Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Neurochem Res ; 37(4): 671-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22101393

ABSTRACT

Tricyclodecan-9-yl-xanthogenate (D609) is known for its antiviral and antitumor properties. D609 actions are widely attributed to inhibiting phosphatidylcholine (PC)-specific phospholipase C (PC-PLC). D609 also inhibits sphingomyelin synthase (SMS). PC-PLC and/or SMS inhibition will affect lipid second messengers 1,2-diacylglycerol (DAG) and/or ceramide. Evidence indicates either PC-PLC and/or SMS inhibition affected the cell cycle and arrested proliferation, and stimulated differentiation in various in vitro and in vivo studies. Xanthogenate compounds are also potent antioxidants and D609 reduced Aß-induced toxicity, attributed to its antioxidant properties. Zn²âº is necessary for PC-PLC enzymatic activity; inhibition by D609 might be attributed to its Zn²âº chelation. D609 has also been proposed to inhibit acidic sphingomyelinase or down-regulate hypoxia inducible factor-1α; however these are down-stream events related to PC-PLC inhibition. Characterization of the mammalian PC-PLC is limited to inhibition of enzymatic activity (frequently measured using Amplex red assay with bacterial PC-PLC as a standard). The mammalian PC-PLC has not been cloned; sequenced and structural information is unavailable. D609 showed promise in cancer studies, reduced atherosclerotic plaques (inhibition of PC-PLC) and cerebral infarction after stroke (PC-PLC or SMS). D609 actions as an antagonist to pro-inflammatory cytokines have been attributed to PC-PLC. The purpose of this review is to comprehensively evaluate the literature and summarize the findings and relevance to cell cycle and CNS pathologies.


Subject(s)
Bridged-Ring Compounds/pharmacology , Cell Cycle/drug effects , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Thiones/pharmacology , Type C Phospholipases/antagonists & inhibitors , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Antioxidants/therapeutic use , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/therapeutic use , Cell Cycle/physiology , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/enzymology , Humans , Norbornanes , Sphingomyelin Phosphodiesterase/metabolism , Thiocarbamates , Thiones/chemistry , Thiones/therapeutic use , Type C Phospholipases/metabolism
2.
BMB Rep ; 41(8): 560-7, 2008 Aug 31.
Article in English | MEDLINE | ID: mdl-18755070

ABSTRACT

The importance of lipids in cell signaling and tissue physiology is demonstrated by the many CNS pathologies involving deregulated lipid metabolism. One such critical metabolic event is the activation of phospholipase A(2) (PLA(2)), which results in the hydrolysis of membrane phospholipids and the release of free fatty acids, including arachidonic acid, a precursor for essential cell-signaling eicosanoids. Reactive oxygen species (ROS, a product of arachidonic acid metabolism) react with cellular lipids to generate lipid peroxides, which are degraded to reactive aldehydes (oxidized phospholipid, 4-hydroxynonenal, and acrolein) that bind covalently to proteins, thereby altering their function and inducing cellular damage. Dissecting the contribution of PLA(2) to lipid peroxidation in CNS injury and disorders is a challenging proposition due to the multiple forms of PLA(2), the diverse sources of ROS, and the lack of specific PLA(2) inhibitors. In this review, we summarize the role of PLA(2) in CNS pathologies, including stroke, spinal cord injury, Alzheimer's, Parkinson's, Multiple sclerosis-Experimental autoimmune encephalomyelitis and Wallerian degeneration.


Subject(s)
Central Nervous System Diseases/metabolism , Lipid Peroxidation , Phospholipases A2/metabolism , Reactive Oxygen Species/metabolism , Animals , Brain Ischemia/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Hypoxia, Brain/metabolism , Neurodegenerative Diseases/metabolism , Reactive Nitrogen Species/metabolism , Spinal Cord Injuries/metabolism , Stroke/metabolism
3.
Subcell Biochem ; 49: 241-68, 2008.
Article in English | MEDLINE | ID: mdl-18751914

ABSTRACT

Deregulated lipid metabolism may be of particular importance for CNS injuries and disorders, as this organ has the highest lipid concentration next to adipose tissue. Atherosclerosis (a risk factor for ischemic stroke) results from accumulation of LDL-derived lipids in the arterial wall. Pro-inflammatory cytokines (TNF-alpha and IL-1), secretory phospholipase A2 IIA and lipoprotein-PLA2 are implicated in vascular inflammation. These inflammatory responses promote atherosclerotic plaques, formation and release of the blood clot that can induce ischemic stroke. TNF-alpha and IL-1 alter lipid metabolism and stimulate production of eicosanoids, ceramide, and reactive oxygen species that potentiate CNS injuries and certain neurological disorders. Cholesterol is an important regulator of lipid organization and the precursor for neurosteroid biosynthesis. Low levels of neurosteroids were related to poor outcome in many brain pathologies. Apolipoprotein E is the principal cholesterol carrier protein in the brain, and the gene encoding the variant Apolipoprotein E4 is a significant risk factor for Alzheimer's disease. Parkinson's disease is to some degree caused by lipid peroxidation due to phospholipases activation. Niemann-Pick diseases A and B are due to acidic sphingomyelinase deficiency, resulting in sphingomyelin accumulation, while Niemann-Pick disease C is due to mutations in either the NPC1 or NPC2 genes, resulting in defective cholesterol transport and cholesterol accumulation. Multiple sclerosis is an autoimmune inflammatory demyelinating condition of the CNS. Inhibiting phospholipase A2 attenuated the onset and progression of experimental autoimmune encephalomyelitis. The endocannabinoid system is hypoactive in Huntington's disease. Ethyl-eicosapetaenoate showed promise in clinical trials. Amyotrophic lateral sclerosis causes loss of motorneurons. Cyclooxygenase-2 inhibition reduced spinal neurodegeneration in amyotrophic lateral sclerosis transgenic mice. Eicosapentaenoic acid supplementation provided improvement in schizophrenia patients, while the combination of (eicosapentaenoic acid + docosahexaenoic acid) provided benefit in bipolar disorders. The ketogenic diet where >90% of calories are derived from fat is an effective treatment for epilepsy. Understanding cytokine-induced changes in lipid metabolism will promote novel concepts and steer towards bench-to-bedside transition for therapies.


Subject(s)
Brain Diseases/metabolism , Brain Injuries/metabolism , Central Nervous System Diseases/metabolism , Lipid Metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Apolipoproteins E/metabolism , Atherosclerosis/complications , Atherosclerosis/metabolism , Bipolar Disorder/metabolism , Cholesterol/metabolism , Cytokines/metabolism , Group II Phospholipases A2/metabolism , Humans , Inflammation/physiopathology , Lipid Peroxidation , Membrane Lipids/metabolism , Niemann-Pick Diseases/metabolism , Oxidative Stress , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Reactive Oxygen Species/metabolism , Risk Factors , Schizophrenia/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Spinal Cord Injuries/metabolism , Stroke/etiology , Stroke/metabolism
4.
Neuroscience ; 146(3): 946-61, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17434680

ABSTRACT

Alterations in lipid metabolism play an integral role in neuronal death in cerebral ischemia. Here we used an in vitro model, oxygen-glucose deprivation (OGD) of rat pheochromocytoma (PC12) cells, and analyzed changes in phosphatidylcholine (PC) and sphingomyelin (SM) metabolism. OGD (4-8 h) of PC12 cells triggered a dramatic reduction in PC and SM levels, and a significant increase in ceramide. OGD also caused increases in phosphatidylcholine-phospholipase C (PC-PLC) and phospholipase D (PLD) activities and PLD2 protein expression, and reduction in cytidine triphosphate:phosphocholine cytidylyltransferase-alpha (CCTalpha, the rate-limiting enzyme in PC synthesis) protein expression and activity. Phospholipase A2 activity and expression were unaltered during OGD. Increased neutral sphingomyelinase activity during OGD could account for SM loss and increased ceramide. Surprisingly, treatment with PC-PLC inhibitor tricyclodecan-9-yl potassium xanthate (D609) aggravated cell death in PC12 cells during OGD. D609 was cytotoxic only during OGD; cell death could be prevented by inclusion of sera, glucose or oxygen. During OGD, D609 caused further loss of PC and SM, depletion of 1,2-diacylglycerol (DAG), increase in ceramide and free fatty acids (FFA), cytochrome c release from mitochondria, increases in intracellular Ca2+ ([Ca2+]i), poly-ADP ribose polymerase (PARP) cleavage and phosphatidylserine externalization, indicative of apoptotic cell death. Exogenous PC during OGD in PC12 cells with D609 attenuated PC, SM loss, restored DAG, attenuated ceramide levels, decreased cytochrome c release, PARP cleavage, annexin V binding, attenuated the increase in [Ca2+]i, FFA release, and significantly increased cell viability. Exogenous PC may have elicited these effects by restoring membrane PC levels. A tentative scheme depicting the mechanism of action of D609 (inhibiting PC-PLC, SM synthase, PC synthesis at the CDP-choline-1,2-diacylglycerol phosphocholine transferase (CPT) step and causing mitochondrial dysfunction) has been proposed based on our observations and literature.


Subject(s)
Bridged-Ring Compounds/pharmacology , Cell Hypoxia/physiology , Glucose/deficiency , Phospholipids/metabolism , Thiones/pharmacology , Animals , Annexin A5/metabolism , Calcium/metabolism , Cell Death , Cell Survival/physiology , Ceramides/metabolism , Culture Media , Cytochromes c/metabolism , Diglycerides/physiology , Fatty Acids, Nonesterified/metabolism , Glutathione/metabolism , L-Lactate Dehydrogenase/metabolism , Mitochondria/enzymology , Norbornanes , PC12 Cells , Phosphatidylcholines/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Rats , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Thiocarbamates , Trypan Blue
5.
Brain Res ; 1134(1): 199-205, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17204250

ABSTRACT

Cerebral ischemia initiates an inflammatory response in the brain that is associated with the induction of a variety of cytokines, including tumor necrosis factor-alpha (TNF-alpha) and interleukin-1alpha/beta (IL-1alpha/beta) that contributes to stroke injury. Transient middle cerebral artery occlusion (tMCAO) in spontaneously hypertensive rat (SHR) resulted in significant increases in TNF-alpha and IL-1beta levels. We have previously demonstrated up-regulation of secretory phospholipase A2 IIA (sPLA2 IIA) mRNA and protein expression, increased PLA2 activity, and loss of phosphatidylcholine after 1-h tMCAO and 24-h reperfusion in SHR. Treatment with TNF-alpha antibody or IL-1 receptor antagonist significantly attenuated infarction volume, sPLA2 IIA protein expression, PLA2 activity and significantly restored phosphatidylcholine levels after tMCAO. This suggests that cytokine induction up-regulates sPLA2 IIA protein expression, resulting in altered lipid metabolism that contributes to stroke injury.


Subject(s)
Cerebral Infarction/enzymology , Cytokines/metabolism , Encephalitis/enzymology , Ischemic Attack, Transient/enzymology , Phospholipases A/metabolism , Up-Regulation/immunology , Animals , Antibodies/pharmacology , Cerebral Infarction/immunology , Cerebral Infarction/physiopathology , Cytokines/antagonists & inhibitors , Cytokines/immunology , Down-Regulation/drug effects , Down-Regulation/immunology , Encephalitis/immunology , Encephalitis/physiopathology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Group II Phospholipases A2 , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/immunology , Interleukin-1alpha/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Ischemic Attack, Transient/immunology , Ischemic Attack, Transient/physiopathology , Male , Membrane Lipids/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/enzymology , Nerve Degeneration/immunology , Phosphatidylcholines/metabolism , Phospholipases A/genetics , Phospholipases A2 , Rats , Rats, Inbred SHR , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
6.
Future Lipidol ; 2(4): 403-422, 2007 Aug.
Article in English | MEDLINE | ID: mdl-18176634

ABSTRACT

Lipid metabolism is of particular interest due to its high concentration in CNS. The importance of lipids in cell signaling and tissue physiology is demonstrated by many CNS disorders and injuries that involve deregulated metabolism. The long suffering lipid field is gaining reputation and respect as evidenced through the Center of Biomedical Research Excellence in Lipidomics and Pathobiology (COBRE), Lipid MAPS (Metabolites And Pathways Strategy) Consortium sponsored by NIH, European initiatives for decoding the lipids through genomic approaches, and Genomics of Lipid-associated Disorder (GOLD) project initiated by Austrian government. This review attempts to provide an overview of the lipid imbalances associated with neurological disorders (Alzheimer's, Parkinson's; Niemann-Pick; Multiple sclerosis, Huntington, amyotrophic lateral sclerosis, schizophrenia, bipolar disorders and epilepsy) and CNS injury (Stroke, traumatic brain injury; and spinal cord injury) and a few provocative thoughts. Lipidomic analyses along with RNA silencing will provide new insights into the role of lipid intermediates in cell signaling and hopefully open new avenues for prevention or treatment options.

7.
AAPS J ; 8(2): E314-21, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16796382

ABSTRACT

Lipidomics is systems-level analysis and characterization of lipids and their interacting moieties. The amount of information in the genomic and proteomic fields is greater than that in the lipidomics field, because of the complex nature of lipids and the limitations of tools for analysis. The main innovation during recent years that has spurred advances in lipid analysis has been the development of new mass spectroscopic techniques, particularly the "soft ionization" techniques electrospray ionization and matrix-assisted laser desorption/ionization. Lipid metabolism may be of particular importance for the central nervous system, as it has a high concentration of lipids. The crucial role of lipids in cell signaling and tissue physiology is demonstrated by the many neurological disorders, including bipolar disorders and schizophrenia, and neurodegenerative diseases such as Alzheimer's, Parkinson's, and Niemann-Pick diseases, that involve deregulated lipid metabolism. Altered lipid metabolism is also believed to contribute to cerebral ischemic (stroke) injury. Lipidomics will provide a molecular signature to a certain pathway or a disease condition. Lipidomic analyses (characterizing complex mixtures of lipids and identifying previously unknown changes in lipid metabolism) together with RNA silencing, using small interfering RNA (siRNA), may provide powerful tools to elucidate the specific roles of lipid intermediates in cell signaling and open new opportunities for drug development.


Subject(s)
Brain Diseases/physiopathology , Brain Injuries/physiopathology , Brain/physiology , Lipids/physiology , Animals , Brain/physiopathology , Genomics , Humans , Lipids/analysis
8.
Free Radic Biol Med ; 40(3): 376-87, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16443152

ABSTRACT

Ischemic stroke is caused by obstruction of blood flow to the brain, resulting in energy failure that initiates a complex series of metabolic events, ultimately causing neuronal death. One such critical metabolic event is the activation of phospholipase A2 (PLA2), resulting in hydrolysis of membrane phospholipids and release of free fatty acids including arachidonic acid, a metabolic precursor for important cell-signaling eicosanoids. PLA2 enzymes have been classified as calcium-dependent cytosolic (cPLA2) and secretory (sPLA2) and calcium-independent (iPLA2) forms. Cardiolipin hydrolysis by mitochondrial sPLA2 disrupts the mitochondrial respiratory chain and increases production of reactive oxygen species (ROS). Oxidative metabolism of arachidonic acid also generates ROS. These two processes contribute to formation of lipid peroxides, which degrade to reactive aldehyde products (malondialdehyde, 4-hydroxynonenal, and acrolein) that covalently bind to proteins/nucleic acids, altering their function and causing cellular damage. Activation of PLA2 in cerebral ischemia has been shown while other studies have separately demonstrated increased lipid peroxidation. To the best of our knowledge no study has directly shown the role of PLA2 in lipid peroxidation in cerebral ischemia. To date, there are very limited data on PLA2 protein by Western blotting after cerebral ischemia, though some immunohistochemical studies (for cPLA2 and sPLA2) have been reported. Dissecting the contribution of PLA2 to lipid peroxidation in cerebral ischemia is challenging due to multiple forms of PLA2, cardiolipin hydrolysis, diverse sources of ROS arising from arachidonic acid metabolism, catecholamine autoxidation, xanthine oxidase activity, mitochondrial dysfunction, activated neutrophils coupled with NADPH oxidase activity, and lack of specific inhibitors. Although increased activity and expression of various PLA2 isoforms have been demonstrated in stroke, more studies are needed to clarify the cellular origin and localization of these isoforms in the brain, their responses in cerebral ischemic injury, and their role in oxidative stress.


Subject(s)
Brain Ischemia/metabolism , Lipid Peroxidation , Oxidative Stress , Phospholipases A/metabolism , Reactive Oxygen Species/metabolism , Animals , Arachidonic Acid/metabolism , Humans , Phospholipases A2
9.
Brain Res ; 1058(1-2): 193-7, 2005 Oct 05.
Article in English | MEDLINE | ID: mdl-16153613

ABSTRACT

Cytidine-5'-diphosphocholine (CDP-choline, Citicoline, Somazina) is in clinical use (intravenous administration) for stroke treatment in Europe and Japan, while USA phase III stroke clinical trials (oral administration) were disappointing. Others showed that CDP-choline liposomes significantly increased brain uptake over the free drug in cerebral ischemia models. Liposomes were formulated as DPPC, DPPS, cholesterol, GM(1) ganglioside; 7/4/7/1.57 molar ratio or 35.8/20.4/35.8/8.0 mol%. GM(1) ganglioside confers long-circulating properties to the liposomes by suppressing phagocytosis. CDP-choline liposomes deliver the agent intact to the brain, circumventing the rate-limiting, cytidine triphosphate:phosphocholine cytidylyltransferase in phosphatidylcholine synthesis. Our data show that CDP-choline liposomes significantly ( P < 0.01) decreased cerebral infarction (by 62%) compared to the equivalent dose of free CDP-choline (by 26%) after 1 h focal cerebral ischemia and 24 h reperfusion in spontaneously hypertensive rats. Beneficial effects of CDP-choline liposomes in stroke may derive from a synergistic effect between the phospholipid components of the liposomes and the encapsulated CDP-choline.


Subject(s)
Brain Infarction/drug therapy , Brain/drug effects , Choline/administration & dosage , Cytidine Diphosphate Choline/administration & dosage , Liposomes/pharmacology , Stroke/drug therapy , Animals , Brain/metabolism , Brain/pathology , Brain Infarction/metabolism , Brain Infarction/physiopathology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cholesterol/administration & dosage , Disease Models, Animal , Drug Combinations , Drug Synergism , G(M1) Ganglioside/administration & dosage , Liposomes/therapeutic use , Male , Phagocytosis/drug effects , Phagocytosis/physiology , Phosphatidylcholines/biosynthesis , Rats , Rats, Inbred SHR , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Stroke/metabolism , Stroke/physiopathology , Treatment Outcome
10.
Neurochem Res ; 30(1): 15-23, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15756928

ABSTRACT

Brain phosphatidylcholine (PC) levels are regulated by a balance between synthesis and hydrolysis. Pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 (IL-1alpha/beta) activate phospholipase A(2) (PLA(2)) and PC-phospholipase C (PC-PLC) to hydrolyze PC. PC hydrolysis by PLA(2) releases free fatty acids including arachidonic acid, and lyso-PC, an inhibitor of CTP-phosphocholine cytidylyltransferase (CCT). Arachidonic acid metabolism by cyclooxygenases/lipoxygenases is a significant source of reactive oxygen species. CDP-choline might increase the PC levels by attenuating PLA(2) stimulation and loss of CCT activity. TNF-alpha also stimulates proteolysis of CCT. TNF-alpha and IL-1beta are induced in brain ischemia and may disrupt PC homeostasis by increasing its hydrolysis (increase PLA(2) and PC-PLC activities) and inhibiting its synthesis (decrease CCT activity). The beneficial effects of CDP-choline may result by counteracting TNF-alpha and/or IL-1 mediated events, integrating cytokine biology and lipid metabolism. Re-evaluation of CDP-choline phase III stroke clinical trial data is encouraging and future trails are warranted. CDP-choline is non-xenobiotic, safe, well tolerated, and can be considered as one of the agents in multi-drug treatment of stroke.


Subject(s)
Cytidine Diphosphate Choline/metabolism , Cytidine Diphosphate Choline/therapeutic use , Stroke/drug therapy , Stroke/metabolism , Animals , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Humans
11.
Neuroscience ; 115(2): 587-602, 2002.
Article in English | MEDLINE | ID: mdl-12421624

ABSTRACT

Cardiolipin, a lipid of the mitochondrial inner membrane, is lost from many types of cells during apoptotic death. Here we show that the cardiolipin content of nerve growth factor (NGF)-deprived rat sympathetic neurons undergoing apoptotic death in cell culture decreased before extensive loss of mitochondria from the cells. By 18-24 h after NGF deprivation, many neurons did not stain with the cardiolipin-specific dye, Nonyl Acridine Orange, suggesting complete loss of cardiolipin. Gas chromatography confirmed the decline of cardiolipin content in NGF-deprived neurons. Electron microscopy and immunoblots for the mitochondrial-specific protein, heat shock protein 60 (HSP60), revealed that there was only a slight decrease in mitochondrial mass at this time. Cardiolipin loss after NGF deprivation was concurrent with increased production of mitochondrial-derived reactive oxygen species [Kirkland, R.A., Franklin, J.L., 2001. J. Neurosci. 21, 1949-1963] and increased lipid peroxidation. Compounds having antioxidant effects blocked peroxidation, loss of cardiolipin, and the decrease of mitochondrial mass in NGF-deprived neurons. These compounds also blocked an increase in the number of lysosomes and autophagosomes in NGF-deprived cells. The findings reported here show that the important mitochondrial inner membrane lipid, cardiolipin, is lost from mitochondria during neuronal apoptosis and that this loss occurs before significant loss of mitochondria from cells. They suggest that the loss of cardiolipin is mediated by free radical oxygen.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Cardiolipins/metabolism , Mitochondria/metabolism , Neurons/cytology , Animals , Apoptosis/drug effects , Cells, Cultured , Cytochrome c Group/metabolism , Female , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/ultrastructure , Nerve Growth Factor/pharmacology , Neurons/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
12.
J Neurosci Res ; 70(2): 133-9, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12271462

ABSTRACT

Citicoline, an intermediate in the biosynthesis of phosphatidylcholine (PtdCho), has shown beneficial effects in various CNS injury models and neurodegenerative diseases. PtdCho hydrolysis by phospholipase A(2) (PLA(2)) after cerebral ischemia and reperfusion yields arachidonic acid (ArAc) and lyso-PtdCho. ArAc oxidative metabolism results in formation of reactive oxygen species and lipid peroxides. Lyso-PtdCho could inhibit activity of cytidine triphosphate-phosphocholine cytidylyltransferase (the rate-limiting enzyme in PtdCho biosynthesis), resulting in impaired PtdCho synthesis. Citicoline significantly increased glutathione levels and attenuated release of ArAc and the loss of PtdCho, cardiolipin, and sphingomyelin following transient cerebral ischemia. These effects could be explained by an effect of citicoline on PLA(2). Based on these observations, a mechanism has been hypothesized. This Mini-Review summarizes recent experimental data on the effects of citicoline in cerebral ischemia and evaluates several factors that might have hindered efficacy of citicoline in stroke clinical trials in the United States. Clinical stroke trials of citicoline in Europe and Japan have demonstrated beneficial effects. U.S. trials shown only marginal effects, which might be due to the 24 hr time window, the dose and route of administration, and the stringency of the primary outcome parameters. Recent evaluation of U.S. clinical data suggests that reduction of infarct growth may be a more sensitive measure of the citicoline effect than improvement on the NIH Stroke Scale (NIHSS) by > or =7 points. The citicoline neuroprotective mechanism has not been clearly identified, and its potential in stroke treatment might still be fully recognized in the United States. The clinical efficacy of citicoline should be examined further in light of the recent phase III stroke clinical trials and experimental data for cerebral ischemia.


Subject(s)
Brain Ischemia/drug therapy , Cytidine Diphosphate Choline/pharmacology , Cytidine Diphosphate Choline/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Animals , Arachidonic Acid/metabolism , Brain Ischemia/metabolism , Clinical Trials as Topic , Disease Models, Animal , Humans , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/metabolism , Phosphatidylcholines/metabolism , Phospholipases A/drug effects , Phospholipases A/metabolism , Rats , Treatment Outcome
13.
J Neurochem ; 80(1): 12-23, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11796739

ABSTRACT

Cytidine-5'-diphosphocholine (citicoline or CDP-choline), an intermediate in the biosynthesis of phosphatidylcholine (PtdCho), has shown beneficial effects in a number of CNS injury models and pathological conditions of the brain. Citicoline improved the outcome in several phase-III clinical trials of stroke, but provided inconclusive results in recent clinical trials. The therapeutic action of citicoline is thought to be caused by stimulation of PtdCho synthesis in the injured brain, although the experimental evidence for this is limited. This review attempts to shed some light on the properties of citicoline that are responsible for its effectiveness. Our studies in transient cerebral ischemia suggest that citicoline might enhance reconstruction (synthesis) of PtdCho and sphingomyelin, but could act by inhibiting the destructive processes (activation of phospholipases). Citicoline neuroprotection may include: (i) preserving cardiolipin (an exclusive inner mitochondrial membrane component) and sphingomyelin; (ii) preserving the arachidonic acid content of PtdCho and phosphatidylethanolamine; (iii) partially restoring PtdCho levels; (iv) stimulating glutathione synthesis and glutathione reductase activity; (v) attenuating lipid peroxidation; and (vi) restoring Na(+)/K(+)-ATPase activity. These observed effects of citicoline could be explained by the attenuation of phospholipase A(2) activation. Based on these findings, a singular unifying mechanism has been hypothesized. Citicoline also provides choline for synthesis of neurotransmitter acetylcholine, stimulation of tyrosine hydroxylase activity and dopamine release.


Subject(s)
Brain Ischemia/physiopathology , Cytidine Diphosphate Choline/pharmacology , Neuroprotective Agents/pharmacology , Animals , Cytidine Diphosphate Choline/metabolism , Humans , Neuroprotective Agents/metabolism
14.
Stroke ; 32(10): 2376-81, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11588329

ABSTRACT

BACKGROUND AND PURPOSE: Cytidine-5'-diphosphocholine (citicoline or CDP-choline) is an essential intermediate in the biosynthesis of phosphatidylcholine, an important component of the neural cell membrane. Citicoline provided significant neuroprotection after transient forebrain ischemia in gerbils. This study was undertaken to examine changes and effects of citicoline on phospholipids and glutathione synthesis after transient cerebral ischemia and reperfusion. METHODS: Ten-minute transient forebrain ischemia was induced by bilateral carotid artery occlusion in male Mongolian gerbils with reperfusion up to 6 days. Citicoline (500 mg/kg IP in saline) was given to gerbils just after the end of ischemia, at 3-hour reperfusion, and daily thereafter until 1 day before euthanasia. Hippocampal lipids were extracted and analyzed by thin-layer and gas chromatography. Glutathione was measured by using an enzymatic recycling assay. Glutathione reductase activity was determined by measuring NADPH oxidation. RESULTS: Significant decreases in phospholipids occurred at 1-day reperfusion. Citicoline significantly restored the phosphatidylcholine, sphingomyelin, and cardiolipin levels but did not affect phosphatidylinositol and phosphatidylserine at 1 day. The phospholipids returned to sham levels over days 2 to 6 and were unaffected by citicoline. Ceramide levels significantly increased by 3 and 6 days of reperfusion and were unaltered by citicoline. Ischemia resulted in significant decreases in glutathione and glutathione reductase activity over 3 days of reperfusion. Citicoline significantly increased total glutathione and glutathione reductase activity and decreased the glutathione oxidation ratio, an indicator of glutathione redox status. CONCLUSIONS: Our data indicated that the effects of citicoline on phospholipids occurred primarily during the first day of reperfusion, with effects on glutathione being important over the 3-day reperfusion period.


Subject(s)
Cytidine Diphosphate Choline/pharmacology , Glutathione/metabolism , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/metabolism , Phospholipids/metabolism , Animals , Disease Models, Animal , Fatty Acids/metabolism , Gerbillinae , Glutathione Reductase/metabolism , Hippocampus/chemistry , Hippocampus/metabolism , Ischemic Attack, Transient/pathology , Male , Prosencephalon/blood supply , Prosencephalon/metabolism , Prosencephalon/pathology , Reperfusion
15.
Brain Res ; 893(1-2): 268-72, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11223016

ABSTRACT

Ten min forebrain ischemia/1-day reperfusion resulted in significant decreases in total phosphatidylcholine (PtdCho), phosphatidylinositol (PtdIns), and cardiolipin in gerbil hippocampus. CDP-choline restored cardiolipin levels, arachidonic acid content of PtdCho, partially but significantly restored total PtdCho, and had no effect on PtdIns. These data suggest that CDP-choline prevented the activation of phospholipase A(2) (rather than inhibiting phospholipase A(2) activity) but did not affect activities of PtdCho-phospholipases C and/or D, or phosphoinositide-phospholipase C. CDP-choline also provided significant protection for hippocampal CA(1) neurons.


Subject(s)
Cytidine Diphosphate Choline/administration & dosage , Ischemic Attack, Transient/metabolism , Nootropic Agents/administration & dosage , Phospholipases/metabolism , Prosencephalon/metabolism , Animals , Arachidonic Acid/metabolism , Cardiolipins/chemistry , Cardiolipins/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Fatty Acids/analysis , Fatty Acids/metabolism , Gerbillinae , Hippocampus/drug effects , Hippocampus/metabolism , Injections, Intraperitoneal , Isoenzymes/metabolism , Male , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Phospholipase D/metabolism , Phospholipases A/metabolism , Prosencephalon/blood supply , Type C Phospholipases/metabolism
16.
Neurosci Lett ; 293(1): 1-4, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11065123

ABSTRACT

Stimulation of group I metabotropic glutamate receptors (mGluR 1 and 5) activates G-protein coupled-phospholipase C (PLC) to release 1,2-diacylglycerol (DAG) and arachidonic acid (ArAc). To elucidate the role of group I mGluR, we tested the effects of (S)-alpha-methyl-4-carboxy-phenylglycine (MCPG, mGluR 1 and 5 antagonist), 1-aminoindan-1,5-dicarboxylic acid (AIDA, mGluR 1a specific antagonist) and 2-methyl-6-(phenylethynyl) pyridine (MPEP, mGluR 5 antagonist) on ArAc release and neuronal survival after transient forebrain ischemia in gerbils. Ischemia resulted in (a) significant release of ArAc at 1-day reperfusion and (b) significant neuronal death in the hippocampal CA1 subfield after 6-day reperfusion. MCPG and MPEP decreased ArAc release and also significantly increased neuronal survival. AIDA was less effective in decreasing ArAc release and had no effect on neuronal death. These results suggest that activation of mGluR 5 may be an important pathway in ArAc release and neuronal death after transient ischemia.


Subject(s)
Benzoates/therapeutic use , Brain Ischemia/drug therapy , Glycine/therapeutic use , Indans/therapeutic use , Neuroprotective Agents/therapeutic use , Prosencephalon/blood supply , Pyridines/therapeutic use , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Arachidonic Acid/antagonists & inhibitors , Brain/metabolism , Brain/physiopathology , Brain Ischemia/metabolism , Cell Survival/drug effects , Excitatory Amino Acid Antagonists/therapeutic use , Gerbillinae , Glycine/analogs & derivatives , Male , Neurons/physiology
17.
J Neurochem ; 75(6): 2528-35, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11080206

ABSTRACT

We have previously demonstrated that cytidine 5'-diphosphocholine (CDP-choline or citicoline) attenuated arachidonic acid (ArAc) release and provided significant protection for the vulnerable hippocampal CA(1) neurons of the cornu ammonis after transient forebrain ischemia of gerbil. ArAc is released by the activation of phospholipases and the alteration of phosphatidylcholine (PtdCho) synthesis. Released ArAc is metabolized by cyclooxygenases/lipoxygenases to form eicosanoids and reactive oxygen species (ROS). ROS contribute to neurotoxicity through generation of lipid peroxides and the cytotoxic byproducts 4-hydroxynonenal and acrolein. ArAc can also stimulate sphingomyelinase to produce ceramide, a potent pro-apoptotic agent. In the present study, we examined the changes and effect of CDP-choline on ceramide and phospholipids including PtdCho, phosphatidylethanolamine (PtdEtn), phosphatidylinositol (PtdIns), phosphatidylserine (PtdSer), sphingomyelin, and cardiolipin (an exclusive inner mitochondrial membrane lipid essential for electron transport) following ischemia/1-day reperfusion. Our studies indicated significant decreases in total PtdCho, PtdIns, PtdSer, sphingomyelin, and cardiolipin and loss of ArAc from PtdEtn in gerbil hippocampus after 10-min forebrain ischemia/1-day reperfusion. CDP-choline (500 mg/kg i.p. immediately after ischemia and at 3-h reperfusion) significantly restored the PtdCho, sphingomyelin, and cardiolipin levels as well as the ArAc content of PtdCho and PtdEtn but did not affect PtdIns and PtdSer. These data suggest multiple beneficial effects of CDP-choline: (1) stabilizing the cell membrane by restoring PtdCho and sphingomyelin (prominent components of outer cell membrane), (2) attenuating the release of ArAc and limiting its oxidative metabolism, and (3) restoring cardiolipin levels.


Subject(s)
Cytidine Diphosphate Choline/metabolism , Ischemic Attack, Transient/metabolism , Phospholipids/metabolism , Prosencephalon/metabolism , Animals , Arachidonic Acid/metabolism , Cardiolipins/metabolism , Ceramides/metabolism , Cytidine Diphosphate Choline/pharmacology , Gerbillinae , Male , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Phosphatidylinositols/metabolism , Phosphatidylserines/metabolism , Prosencephalon/blood supply , Reperfusion , Sphingomyelins/metabolism
18.
J Neurochem ; 74(3): 1106-11, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10693942

ABSTRACT

The polyamine system is very sensitive to different pathological states of the brain and is perturbed after CNS injury. The main modifications are significant increases in ornithine decarboxylase activity and an increase in tissue putrescine levels. Previously we have shown that the specific polyamine oxidase (PAO) inhibitor N1,N4-bis(2,3-butadienyl)-1,4-butanediamine (MDL 72527) reduced the tissue putrescine levels, edema, and infarct volume after transient focal cerebral ischemia in spontaneously hypertensive rats and traumatic brain injury of Sprague-Dawley rats. In the present study, N1-acetyl-spermidine accumulation was greater in injured brain regions compared with sham or contralateral regions following inhibition of PAO by MDL 72527. This indicates spermidine/spermine-N1-acetyltransferase (SSAT) activation after CNS injury. The observed increase in N1-acetylspermidine levels at 1 day after CNS trauma paralleled the decrease in putrescine levels after treatment with MDL 72527. This suggests that the increased putrescine formation at 1 day after CNS injury is mediated by the SSAT/PAO pathway, consistent with increased SSAT mRNA after transient ischemia.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Spermidine/analogs & derivatives , Animals , Arterial Occlusive Diseases/metabolism , Blood-Brain Barrier , Cerebral Arteries , Gerbillinae , Ischemic Attack, Transient/metabolism , Male , Prosencephalon/blood supply , Putrescine/analogs & derivatives , Putrescine/antagonists & inhibitors , Putrescine/metabolism , Putrescine/pharmacokinetics , Putrescine/pharmacology , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Spermidine/metabolism
19.
J Neurosci Res ; 58(5): 697-705, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10561698

ABSTRACT

CDP-choline is a rate-limiting intermediate in the biosynthesis of phosphatidylcholine (PtdCho), an important component of the neural cell membrane. The ability of CDP-choline to alter phospholipid metabolism is an important function in the treatment of ischemic injury. Exogenous treatment with CDP-choline stimulates PtdCho synthesis and prevents release of free fatty acids (FFA), especially arachidonic acid (AA), after ischemia/reperfusion. Phase III clinical trials of CDP-choline in the treatment of stroke are currently underway. Here we report the neuroprotection by CDP-choline in transient forebrain ischemia of gerbils. CDP-choline significantly attenuated the blood-brain barrier (BBB) dysfunction after ischemia with 6-hr reperfusion, and considerably reduced the increase of AA in FFA and leukotriene C(4) (LTC(4)) synthesis at 1 day. Edema was significantly elevated after 1 and 2 days, but attained maximum at 3-day reperfusion. CDP-choline substantially attenuated edema at 3 days. Ischemia resulted in 80 +/- 8% CA(1) hippocampal neuronal death after 6-day reperfusion, and CDP-choline provided 65 +/- 6% neuroprotection. CDP-choline may act by increasing PtdCho synthesis via two pathways: (1) conversion of 1, 2-diacylglycerol to PtdCho, and (2) biosynthesis of S-adenosyl-L-methionine, thus stabilizing the membrane and reducing AA release and metabolism to leukotriene C(4). This would result in decreased toxicity due to AA, leukotrienes, oxygen radicals, lipid peroxidation, and altered glutamate uptake, thus limiting BBB dysfunction, edema and providing neuroprotection.


Subject(s)
Cytidine Diphosphate Choline/pharmacology , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/pharmacology , Prosencephalon/physiopathology , Animals , Arachidonic Acid/metabolism , Blood-Brain Barrier/drug effects , Cerebrovascular Circulation/physiology , Gerbillinae , Hippocampus/drug effects , Hippocampus/pathology , Ischemic Attack, Transient/physiopathology , Leukotriene C4/metabolism , Neurons/drug effects , Phosphatidylcholines/metabolism
20.
Neurochem Res ; 24(10): 1225-32, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10492517

ABSTRACT

Accumulation of arachidonic acid (AA) is greatest in brain regions most sensitive to transient ischemia. Free AA released after ischemia is either: 1) reincorporated into the membrane phospholipids, or 2) oxidized during reperfusion by lipoxygenases and cyclooxygenases, producing leukotrienes (LT), prostaglandins, thromboxanes and oxygen radicals. AA, its metabolite LTC4 and lipid peroxides (generated during AA metabolism) have been implicated in the blood-brain barrier (BBB) dysfunction, edema and neuronal death after ischemia/reperfusion. This report describes the time course of AA release, LTC4 accumulation and association with the physiological outcome during transient cerebral ischemia of gerbils. Significant amount of AA was detected immediately after 10 min ischemia (0 min reperfusion) which returned to sham levels within 30 min reperfusion. A later release of AA occurred after 1 d. LTC4 levels were elevated at 0-6 h and 1 d after ischemia. Increased lipid peroxidation due to AA metabolism was observed between 2-6 h. BBB dysfunction occurred at 6 h. Significant edema developed at 1 and 2 d after ischemia and reached maximum at 3 d. Ischemia resulted in approximately 80% neuronal death in the CA1 hippocampal region. Pretreatment with a 5-lipoxygenase inhibitor, AA861 resulted in significant attenuation of LTC4 levels (Baskaya et al. 1996. J. Neurosurg. 85: 112-116) and CA1 neuronal death. Accumulation of AA and LTC4, together with highly reactive oxygen radicals and lipid peroxides, may alter membrane permeability, resulting in BBB dysfunction, edema and ultimately to neuronal death.


Subject(s)
Arachidonic Acid/physiology , Ischemic Attack, Transient/physiopathology , Leukotriene C4/physiology , Animals , Blood-Brain Barrier , Brain Edema/metabolism , Brain Edema/pathology , Gerbillinae , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Lipid Peroxidation , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...