophysiology, Johannes Gutenberg University, Duesbergweg 6, D-55099 Mainz, Germany
| | | |
Collapse
|
24
|
Phillis JW, Horrocks LA, Farooqui AA. Cyclooxygenases, lipoxygenases, and epoxygenases in CNS: Their role and involvement in neurological disorders. ACTA ACUST UNITED AC 2006; 52:201-43. [PMID: 16647138 DOI: 10.1016/j.brainresrev.2006.02.002] [Citation(s) in RCA: 266] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2005] [Revised: 02/23/2006] [Accepted: 02/24/2006] [Indexed: 01/01/2023]
Abstract
Three enzyme systems, cyclooxygenases that generate prostaglandins, lipoxygenases that form hydroxy derivatives and leukotrienes, and epoxygenases that give rise to epoxyeicosatrienoic products, metabolize arachidonic acid after its release from neural membrane phospholipids by the action of phospholipase A(2). Lysophospholipids, the other products of phospholipase A(2) reactions, are either reacylated or metabolized to platelet-activating factor. Under normal conditions, these metabolites play important roles in synaptic function, cerebral blood flow regulation, apoptosis, angiogenesis, and gene expression. Increased activities of cyclooxygenases, lipoxygenases, and epoxygenases under pathological situations such as ischemia, epilepsy, Alzheimer's disease, Parkinson disease, amyotrophic lateral sclerosis, and Creutzfeldt-Jakob disease produce neuroinflammation involving vasodilation and vasoconstriction, platelet aggregation, leukocyte chemotaxis and release of cytokines, and oxidative stress. These are closely associated with the neural cell injury which occurs in these neurological conditions. The metabolic products of docosahexaenoic acid, through these enzymes, generate a new class of lipid mediators, namely docosatrienes and resolvins. These metabolites antagonize the effect of metabolites derived from arachidonic acid. Recent studies provide insight into how these arachidonic acid metabolites interact with each other and other bioactive mediators such as platelet-activating factor, endocannabinoids, and docosatrienes under normal and pathological conditions. Here, we review present knowledge of the functions of cyclooxygenases, lipoxygenases, and epoxygenases in brain and their association with neurodegenerative diseases.
Collapse
Affiliation(s)
- John W Phillis
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| | | | | |
Collapse
|
25
|
Alexander SPH, Mathie A, Peters JA. Platelet-activating factor. Br J Pharmacol 2006. [DOI: 10.1038/sj.bjp.0706539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
26
|
Nakanishi K. Terpene trilactones from Gingko biloba: from ancient times to the 21st century. Bioorg Med Chem 2005; 13:4987-5000. [PMID: 15990319 DOI: 10.1016/j.bmc.2005.06.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2005] [Accepted: 06/01/2005] [Indexed: 11/19/2022]
Abstract
Ginkgolides were always close to my heart. I continue to be strongly attracted to Ginkgo biloba, the ginkgolides and bilobalide. Starting in 1963, I became fascinated by these molecules while working on their isolation and structure elucidation in Sendai. Presumably, due to the ginkgolide studies, I received an invitation to join the faculty at Columbia University. After almost three decades of not touching the ginkgolide project, we have unexpectedly resumed the studies, at this time because of their enigmatic biological effects. This account is a reflection on earlier studies, as well as an outline of our current work.
Collapse
Affiliation(s)
- Koji Nakanishi
- Department of Chemistry, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
27
|
Strømgaard K, Suehiro M, Nakanishi K. Preparation of a tritiated ginkgolide. Bioorg Med Chem Lett 2004; 14:5673-5. [PMID: 15482945 DOI: 10.1016/j.bmcl.2004.08.042] [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] [Received: 06/18/2004] [Revised: 08/17/2004] [Accepted: 08/18/2004] [Indexed: 11/20/2022]
Abstract
Ginkgolide B, a constituent of the tree Ginkgo biloba, was radiolabeled with the beta-emitter tritium ([(3)H]) in two steps from ginkgolide C. First, a triflate precursor was prepared utilizing the selective reactivity of 7-OH in ginkgolide C; the triflate was then reduced with sodium borotritide to yield tritiated ginkgolide B ([(3)H]GB) in good yield and high specific activity. The tritiated ginkgolide will be an important tool for studying neuromodulatory properties of ginkgolides.
Collapse
|
28
|
Ranasinghe S, Bolsover S. Microglial Calcium Responses to Platelet-Activating Factor are Inhibited by Analogue CAS 99103-16-9 and Dihydropyridine PCA 4248 but Not by Ginkgolide A. ACTA ACUST UNITED AC 2004; 95:87-91. [PMID: 15379786 DOI: 10.1111/j.1742-7843.2004.950208.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Calcium signals evoked in N9 microglial cells were monitored using the calcium indicator dye Fluo-4 in a fluorescence imaging plate reader. Platelet activating factor in the range 100 nM to 20 microM elicited graded calcium responses. The analogue CAS 99103-16-9 inhibited the evoked calcium rise with an apparent KB of 1.3 +/- 0.4 microM. The dihydropyridine PCA 4248 inhibited the evoked calcium rise with an apparent KB of 1.2 +/- 0.2 microM. Ginkgolide A at concentrations up to 18 microM had no effect on the evoked calcium rise. While CAS 99103-16-9 and PCA 4248 appear to be simple competitive inhibitors of platelet-activating factor responses, the efficacy of ginkgolide in more complex pharmacological situations may result from an action at a site other than the platelet-activating factor receptor.
Collapse
Affiliation(s)
- Saman Ranasinghe
- Department of Physiology, University College London, Gower Street, London WC1E 6BT, U.K
| | | |
Collapse
|
29
|
Row BW, Kheirandish L, Li RC, Guo SZ, Brittian KR, Hardy M, Bazan NG, Gozal D. Platelet-activating factor receptor-deficient mice are protected from experimental sleep apnea-induced learning deficits. J Neurochem 2004; 89:189-96. [PMID: 15030403 DOI: 10.1111/j.1471-4159.2004.02352.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Intermittent hypoxia (IH) during sleep, a hallmark of sleep apnea, is associated with neurobehavioral impairments, regional neurodegeneration and increased oxidative stress and inflammation in rodents. Platelet-activating factor (PAF) is an important mediator of both normal neural plasticity and brain injury. We report that mice deficient in the cell surface receptor for PAF (PAFR-/-), a bioactive mediator of oxidative stress and inflammation, are protected from the spatial reference learning deficits associated with IH. Furthermore, PAFR-/- exhibit attenuated elevations in inflammatory signaling (cyclo-oxygenase-2 and inducible nitric oxide synthase activities), degradation of the ubiquitin-proteasome pathway and apoptosis observed in wild-type littermates (PAFR+/+) exposed to IH. Collectively, these findings indicate that inflammatory signaling and neurobehavioral impairments induced by IH are mediated through PAF receptors.
Collapse
Affiliation(s)
- Barry W Row
- Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Kim SR, Lee MK, Koo KA, Kim SH, Sung SH, Lee NG, Markelonis GJ, Oh TH, Yang JH, Kim YC. Dibenzocyclooctadiene lignans fromSchisandra chinensis protect primary cultures of rat cortical cells from glutamate-induced toxicity. J Neurosci Res 2004; 76:397-405. [PMID: 15079869 DOI: 10.1002/jnr.20089] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
A methanolic extract of dried Schisandra fruit (Schisandra chinensis Baill.; Schisandraceae) significantly attenuated the neurotoxicity induced by L-glutamate in primary cultures of rat cortical cells. Five dibenzocyclooctadiene lignans (deoxyschisandrin, gomisin N, gomisin A, schisandrin, and wuweizisu C) were isolated from the methanolic extract; their protective effects against glutamate-induced neurotoxicity were then evaluated. Among the five lignans, deoxyschisandrin, gomisin N, and wuweizisu C significantly attenuated glutamate-induced neurotoxicity as measured by 1). an inhibition in the increase of intracellular [Ca(2+)]; 2). an improvement in the glutathione defense system, the level of glutathione, and the activity of glutathione peroxidase; and 3). an inhibition in the formation of cellular peroxide. These results suggest that dibenzocyclooctadiene lignans from Schisandra chinensis may possess therapeutic potential against oxidative neuronal damage induced by excitotoxin.
Collapse
Affiliation(s)
- So Ra Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Nayernia K, Vauti F, Meinhardt A, Cadenas C, Schweyer S, Meyer BI, Schwandt I, Chowdhury K, Engel W, Arnold HH. Inactivation of a testis-specific Lis1 transcript in mice prevents spermatid differentiation and causes male infertility. J Biol Chem 2003; 278:48377-85. [PMID: 13129914 DOI: 10.1074/jbc.m309583200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lis1 protein is the non-catalytic component of platelet-activating factor acetylhydrolase 1b (PAF-AH 1B) and associated with microtubular structures. Hemizygous mutations of the LIS1 gene cause type I lissencephaly, a brain abnormality with developmental defects of neuronal migration. Lis1 is also expressed in testis, but its function there has not been determined. We have generated a mouse mutant (LIS1GT/GT) by gene trap integration leading to selective disruption of a Lis1 splicing variant in testis. Homozygous mutant males are infertile with no other apparent phenotype. We demonstrate that Lis1 is predominantly expressed in spermatids, and spermiogenesis is blocked when Lis1 is absent. Mutant spermatids fail to form correct acrosomes and nuclei appear distorted in size and shape. The tissue architecture in mutant testis appears severely disturbed displaying collapsed seminiferous tubules, mislocated germ cells, and increased apoptosis. These results provide evidence for an essential and hitherto uncharacterized role of the Lis1 protein in spermatogenesis, particularly in the differentiation of spermatids into spermatozoa.
Collapse
Affiliation(s)
- Karim Nayernia
- Institute for Human Genetics and Department of Pathology, University of Göttingen, Göttingen 37073, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Logan A, Berry M. Cellular and molecular determinants of glial scar formation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 513:115-58. [PMID: 12575819 DOI: 10.1007/978-1-4615-0123-7_4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ann Logan
- Molecular Neuroscience, Department of Medicine, Wolfson Research Laboratories, Queen Elizabeth Hospital, Edgbaston, Birmingham, B15 2TH, UK
| | | |
Collapse
|
33
|
Maclennan KM, Zheng Y, Sheard PW, Williams SM, Darlington CL, Smith PF. Adrenalectomy-induced cell death in the dentate gyrus: further characterisation using TUNEL and effects of the Ginkgo biloba extract, EGb 761, and ginkgolide B. Hippocampus 2003; 13:212-25. [PMID: 12699329 DOI: 10.1002/hipo.10073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This study investigated the potential neuroprotective effects of the Ginkgo biloba extract, EGb-761, and ginkgolide B, on adrenalectomy (ADX)-induced cell death in the dentate gyrus (DG). Adrenalectomised, sham surgery-treated, and naive controls received either EGb-761 (25, 50, or 100 mg/kg), 0.9% saline vehicle control, ginkgolide B (10 or 25 mg/kg), or a polyethylene glycol vehicle control, i.p, daily for 6 days postsurgery. Cell death in the DG was determined by in situ labelling of DNA fragments, using the TUNEL method; sections were counterstained with hematoxylin. Radioimmunoassay was used to confirm a decrease in plasma corticosterone (CORT) after ADX. TUNEL-positive granule cells were observed in the DG at 1 week, but not at 24 h, post-ADX. The rate of granule cell death at this time was highest in the suprapyramidal blade and increased in a crest tip and a rostrotemporal gradient. Whereas CORT replacement completely prevented the occurrence of TUNEL-positive granule cells, EGb-761 and ginkgolide B did not, at any of the doses used. These results suggest that these drugs may not have substantial neuroprotective effects in the ADX model of neurodegeneration.
Collapse
Affiliation(s)
- Karyn M Maclennan
- Department of Pharmacology and Toxicology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | | | | | | | | | | |
Collapse
|
34
|
Koizumi H, Yamaguchi N, Hattori M, Ishikawa TO, Aoki J, Taketo MM, Inoue K, Arai H. Targeted disruption of intracellular type I platelet activating factor-acetylhydrolase catalytic subunits causes severe impairment in spermatogenesis. J Biol Chem 2003; 278:12489-94. [PMID: 12551946 DOI: 10.1074/jbc.m211836200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Intracellular type I platelet activating factor-acetylhydrolase is a phospholipase that consists of a dimer of two homologous catalytic subunits alpha1 and alpha2 as well as LIS1, a product of the causative gene for type I lissencephaly. LIS1 plays an important role in neuronal migration during brain development, but the in vivo function of the catalytic subunits remains unclear. In this study, we generated alpha1- and a2-deficient mice by targeted disruption. alpha1(-/-) mice are indistinguishable from wild-type mice, whereas alpha2(-/-) male mice show a significant reduction in testis size. Double-mutant male mice are sterile because of severe impairment of spermatogenesis. Histological examination revealed marked degeneration at the spermatocyte stage and an increase of apoptotic cells in the seminiferous tubules. The catalytic subunits are expressed at high levels in testis as well as brain in mice. In wild-type mice, alpha2 is expressed in all seminiferous tubule cell types, whereas alpha1 is expressed only in the spermatogonia. This expression pattern parallels the finding that deletion of both subunits induces a marked loss of germ cells at an early spermatogenic stage. We also found that the LIS1 protein levels, but not the mRNA levels, were significantly reduced in alpha2(-/-) and double-mutant mice, suggesting that the catalytic subunits, especially alpha2, are a determinant of LIS1 expression level.
Collapse
Affiliation(s)
- Hiroyuki Koizumi
- Department of Health Chemistry and Laboratory of Biomedical Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Polazzi E, Contestabile A. Reciprocal interactions between microglia and neurons: from survival to neuropathology. Rev Neurosci 2003; 13:221-42. [PMID: 12405226 DOI: 10.1515/revneuro.2002.13.3.221] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Microglia represent a major cellular component of the brain, where they constitute a widely distributed network of immunoprotective cells. During the last decades, it has become clear that the functions traditionally ascribed to microglia, i.e. to dispose of dead cells and debris and to mediate brain inflammatory states, are only a fraction of a much wider repertoire of functions spanning from brain development to aging and neuropathology. The aim of the present survey is to critically discuss some of these functions, focusing in particular on the reciprocal microglia-neuron interactions and on the complex signaling systems subserving them. We consider first some of the functional interactions dealing with invasion, proliferation and migration of microglia as well as with the establishment of the initial blueprint of neural circuits in the developing brain. The signals related to the suppression of immunological properties of microglia by neurons in the healthy brain, and the derangement from this physiological equilibrium in aging and diseases, are then examined. Finally, we make a closer examination of the reciprocal signaling between damaged neurons and microglia and, on these bases, we propose that microglial activation, consequent to neuronal injury, is primarily aimed at neuroprotection. The loss of specific communication between damaged neurons and microglia is viewed as responsible for the turning of microglia to a hyperactivated state, which allows them to escape neuronal control and to give rise to persistent inflammation, resulting in exacerbation of neuropathology. The data surveyed here point at microglial-neuron interactions as the basis of a complex network of signals conveying messages with high information content and regulating the most important aspects of brain function. This network shares similar features with some fundamental principles governing the activity of brain circuits: it is provided with memory and it continuously evolves in relation to the flow of time and information.
Collapse
|
36
|
McManus LM, Pinckard RN. PAF, a putative mediator of oral inflammation. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 2002; 11:240-58. [PMID: 12002818 DOI: 10.1177/10454411000110020701] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PAF, or platelet-activating factor, is a family of structurally related phospholipids (1-O-alkyl/acyl/alkenyl-2-acetyl-sn-glycero-3-phosphocholine) which possesses a wide spectrum of potent pro-inflammatory actions. These phospholipids are synthesized by a diverse array of cells, including neutrophilic polymorphonuclear leukocytes (PMN), platelets, mast cells, monocytes/macrophages, vascular endothelial cells, and lymphocytes. PAF targets these and other cells via specific, G-protein-coupled receptors to initiate intracrine, autocrine, paracrine, and juxtacrine cell activation. Of importance, these unique acetylated phospholipids are frequently synthesized in concert with pro-inflammatory lipid mediators derived from arachidonic acid. Since PAF synergizes with these and other mediators to amplify the inflammatory response, it seems likely that PAF plays an integral, perhaps pivotal, role in acute and chronic inflammatory processes. PAF is present in the mixed saliva of dentate, but not edentulous, human subjects. The levels of PAF in mixed saliva or in gingival crevicular fluid and tissues are significantly increased during oral inflammatory conditions such as periodontitis and mucositis. Interestingly, the levels of salivary PAF correlate with the extent/severity of these oral diseases. These observations suggest that PAF may participate in pathophysiologic events during the course of oral inflammation. The availability of specific PAF receptor antagonists and human recombinant PAF-acetylhydrolase (PAF-AH), a plasma enzyme which rapidly destroys PAF, should provide clinical tools for the investigation of the role of PAF in these and other inflammatory disorders; and perhaps, ultimately, some of these reagents may prove to be therapeutically useful in the treatment and management of these conditions.
Collapse
Affiliation(s)
- L M McManus
- Department of Pathology, The University of Texas Health Science Center, San Antonio 78229, USA.
| | | |
Collapse
|
37
|
Hostettler ME, Knapp PE, Carlson SL. Platelet-activating factor induces cell death in cultured astrocytes and oligodendrocytes: involvement of caspase-3. Glia 2002; 38:228-39. [PMID: 11968060 DOI: 10.1002/glia.10065] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The biologically active lipid metabolite, platelet-activating factor (PAF), is thought to contribute to inflammatory processes and tissue damage in a variety of central nervous system (CNS) injuries. In previous studies, we found that after contusion spinal cord injury, treatment with a PAF antagonist led to significantly increased white matter tissue sparing as well as decreased mRNA levels for pro-inflammatory cytokines. Some studies suggest that PAF can also have toxic effects on neurons in vitro. Few studies, however, have examined the effects of PAF on glial cells of the CNS. In the present study, the potential for PAF to act as a toxin to cultured astrocytes was examined. Also investigated were the effects of PAF on oligodendrocytes at two different stages of development. Treatment with 0.02-2 microM PAF for 72 h resulted in significant levels of cell death in both cell types (P < 0.05), an effect that was blocked by the PAF receptor antagonists, WEB 2170 and BN 52021. To investigate PAF-induced glial cell death further, we looked for activation of the enzyme, caspase-3, which can be indicative of apoptosis. Immunocytochemistry demonstrated that PAF at all concentrations caused activation of caspase-3 at 24, 48, and 72 h after treatment in both cell types. Caspase-3-dependent cell death was further confirmed using knockout mice (-/-) deficient in the caspase-3 gene. Toxicity was lost when astrocytes (-/-) were exposed to 0.02-2 microM PAF (P < 0.01). Oligodendrocytes (-/-) were not susceptible to toxicity at 2 microM PAF (P < 0.001). The results demonstrate that the pro-inflammatory molecule, PAF, induces cell death in cultured CNS glial cells and that this effect is, in part, dependent on caspase-3 activation.
Collapse
Affiliation(s)
- Mary Ellen Hostettler
- Department of Anatomy and Neurobiology, University of Kentucky Medical Center, Lexington 40536-0398, USA
| | | | | |
Collapse
|
38
|
Grassi S, Pettorossi VE. Synaptic plasticity in the medial vestibular nuclei: role of glutamate receptors and retrograde messengers in rat brainstem slices. Prog Neurobiol 2001; 64:527-53. [PMID: 11311461 DOI: 10.1016/s0301-0082(00)00070-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The analysis of cellular-molecular events mediating synaptic plasticity within vestibular nuclei is an attempt to explain the mechanisms underlying vestibular plasticity phenomena. The present review is meant to illustrate the main results, obtained in vitro, on the mechanisms underlying long-term changes in synaptic strength within the medial vestibular nuclei. The synaptic plasticity phenomena taking place at the level of vestibular nuclei could be useful for adapting and consolidating the efficacy of vestibular neuron responsiveness to environmental requirements, as during visuo-vestibular recalibration and vestibular compensation. Following a general introduction on the most salient features of vestibular compensation and visuo-vestibular adaptation, which are two plastic events involving neuronal circuitry within the medial vestibular nuclei, the second and third sections describe the results from rat brainstem slice studies, demonstrating the possibility to induce long-term potentiation and depression in the medial vestibular nuclei, following high frequency stimulation of the primary vestibular afferents. In particular the mechanisms sustaining the induction and expression of vestibular long-term potentiation and depression, such as the role of various glutamate receptors and retrograde messengers have been described. The relevant role of the interaction between the platelet-activating factor, acting as a retrograde messenger, and the presynaptic metabotropic glutamate receptors, in determining the full expression of vestibular long-term potentiation is also underlined. In addition, the mechanisms involved in vestibular long-term potentiation have been compared with those leading to long-term potentiation in the hippocampus to emphasize the most significant differences emerging from vestibular studies. The fourth part, describes recent results demonstrating the essential role of nitric oxide, another retrograde messenger, in the induction of vestibular potentiation. Finally the fifth part suggests the possible functional significance of different action times of the two retrograde messengers and metabotropic glutamate receptors, which are involved in mediating the presynaptic mechanism sustaining vestibular long-term potentiation.
Collapse
Affiliation(s)
- S Grassi
- Dipartimento di Medicina Interna, Sezione di Fisiologia Umana, Università di Perugia, I-06100, Perugia, Italy.
| | | |
Collapse
|
39
|
Farooqui AA, Horrocks LA, Farooqui T. Glycerophospholipids in brain: their metabolism, incorporation into membranes, functions, and involvement in neurological disorders. Chem Phys Lipids 2000; 106:1-29. [PMID: 10878232 DOI: 10.1016/s0009-3084(00)00128-6] [Citation(s) in RCA: 328] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neural membranes contain several classes of glycerophospholipids which turnover at different rates with respect to their structure and localization in different cells and membranes. The glycerophospholipid composition of neural membranes greatly alters their functional efficacy. The length of glycerophospholipid acyl chain and the degree of saturation are important determinants of many membrane characteristics including the formation of lateral domains that are rich in polyunsaturated fatty acids. Receptor-mediated degradation of glycerophospholipids by phospholipases A(l), A(2), C, and D results in generation of second messengers such as arachidonic acid, eicosanoids, platelet activating factor and diacylglycerol. Thus, neural membrane phospholipids are a reservoir for second messengers. They are also involved in apoptosis, modulation of activities of transporters, and membrane-bound enzymes. Marked alterations in neural membrane glycerophospholipid composition have been reported to occur in neurological disorders. These alterations result in changes in membrane fluidity and permeability. These processes along with the accumulation of lipid peroxides and compromised energy metabolism may be responsible for the neurodegeneration observed in neurological disorders.
Collapse
Affiliation(s)
- A A Farooqui
- Department of Molecular and Cellular Biochemistry, Ohio State University, 1645 Neil Avenue, 465 Hamilton Hall, 43210, Columbus, OH, USA
| | | | | |
Collapse
|
40
|
Abstract
In this review properties of lipid acetyltransferase enzymes are outlined. The three activities of interest are lyso PAF acetyltransferase (acetyl CoA: 1-alkyl-sn-glycero-3-phosphocholine acetyltransferase), AGP acetyltransferase (acetyl CoA: 1-alkyl sn-glycero-3-phosphate acetyltransferase) and a transacetylase activity that can transfer acetyl groups from PAF to lipid acceptors in the formation of 1-alkenyl-2-acetyl-sn-glycero-3-phosphoethanolamine and N-acetyl sphingosine (C2 ceramide). This review focuses on the role of acetyltransferases and transacetylases within the metabolism of platelet-activating factor and specifically addresses characteristics of the enzymes, including subcellular localization, substrate selectivity, and enzymatic regulation.
Collapse
Affiliation(s)
- R R Baker
- Department of Biochemistry, University of Toronto, Canada
| |
Collapse
|
41
|
Baker RR, Chang HY. A metabolic path for the degradation of lysophosphatidic acid, an inhibitor of lysophosphatidylcholine lysophospholipase, in neuronal nuclei of cerebral cortex. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1483:58-68. [PMID: 10601695 DOI: 10.1016/s1388-1981(99)00185-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neuronal nuclei isolated from rabbit cerebral cortex were found to be enriched in an NEM-insensitive lysophosphatidic acid (lysoPA) phosphohydrolase activity. LysoPA is an inhibitor of the nuclear lysophosphatidylcholine (lysoPC) lysophospholipase, and by preserving lysoPC levels, lysoPA boosted the nuclear production of the acyl analogue of platelet-activating factor by promoting the acetylation of lysoPC (Baker and Chang, Mol. Cell Biochem., 1999, in press). The nuclear phosphohydrolase converts lysoPA to 1-monoacylglycerol, and thus eliminates this lysoPA inhibition of lysoPC lysophospholipase. The nuclear lysoPA phosphohydrolase specific activity was more than three times that observed for the nuclear lysoPA lysophospholipase (Baker and Chang, Biochim. Biophys. Acta 1438 (1999) 253-263) and represents a more active route for nuclear lysoPA removal. The neuronal nuclear lysoPA phosphohydrolase was inhibited at acidic pH, and also inhibited by calcium ions. The 1-monoacylglycerol product of the phosphohydrolase is rapidly degraded by neuronal monoacylglycerol lipase, an enzyme some sevenfold more active than the phosphohydrolase and sensitive to inhibition by arachidonoyl trifluoromethyl ketone (AACOCF(3)). Both acidic pH and free fatty acid inhibited the lipase. In the absence of AACOCF(3), production of fatty acid from lysoPA substrate could be largely attributed to the sequential actions of the nuclear phosphohydrolase and lipase. This facilitates fatty acid recycling back into phospholipid by lysophospholipid acylation when ATP levels are restored following periods of brain ischemia. At relatively low concentrations, sphingosine-1-phosphate, and alkylglycerophosphate were the most effective phosphohydrolase inhibitors while phosphatidic acid, alkylacetylglycerophosphate and ceramide were without effect. LysoPA is an interesting regulatory molecule that can potentially preserve lysophosphatidylcholine within the nuclear membrane for use in acetylation reactions. Thus conditions relevant to brain ischemia such as falling pH, falling ATP concentrations, rising fatty acid and intracellular calcium levels may, by slowing this metabolic path for lysoPA loss, promote the production of acyl PAF and contribute to the increased levels of the acetylated lipids noted in ischemia.
Collapse
Affiliation(s)
- R R Baker
- Department of Biochemistry, Room 5202, Medical Sciences Bldg., University of Toronto, Toronto, Ont., Canada.
| | | |
Collapse
|
42
|
Grassi S, Francescangeli E, Goracci G, Pettorossi VE. Platelet-activating factor and group I metabotropic glutamate receptors interact for full development and maintenance of long-term potentiation in the rat medial vestibular nuclei. Neuroscience 1999; 94:549-59. [PMID: 10579215 DOI: 10.1016/s0306-4522(99)00284-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In rat brainstem slices, we investigated the interaction between platelet-activating factor and group I metabotropic glutamate receptors in mediating long-term potentiation within the medial vestibular nuclei. We analysed the N1 field potential wave evoked in the ventral portion of the medial vestibular nuclei by primary vestibular afferent stimulation. The group I metabotropic glutamate receptor antagonist, (R,S)-1-aminoindan-1,5-dicarboxylic acid, prevented long-term potentiation induced by a platelet-activating factor analogue [1-O-hexadecyl-2-O-(methylcarbamyl)-sn-glycero-3-phosphocholine], as well as the full development of potentiation, induced by high-frequency stimulation under the blocking agent for synaptosomal platelet-activating factor receptors (ginkolide B), at drug washout. However, potentiation directly induced by the group I glutamate metabotropic receptor agonist, (R,S)-3,5-dihydroxyphenylglycine, was reduced by ginkolide B. These findings suggest that platelet-activating factor, whether exogenous or released following potentiation induction, exerts its effect through presynaptic group I metabotropic glutamate receptors, mediating the increase of glutamate release. In addition, we found that this mechanism, which led to full potentiation through presynaptic group I metabotropic glutamate receptor activation, was inactivated soon after application of potentiation-inducing stimulus. In fact, the long-lasting block of the platelet-activating factor and metabotropic glutamate receptors prevented the full potentiation development and the induced potentiation progressively declined to null. Moreover, ginkolide B, given when high-frequency-dependent potentiation was established, only reduced it within 5 min after potentiation induction. We conclude that to fully develop vestibular long-term potentiation requires presynaptic events. Platelet-activating factor, released after the activation of postsynaptic mechanisms which induce potentiation, is necessary for coupling postsynaptic and presynaptic phenomena, through the activation of group I metabotropic glutamate receptors, and its action lasts only for a short period. If this coupling does not occur, a full and long-lasting potentiation cannot develop.
Collapse
Affiliation(s)
- S Grassi
- Institute of Human Physiology, University of Perugia, Italy.
| | | | | | | |
Collapse
|
43
|
Baker RR, Chang HY. Lysophosphatidic acid, alkylglycerophosphate and alkylacetylglycerophosphate increase the neuronal nuclear acetylation of 1-acyl lysophosphatidyl choline by inhibition of lysophospholipase. Mol Cell Biochem 1999; 198:47-55. [PMID: 10497877 DOI: 10.1023/a:1006933625802] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Neuronal nuclei were isolated from rabbit cerebral cortex, and lipid acetylation reactions were studied because of the high nuclear concentration of acetyltransferases that generate platelet activating factor (PAF) and its acyl analogue AcylPAF. The neuronal nuclear acetylation of 1-palmitoyl lysophosphatidylcholine (lyso PC) was found to be increased more than twofold when low concentrations of lyso PC were incubated in acetylation assays in the presence of 1-palmitoyl lysophosphatidic acid (lyso PA) or 1-hexadecyl glycerophosphate (AGP). This effect was not found for a variety of other acidic and neutral 1-acyl lysoglycerophospholipids. At 4 microM concentrations, AGP was the more effective in increasing rates of lyso PC acetylation, while lyso PA was more effective at 25-35 microM. 1-Stearoyl, 1-alkenyl and 1-decanoyl analogues of lyso PA were all less effective than 1-palmitoyl lyso PA. Phosphatidic acid was considerably less effective than lyso PA, while the acetylated analogue of AGP, AAcGP (alkylacetylglycerophosphate), increased rates of lyso PC acetylation to maxima similar to those seen with lyso PA or AGP. In addition, AAcGP promoted these maxima at considerably lower concentrations (2-4 microM). A mechanism for these effects was suggested when nuclear envelopes (NE), isolated in the presence of PMSF, showed these maximal acetylation rates at low lyso PC concentrations, and these rates were not elevated by the presence of lyso PA. PMSF is a protease inhibitor but can also inhibit lysophospholipase activity. We found a nuclear lysophospholipase that degraded lyso PC at rates more than 13 times those of nuclear lyso PC acetylation. PMSF did inhibit this nuclear lysophospholipase, as did lyso PA, AGP and AAcGP. Kinetic analyses of the effects of lyso PA, AGP and AAcGP on lyso PC lysophospholipase indicated that these three lipids acted as competitive inhibitors for the lyso PC substrate. It is possible that low rates of lyso PC acetylation seen in neuronal nuclei at low lyso PC concentrations, are caused by lyso PC loss mediated by a very strong nuclear lysophospholipase. The effects of lyso PA, AGP and AAcGP in boosting rates of lyso PC acetylation likely come from the inhibition of nuclear lysophospholipase and a preservation of lyso PC concentrations. Competing neuronal nuclear reactions for low endogenous levels of lyso PC may regulate the formation of AcylPAF, and rising lyso PA, AGP or AAcGP concentrations can increase rates of nuclear AcylPAF synthesis.
Collapse
Affiliation(s)
- R R Baker
- Department of Biochemistry, University of Toronto, ON, Canada
| | | |
Collapse
|
44
|
Baker RR, Chang HY. Evidence for two distinct lysophospholipase activities that degrade lysophosphatidylcholine and lysophosphatidic acid in neuronal nuclei of cerebral cortex. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1438:253-63. [PMID: 10320808 DOI: 10.1016/s1388-1981(99)00057-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuronal nuclei were isolated from immature rabbit cerebral cortex and nuclear lysophospholipase activities studied using two different 1-acyl lysophospholipids: lysophosphatidylcholine (lysoPC) and lysophosphatidic acid (lysoPA). Our interest in these two lysolipids arose from the observation that lysoPA could promote the acetylation of lysoPC by substantially inhibiting a very active nuclear lysoPC lysophospholipase activity, in a competitive manner (R.R. Baker, H. -y. Chang, Mol. Cell. Biochem. (1999) in press). As there was also evidence for nuclear lysoPA deacylation, it was of interest to see whether one activity could possibly utilize both lysolipid substrates. We now have evidence for two separate lysophospholipase activities in neuronal nuclei. The lysoPC lysophospholipase activity was the more active, more highly enriched in the neuronal nuclei, and showed optimal activity at pH 8.4-9, while the lysoPA lysophospholipase activity was maintained over a much broader pH range. The lysoPC activity was substantially inhibited by free fatty acid, and showed considerable stimulation by serum albumin, while the activity utilizing lysoPA was much less affected by these agents. When lysoPC was added to incubations containing radioactive lysoPA, there was no significant inhibition found in rates of release of radioactive fatty acid, indicating that the lysoPA lysophospholipase activity did not utilize the lysoPC substrate. In incubations with lysoPC, MgATP and CoA brought about a sizable formation of phosphatidylcholine whose radioactivity was equally distributed between the sn-1 and sn-2 positions suggesting labelling both directly from the lysoPC substrate and from fatty acid produced by the lysophospholipase activity. By comparison, with the radioactive lysoPA substrate, MgATP and CoA promoted relatively lower levels of phosphatidic acid formation whose principal labelling came directly from the radioactive lysoPA. Largely because of the high activity of the nuclear lysoPC lysophospholipase, there is considerable potential in the neuronal nucleus to limit the use of lysoPC in other reactions, such as the formation of acylPAF (1-acyl analogue of platelet activating factor). It is of interest that conditions associated with brain ischaemia such as increased free fatty acid levels, falling pH and declines in MgATP may allow a preservation of neuronal nuclear lysoPC levels for acetylation. The existence of a separate lysophospholipase activity for lysoPA allows an independent control of lysoPA which can serve as an important regulator of the nuclear lysoPC lysophospholipase.
Collapse
Affiliation(s)
- R R Baker
- Department of Biochemistry, Room 5202, Medical Sciences Bldg., University of Toronto, Toronto, Ont. M5S 1A8, Canada
| | | |
Collapse
|
45
|
Le Gouill C, Parent JL, Caron CA, Gaudreau R, Volkov L, Rola-Pleszczynski M, Stanková J. Selective modulation of wild type receptor functions by mutants of G-protein-coupled receptors. J Biol Chem 1999; 274:12548-54. [PMID: 10212233 DOI: 10.1074/jbc.274.18.12548] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Members of the G-protein-coupled receptor (GPCR) family are involved in most aspects of higher eukaryote biology, and mutations in their coding sequence have been linked to several diseases. In the present study, we report that mutant GPCR can affect the functional properties of the co-expressed wild type (WT) receptor. Mutants of the human platelet-activating factor receptor that fail to show any detectable ligand binding (N285I and K298stop) or coupling to a G-protein (D63N, D289A, and Y293A) were co-expressed with the WT receptor in Chinese hamster ovary and COS-7 cells. In this context, N285I and K298stop mutant receptors inhibited 3H-WEB2086 binding and surface expression. Co-transfection with D63N resulted in a constitutively active receptor phenotype. Platelet-activating factor-induced inositol phosphate production in cells transfected with a 1:1 ratio of WT:D63N was higher than with the WT cDNA alone but was abolished with a 1:3 ratio. We confirmed that these findings could be extended to other GPCRs by showing that co-expression of the WT C-C chemokine receptor 2b with a carboxyl-terminal deletion mutant (K311stop), resulted in a decreased affinity and responsiveness to MCP-1. A better understanding of this phenomenon could lead to important tools for the prevention or treatment of certain diseases.
Collapse
Affiliation(s)
- C Le Gouill
- Immunology Division, Department of Pediatrics, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada. sternsdo@
| | | | | | | | | | | | | |
Collapse
|
46
|
Kobayashi K, Ishii S, Kume K, Takahashi T, Shimizu T, Manabe T. Platelet-activating factor receptor is not required for long-term potentiation in the hippocampal CA1 region. Eur J Neurosci 1999; 11:1313-6. [PMID: 10103126 DOI: 10.1046/j.1460-9568.1999.00538.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
From pharmacological studies, platelet-activating factor (PAF) has been proposed as a retrograde messenger for long-term potentiation (LTP) in the hippocampal CA1 region. We re-examined a possible contribution of PAF to LTP with a more specific approach using mice deficient in the PAF receptor. The PAF receptor-deficient mice exhibited normal LTP and showed no obvious abnormality in excitatory synaptic transmission. We also performed pharmacological experiments on the wild-type mice. Two structurally different antagonists of PAF receptors had no effects on LTP. Furthermore, the application of PAF itself caused no detectable changes in excitatory synaptic transmission. Thus, we conclude that the PAF receptor is not required for LTP in the CA1 region. Introduction
Collapse
Affiliation(s)
- K Kobayashi
- Department of Neurophysiology, Faculty of Medicine, University of Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Roy Baker R, Chang H. Substrate specificities of neuronal nuclear acetyltransferases involved in the synthesis of platelet-activating factor: differences in the use of 1-alkyl and 1-acyl lysophospholipid acceptors. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1390:215-24. [PMID: 9507140 DOI: 10.1016/s0005-2760(97)00168-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The selectivity of alkylglycerophosphate (AGP) acetyltransferase and lyso-platelet-activating factor (lyso-PAF) acetyltransferase was studied in neuronal nuclei isolated from cerebral cortices of 15-day-old rabbits. Specifically, 1-alkyl and 1-acyl analogues were compared as acceptors in these acetylation reactions. A number of observations supported one nuclear activity in the acetylation of AGP and lyso-PA. Lyso-PA was a competitive substrate for AGP, Km values for AGP and lyso-PA were similar, as were acetylation rates measured at individual AGP or lyso-PA concentrations, and the acetylation of both substrates was unaffected by preincubations with protein phosphatase 1 (PP-1). In contrast, there were a number of differences seen in the acetylation of lyso-PAF and lyso-PC. The kinetics for lyso-PC acetylation (as a function of lyso-PC concentration) were not hyperbolic, and lyso-PC was not a competitive substrate for the acetylation of lyso-PAF. Unlike acetylation rates with lyso-PAF, lyso-PC acetylation was not reduced by preincubations with PP-1, and was less susceptible to inhibition particularly at high levels of free fatty acid. In addition, rates of acetylation of lyso-PC were selectively increased by the presence of lyso-PA. When neuronal nuclear envelope fractions (NE) were prepared from N1, the specific acetylation activity with lyso-PAF was significantly lower in NE, while the activities for lyso-PC were comparable in NE and the parent N1 fraction. The results with the acetylation of lyso-PC and lyso-PAF suggest that the lyso-PC acetyltransferase may be in a uniquely sequestered state within the neuronal nucleus. This could explain the smaller inhibition of lyso-PC acetylation by free fatty acid, the maintenance of lyso-PC acetylation during PP-1 preincubations, the non-hyperbolic response to lyso-PC concentrations and the selective preservation of lyso-PC acetylation during NE isolation. This protected status could result from a more internal location for this acetyltransferase within the membranes of the nuclear envelope, or possibly an association of the enzyme with the nuclear matrix that is disrupted with the exposure of N1 to lyso-PA.
Collapse
Affiliation(s)
- R Roy Baker
- Division of Neurology, Department of Medicine, Clinical Science Division, Room 6368, Medical Sciences Bldg., University of Toronto, Toronto, Ont., Canada
| | | |
Collapse
|
48
|
Cowen MS, Beart PM. Cyclothiazide and AMPA receptor desensitization: analyses from studies of AMPA-induced release of [3H]-noradrenaline from hippocampal slices. Br J Pharmacol 1998; 123:473-80. [PMID: 9504388 PMCID: PMC1565195 DOI: 10.1038/sj.bjp.0701638] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
1. Responses in brain produced by the activation of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) subtype of ionotropic receptor for L-glutamate are often rapidly desensitizing. AMPA-induced desensitization and its characteristics, and the potentiating effect of cyclothiazide were investigated in vitro by analysing AMPA-induced release of [3H]-noradrenaline from prisms of rat hippocampus. 2. AMPA (1-1000 microM) stimulated the release of [3H]-noradrenaline in a concentration-dependent manner that was both calcium-dependent and tetrodotoxin-sensitive, and attenuated by the AMPA-selective antagonists, NBQX (1 and 10 microM), LY 293558 (1 and 10 microM) and GYKI 52466 (10 and 30 microM). 3. By use of an experimental procedure with consecutive applications of AMPA (100 microM, 28 min apart), the second response was reduced, indicative of receptor desensitization, and was reversed by cyclothiazide in a concentration-dependent manner (1-300 microM). The concentration-response curve for AMPA-induced release of [3H]-noradrenaline was shifted leftwards, but the reversal by cyclothiazide of the desensitized response was partial and failed to reach the maximal response of the first stimulus. 4. Observations made with various schedules of cyclothiazide application indicated that the initial AMPA-evoked response was already partially desensitized (150% potentiation by 100 microM cyclothiazide) and that the desensitization was not likely to be due to a time-dependent diminution and was longlasting (second application of cyclothiazide was ineffective). 5. Co-application of a number of drugs with actions on second messenger systems, in association with the second AMPA stimulus, revealed significant potentiation of the AMPA-induced release of [3H]-noradrenaline: forskolin (10 microM, +78%), Rp-cAMPS (100 microM, +65%), Ro 31-8220 (10 microM, +163%) and thapsigargin (100 pM, + 161%). 6. The AMPA receptor-mediated response regulating the release of [3H]-noradrenaline from rat hippocampal slices was desensitized and cyclothiazide acted to reverse partially the desensitization in a concentration-dependent manner. Since the time-course of desensitization was longer lasting than that noted in previous electrophysiological studies, multiple events may be involved in the down-regulation of AMPA receptor activity including receptor phosphorylation and depletion of intracellular Ca2+ stores.
Collapse
Affiliation(s)
- M S Cowen
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
49
|
Köller H, Siebler M, Hartung HP. Immunologically induced electrophysiological dysfunction: implications for inflammatory diseases of the CNS and PNS. Prog Neurobiol 1997; 52:1-26. [PMID: 9185232 DOI: 10.1016/s0301-0082(96)00065-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
During inflammation of the central or peripheral nervous system, a high number of immunologically active molecules, including bacterial or viral products as well as host-derived cytokines, are released. Patients suffering from inflammatory CNS or PNS diseases often develop transient symptoms with a rapid recovery, which obviously cannot be accounted for by immunologically induced tissue damage. These observations led to the hypothesis that immunologically active molecules can affect directly the electrophysiological functions of neurons and glial cells. Evidence for this hypothesis came from in vitro studies showing that cytokines, such as interleukins or tumor necrosis factors, arachidonic acid and its metabolites, interfere with electrophysiological properties of neurons or glial cells. These molecules affect ion currents, intracellular Ca2+ homeostasis, membrane potentials, and suppress or enhance the induction and maintenance of long-term potentiation. Similarly, virus proteins from human immunodeficiency virus type I were found to alter intracellular Ca2+ concentrations of neurons and astrocytes by modulating either transmitter receptors and channels or membrane transporters. Cerebrospinal fluid from MS patients contains factors which increase Na+ current inactivation and thereby reduce neuronal excitability. Immunoglobulins in sera of patients suffering from multifocal motor neuropathy and from acquired neuromyotonia interfere with nerve fibers, inducing alterations of conduction. Increased knowledge of these mechanisms will help to explain the pathogenesis of neurological symptoms and may provide a rationale for new therapeutic strategies.
Collapse
Affiliation(s)
- H Köller
- Department of Neurology, Heinrich-Heine University Düsseldorf, Germany
| | | | | |
Collapse
|
50
|
Maclennan K, Smith PF, Darlington CL. The effects of ginkgolide B (BN52021) on guinea pig vestibular nucleus neurons in vitro: importance of controlling for effects of dimethylsulphoxide (DMSO) vehicles. Neurosci Res 1996; 26:395-9. [PMID: 9004279 DOI: 10.1016/s0168-0102(96)01118-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The present study investigated the effects of the platelet-activating factor (PAF) receptor antagonist, ginkgolide B, on guinea pig medial vestibular nucleus (MVN) neurons in vitro and addressed the potential problem of using dimethylsulphoxide (DMSO) as a solvent. Using DMSO as a solvent, 10(-10), 10(-8) and 10(-4) M ginkgolide B had no effect on extracellularly recorded MVN neurons in brainstem slices in vitro. Using 10(-6) M ginkgolide B, 50% (5/10) of MVN neurons exhibited a decrease in firing rate and 10% (1/10) of neurons responded with an increase in firing rate. While attempting to control for possible DMSO vehicle effects, 40% (4/10) of MVN neurons displayed a decrease in firing rate when ginkgolide B (10(-6) M) was presented before the DMSO control solution. When the DMSO vehicle was presented first, only 10% (1/10) responded similarly. In some cases the DMSO vehicle alone also produced a decrease in MVN neuron firing rate. These results emphasise the importance of using adequate DMSO control conditions in ginkgolide research.
Collapse
Affiliation(s)
- K Maclennan
- Department of Pharmacology, School of Medical Sciences, University of Otago Medical School, Dunedin, New Zealand
| | | | | |
Collapse
|