Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 15(1): 272, 2018 Sep 20.
Article in English | MEDLINE | ID: mdl-30236122

ABSTRACT

BACKGROUND: Prostaglandins are products of the cyclooxygenase pathway, which is implicated in Parkinson's disease (PD). Limited knowledge is available on mechanisms by which prostaglandins contribute to PD neurodegeneration. To address this gap, we focused on the prostaglandin PGD2/J2 signaling pathway, because PGD2 is the most abundant prostaglandin in the brain, and the one that increases the most under pathological conditions. Moreover, PGJ2 is spontaneously derived from PGD2. METHODS: In this study, we determined in rats the impact of unilateral nigral PGJ2-microinfusions on COX-2, lipocalin-type PGD2 synthase (L-PGDS), PGD2/J2 receptor 2 (DP2), and 15 hydroxyprostaglandin dehydrogenase (15-PGDH). Nigral dopaminergic (DA) and microglial distribution and expression levels of these key factors of the prostaglandin D2/J2 pathway were evaluated by immunohistochemistry. PGJ2-induced motor deficits were assessed with the cylinder test. We also determined whether oral treatment with ibuprofen improved the PD-like pathology induced by PGJ2. RESULTS: PGJ2 treatment induced progressive PD-like pathology in the rats. Concomitant with DA neuronal loss in the substantia nigra pars compacta (SNpc), PGJ2-treated rats exhibited microglia and astrocyte activation and motor deficits. In DA neurons, COX-2, L-PGDS, and 15-PGDH levels increased significantly in PGJ2-treated rats compared to controls, while DP2 receptor levels were unchanged. In microglia, DP2 receptors were basically non-detectable, while COX-2 and L-PGDS levels increased upon PGJ2-treatment, and 15-PGDH remained unchanged. 15-PGDH was also detected in oligodendrocytes. Notably, ibuprofen prevented most PGJ2-induced PD-like pathology. CONCLUSIONS: The PGJ2-induced rat model develops progressive PD pathology, which is a hard-to-mimic aspect of this disorder. Moreover, prevention of most PGJ2-induced PD-like pathology with ibuprofen suggests a positive feedback mechanism between PGJ2 and COX-2 that could lead to chronic neuroinflammation. Notably, this is the first study that analyzes the nigral dopaminergic and microglial distribution and levels of factors of the PGD2/J2 signaling pathway in rodents. Our findings support the notions that upregulation of COX-2 and L-PGDS may be important in the PGJ2 evoked PD-like pathology, and that neuronal DP2 receptor antagonists and L-PGDS inhibitors may be novel pharmacotherapeutics to relieve neuroinflammation-mediated neurodegeneration in PD, circumventing the adverse side effects of cyclooxygenase inhibitors.


Subject(s)
Encephalitis/complications , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Signal Transduction/physiology , Substantia Nigra/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/toxicity , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/drug therapy , Encephalitis/metabolism , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Functional Laterality/drug effects , Ibuprofen/therapeutic use , Male , Microglia/drug effects , Neurons/drug effects , Neurons/metabolism , Parkinsonian Disorders/etiology , Parkinsonian Disorders/pathology , Phosphopyruvate Hydratase/metabolism , Prostaglandin D2/toxicity , Psychomotor Performance/drug effects , Rats , Signal Transduction/drug effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
2.
Mol Neurobiol ; 55(3): 2227-2248, 2018 03.
Article in English | MEDLINE | ID: mdl-28299574

ABSTRACT

Prostaglandins (PGs) are divided into conventional PGs, e.g., PGD2, and cyclopentenone-type PGs, e.g., 15-deoxy-Δ12,14 prostaglandin J2 (15d-PGJ2). PGD2 is non-enzymatically metabolized to PGJ2, Δ12-PGJ2, and 15d-PGJ2. In the central nervous system, 15d-PGJ2 differentiates embryonic midbrain cells into dopaminergic neuronal cells via its nuclear peroxysome proliferator-activated receptor-γ (PPARγ). 15d-PGJ2 exerts conflict actions: proinflammatory and anti-inflammatory activities. In the brain, 15d-PGJ2 possesses opposite functions as a neuroprotectant at low concentrations and a neurotoxicant at high concentrations in the brain. PPARγ contributes to the neuroprotective effect of 15d-PGJ2 but not to the neurotoxic effect. Its membrane receptor, chemoattractant receptor-homologous molecule expressed on T-helper type 2 cells (CRTH2), is not also involved in the neurotoxicity of 15d-PGJ2. 15d-PGJ2 induces neuronal apoptosis via inactivating ubiquitin proteasome pathway and activating caspase cascade. Alternatively, 15d-PGJ2 downregulates phosphoinositide 3-kinase (PI3K)-Akt pathway and suppresses neurite outgrowth. 15d-PGJ2 possesses α,ß-unsaturated ketone moiety in its cyclopentenone ring and acts an endogenous electrophile. By the Michael addition reaction, 15d-PGJ2 is covalently bound to cellular nucleophiles, such as free cysteine residues of proteins that regulate intracellular signaling pathways. There are specific binding sites of [3H]15d-PGJ2 in the plasma membrane of cerebral cortices. Besides CRTH2, plasmalemmal glycolytic enzymes, respiratory chain enzymes, molecular chaperones, adaptor proteins and cytoskeletons are identified as membrane targets for 15d-PGJ2. In the present review, we provide evidences for pathophysiological roles of 15d-PGJ2 in the central nervous system and neurological diseases.


Subject(s)
Central Nervous System/physiology , Immunologic Factors/physiology , Immunologic Factors/toxicity , Nervous System Diseases/physiopathology , Prostaglandin D2/analogs & derivatives , Animals , Cell Survival/drug effects , Cell Survival/physiology , Central Nervous System/drug effects , Central Nervous System/pathology , Humans , Nervous System Diseases/chemically induced , Nervous System Diseases/pathology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Prostaglandin D2/physiology , Prostaglandin D2/toxicity
3.
J Biol Chem ; 292(1): 205-216, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27872193

ABSTRACT

Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor that plays a crucial role in protection of cells from electrophile-induced toxicity through up-regulating phase II detoxifying enzymes and phase III transporters. We previously reported that oxidative stress induces up-regulation of interleukin-11 (IL-11), a member of the IL-6 family that ameliorates acetaminophen-induced liver toxicity. However, a role for IL-11 in protection of cells from electrophile-induced toxicity remains unclear. Here we show that an environmental electrophile, 1,2-naphthoquinone (1,2-NQ), but not 15d-prostaglandin J2 (PGJ2) or tert-butylhydroxyquinone (tBHQ), induced IL-11 production. Consistent with a crucial role for prolonged ERK activation in H2O2-induced IL-11 production, 1,2-NQ, but not 15d-PGJ2 or tBHQ, elicited prolonged ERK activation. Conversely, inhibition of the ERK pathway by a MEK inhibitor completely blocked 1,2-NQ-induced IL-11 production at both protein and mRNA levels, further substantiating an intimate cross-talk between ERK activation and 1,2-NQ-induced IL-11 production. Promoter analysis of the Il11 gene revealed that two AP-1 sites were essential for 1,2-NQ-induced promoter activities. Among various members of the AP-1 family, Fra-1 was up-regulated by 1,2-NQ, and its up-regulation was blocked by a MEK inhibitor. Although NRF2 was not required for H2O2-induced IL11 up-regulation, NRF2 was essential for 1,2-NQ-induced IL11 up-regulation by increasing Fra-1 proteins possibly through promoting mRNA translation of FOSL1 Finally, intraperitoneal administration of 1,2-NQ induced body weight loss in wild-type mice, which was further exacerbated in Il11ra1-/- mice compared with Il11ra1+/- mice. Together, both Fra-1 and NRF2 play crucial roles in IL-11 production that protects cells from 1,2-NQ intestinal toxicity.


Subject(s)
Interleukin-11/biosynthesis , Intestinal Diseases/prevention & control , NF-E2-Related Factor 2/metabolism , Naphthoquinones/toxicity , Peritonitis/prevention & control , Prostaglandin D2/analogs & derivatives , Animals , Antineoplastic Agents/toxicity , Cells, Cultured , Gene Expression Regulation/drug effects , HEK293 Cells , Hep G2 Cells , Humans , Hydrogen Peroxide/pharmacology , Interleukin-11 Receptor alpha Subunit/physiology , Intestinal Diseases/chemically induced , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Oxidants/pharmacology , Oxidative Stress/drug effects , Peritonitis/chemically induced , Peritonitis/metabolism , Peritonitis/pathology , Prostaglandin D2/toxicity , Reactive Oxygen Species/metabolism
4.
Neuropharmacology ; 113(Pt A): 416-425, 2017 02.
Article in English | MEDLINE | ID: mdl-27771378

ABSTRACT

15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) induces neuronal cell death via apoptosis independently of its receptors. 15d-PGJ2 inhibits growth factor-induced cell proliferation of primary astrocytes via down-regulating phosphoinositide 3-kinase (PI3K)-Akt pathway. Although 15d-PGJ2-reduced cell viability is accompanied with attenuation of the PI3K signaling in neuroblastoma, it has not been sufficiently clarified how 15d-PGJ2 induces cell death in primary neurons. Here, we found that 15d-PGJ2 exhibited neurotoxicity via inhibiting the PI3K signaling in the primary culture of rat cortical neurons. A PI3K inhibitor induced neuronal cell death regardless serum throughout maturation, confirming that PI3K is required for neuronal cell survival. The inhibitor disrupted neuronal cell bodies, shortened neurites thinly, damaged plasma membranes and activated caspase-3 similarly to 15d-PGJ2. Little additive or synergistic neurotoxicity was detected between 15d-PGJ2 and the PI3K inhibitor. A PI3K activator prevented neurons from undergoing the 15d-PGJ2-induced cell death in vitro. In vivo, the PI3K signaling is required for contextual memory retrieval, which was impaired by bilateral injection of 15d-PGJ2 into hippocampus. The activator suppressed the 15d-PGJ2-impaired memory retrieval significantly. In neurons as well as primary astrocytes and neuroblastomas, 15d-PGJ2 exhibited cytotoxicity via suppressing the PI3K-Akt pathway in vivo and in vitro.


Subject(s)
Neurons/drug effects , Neurons/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Prostaglandin D2/analogs & derivatives , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Dose-Response Relationship, Drug , Fear/drug effects , Fear/physiology , Female , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Injections, Intraventricular , Male , Neurons/pathology , Pregnancy , Prostaglandin D2/administration & dosage , Prostaglandin D2/toxicity , Rats , Rats, Wistar
5.
Naunyn Schmiedebergs Arch Pharmacol ; 389(7): 809-13, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27185495

ABSTRACT

Recent studies have shown that prostaglandin D2 (PGD2) and its nonenzymatic metabolite, 15-deoxy-Δ(12,14)-prostaglandin J2 (15-dPGJ2), inhibit in vitro growth of explanted human hair follicles and inhibit hair growth in mice through the GPR44 (DP2). However, the underlying mechanism is still unclear. In this study, we first investigated the expression of DP2 in human hair follicles and in cultured follicular cells. We found that DP2 is strongly expressed in the outer root sheath (ORS) cells and weakly expressed in the dermal papilla (DP) cells. We observed slight growth stimulation when ORS and DP cells were treated with PGD2. We also observed slight growth stimulation when DP and ORS cells were treated with low concentrations (0.5 and 1 µM) of 15-dPGJ2. However, 5 µM 15-dPGJ2 inhibited the viability and caused apoptosis of both cell types. Exposure of cultured human hair follicles to 15-dPGJ2 resulted in significant apoptosis in follicular keratinocytes. Altogether, our data provide an evidence that 15-dPGJ2 promotes apoptosis in follicular keratinocytes and provide rationale for developing remedies for the prevention and treatment of hair loss based on DP2 antagonism.


Subject(s)
Apoptosis/drug effects , Hair Follicle/drug effects , Keratinocytes/drug effects , Prostaglandin D2/analogs & derivatives , Alopecia/metabolism , Alopecia/pathology , Alopecia/physiopathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Hair/growth & development , Hair/metabolism , Hair Follicle/pathology , Humans , Keratinocytes/pathology , Prostaglandin D2/toxicity , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism
6.
J Neurochem ; 133(3): 343-51, 2015 May.
Article in English | MEDLINE | ID: mdl-25662180

ABSTRACT

Obesity is a worldwide health problem that urgently needs to be solved. Leptin is an anti-obesity hormone that activates satiety signals to the brain. Evidence to suggest that leptin resistance is involved in the development of obesity is increasing; however, the molecular mechanisms involved remain unclear. We herein demonstrated that 15-deoxy-Δ(12,14) -prostaglandin J2 (15d-PGJ2 ) was involved in the development of leptin resistance. A treatment with 15d-PGJ2 inhibited the leptin-induced activation of signal transducer and activator of transcription 3 (STAT3) in neuronal cells (SH-SY5Y-Ob-Rb cells). Furthermore, the intracerebroventricular administration of 15d-PGJ2 reversed the inhibitory effects of leptin on food intake in rats. The peroxisome proliferator-activated receptor gamma (PPAR-γ) antagonist, GW9662, slightly reversed the inhibitory effects of 15d-PGJ2 on the leptin-induced activation of STAT3 in neuronal cells. The PPAR-γ agonist, rosiglitazone, also inhibited leptin-induced STAT3 phosphorylation. Therefore, the inhibitory effects of 15d-PGJ2 may be mediated through PPAR-γ. On the other hand, 15d-PGJ2 -induced leptin resistance may not be mediated by endoplasmic reticulum stress or suppressor of cytokine signaling 3. The results of the present study suggest that 15d-PGJ2 is a novel factor for the development of leptin resistance in obesity. Leptin resistance, an insensitivity to the actions of leptin, is involved in the development of obesity. Here, we found 15-deoxy-Δ(12,14) -prostaglandin J2 (15d-PGJ2 ) may be involved in the development of leptin resistance. The present results suggest that the 15d-PGJ2 may be a novel factor for the development of leptin resistance in obesity. 15d-PGJ2 , 15-Deoxy-Δ(12,14) -prostaglandin J2; STAT3, signal tranducer and activator of transcription 3.


Subject(s)
Eating/drug effects , Eating/physiology , Leptin/administration & dosage , Prostaglandin D2/analogs & derivatives , Animals , Cell Line, Tumor , Humans , Injections, Intraventricular , Male , Obesity/chemically induced , Obesity/metabolism , Prostaglandin D2/administration & dosage , Prostaglandin D2/toxicity , Rats , Rats, Wistar , Receptors, Leptin/agonists , Receptors, Leptin/metabolism
7.
Biochim Biophys Acta ; 1842(9): 1707-19, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24970746

ABSTRACT

Neuroinflammation is a major risk factor in Parkinson's disease (PD). Alternative approaches are needed to treat inflammation, as anti-inflammatory drugs such as NSAIDs that inhibit cyclooxygenase-2 (COX-2) can produce devastating side effects, including heart attack and stroke. New therapeutic strategies that target factors downstream of COX-2, such as prostaglandin J2 (PGJ2), hold tremendous promise because they will not alter the homeostatic balance offered by COX-2 derived prostanoids. In the current studies, we report that repeated microinfusion of PGJ2 into the substantia nigra of non-transgenic mice, induces three stages of pathology that mimic the slow-onset cellular and behavioral pathology of PD: mild (one injection) when only motor deficits are detectable, intermediate (two injections) when neuronal and motor deficits as well as microglia activation are detectable, and severe (four injections) when dopaminergic neuronal loss is massive accompanied by microglia activation and motor deficits. Microglia activation was evaluated in vivo by positron emission tomography (PET) with [(11)C](R)PK11195 to provide a regional estimation of brain inflammation. PACAP27 reduced dopaminergic neuronal loss and motor deficits induced by PGJ2, without preventing microglia activation. The latter could be problematic in that persistent microglia activation can exert long-term deleterious effects on neurons and behavior. In conclusion, this PGJ2-induced mouse model that mimics in part chronic inflammation, exhibits slow-onset PD-like pathology and is optimal for testing diagnostic tools such as PET, as well as therapies designed to target the integrated signaling across neurons and microglia, to fully benefit patients with PD.


Subject(s)
Encephalitis/prevention & control , Microglia/drug effects , Motor Skills Disorders/prevention & control , Neurons/metabolism , Parkinson Disease , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Prostaglandin D2/analogs & derivatives , Animals , Antineoplastic Agents/toxicity , Behavior, Animal/drug effects , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/metabolism , Encephalitis/pathology , Immunoenzyme Techniques , Male , Mice , Microglia/metabolism , Microglia/pathology , Motor Skills Disorders/chemically induced , Motor Skills Disorders/metabolism , Neurons/drug effects , Neurons/pathology , Positron-Emission Tomography , Prostaglandin D2/toxicity
8.
Int J Med Sci ; 9(7): 555-66, 2012.
Article in English | MEDLINE | ID: mdl-22991494

ABSTRACT

INTRODUCTION: Agonists of peroxisome proliferator-activated receptor gamma (PPARγ) have been examined as chemopreventive and chemotherapeutic agents. The aim was to investigate the cytotoxicity and action mechanisms of 15-deoxy-Δ(12,14)-prostaglandin J(2) (15d-PGJ(2)), one of endogenous ligands for PPARγ, in terms of PPARγ-dependency and the mitogen-activated protein kinase (MAPK) and Akt pathway in three human renal cell carcinoma (RCC)-derived cell lines. METHODS: 786-O, Caki-2 and ACHN cells were used as human RCC-derived cell lines. Cell viability and caspase-3 activity was detected by fluorescent reagents, and chromatin-condensation was observed with a brightfield fluorescent microscope after staining cells with Hoechst33342. The expression levels of proteins were detected by Western blot analysis. RESULTS: 15d-PGJ(2) showed cytotoxicity in dose-dependent manner. 15d-PGJ(2) induced chromatin-condensation and elevated caspase-3 activity, and the cell viability was restored by co-treatment with a pan-caspase inhibitor, Z-VAD-FMK, indicating the involvement of caspase-dependent apoptosis. The cytotoxicity was not impaired by a PPARγ inhibitor, GW9662, suggesting that 15d-PGJ(2) exerted the cytotoxicity in a PPARγ-independent manner. Some antioxidants rescued cells from cell death induced by 15d-PGJ(2), but some did not, suggesting that reactive oxygen species (ROS) did not contribute to the apoptosis. 15d-PGJ(2) also increased the expression levels of phospho-c-Jun N terminal kinase (JNK) in Caki-2 cells, and decreased those of phospho-Akt in 786-O cells, indicating that the JNK MAPK and the Akt pathways participated in the anticancer effects of 15d-PGJ(2) in some cell lines. CONCLUSION: 15d-PGJ(2) exerted cytotoxic effects accompanying caspase-dependent apoptosis, and this effect was elicited in a PPARγ-independent manner in three cell lines. In addition, the JNK MAPK and Akt pathway was involved in the cytotoxicity of 15d-PGJ(2) to some extent in some cell line. Therefore, our study showed the 15d-PGJ(2) to potentially be an interesting approach for RCC treatment.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , MAP Kinase Kinase 4/metabolism , PPAR gamma/physiology , Prostaglandin D2/analogs & derivatives , Proto-Oncogene Proteins c-akt/metabolism , Antioxidants/pharmacology , Blotting, Western , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Fluorometry , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , L-Lactate Dehydrogenase/metabolism , Microscopy, Fluorescence , Prostaglandin D2/toxicity , Reactive Oxygen Species/metabolism
9.
Inflamm Allergy Drug Targets ; 11(1): 58-65, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22309084

ABSTRACT

Cyclopentenone prostaglandins play a modulatory role in inflammation, in part through their ability to covalently modify key proinflammatory proteins. Using mesangial cells as a cellular model of inflammation we have observed that 15-deoxy-Δ(12,14)-prostaglandin J(2) (15d-PGJ(2)) exerts a biphasic effect on cell activation by cytokines, with nanomolar concentrations eliciting an amplification of nitric oxide (NO) production and iNOS and COX-2 levels, and concentrations of 5 µM and higher inhibiting proinflammatory gene expression. An analog of 15d-PGJ(2) lacking the cyclopentenone structure (9,10-dihydro-15d-PGJ(2)) showed reduced ability to elicit both types of effects, suggesting that the electrophilic nature of 15d-PGJ(2) is important for its biphasic action. Interestingly, the switch from stimulatory to inhibitory actions occurred within a narrow concentration range and correlated with the ability of 15d-PGJ(2) to induce heme oxygenase 1 and γ-GCSm expression. These events are highly dependent on the triggering of the antioxidant response, which is considered as a sensor of thiol group modification. Indeed, the levels of the master regulator of the antioxidant response Nrf2 increased upon treatment with concentrations of 15d-PGJ(2) above 5 µM, an effect that could not be mimicked by 9,10-dihydro-15d-PGJ(2). Thus, an interplay of redox and electrophilic signalling mechanisms can be envisaged by which 15d-PGJ(2), as several other redox mediators, could contribute both to the onset and to the resolution of inflammation in a context or concentration-dependent manner.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Immunologic Factors/pharmacology , Mesangial Cells/drug effects , Mesangial Cells/immunology , Prostaglandin D2/analogs & derivatives , Animals , Antioxidants/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Immunologic Factors/toxicity , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Mesangial Cells/metabolism , Prostaglandin D2/pharmacology , Prostaglandin D2/toxicity , Rats
10.
Neurobiol Dis ; 41(2): 318-28, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20933087

ABSTRACT

Cyclopentenone prostaglandins (CyPGs), such as 15-deoxy-Δ(12,14) -prostaglandin J(2) (15d-PGJ(2)), are active prostaglandin metabolites exerting a variety of biological effects that may be important in the pathogenesis of neurological diseases. Ubiquitin-C-terminal hydrolase L1 (UCH-L1) is a brain specific deubiquitinating enzyme whose aberrant function has been linked to neurodegenerative disorders. We report that [15d-PGJ(2)] detected by quadrapole mass spectrometry (MS) increases in rat brain after temporary focal ischemia, and that treatment with 15d-PGJ(2) induces accumulation of ubiquitinated proteins and exacerbates cell death in normoxic and hypoxic primary neurons. 15d-PGJ(2) covalently modifies UCH-L1 and inhibits its hydrolase activity. Pharmacologic inhibition of UCH-L1 exacerbates hypoxic neuronal death while transduction with a TAT-UCH-L1 fusion protein protects neurons from hypoxia. These studies indicate that UCH-L1 function is important in hypoxic neuronal death and that excessive production of CyPGs after stroke may exacerbate ischemic injury by modification and inhibition of UCH-L1.


Subject(s)
Cell Hypoxia/physiology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Prostaglandin D2/analogs & derivatives , Ubiquitin Thiolesterase/metabolism , Animals , Cell Hypoxia/drug effects , Cells, Cultured , Disease Models, Animal , Hypoxia-Ischemia, Brain/enzymology , Nerve Degeneration/chemically induced , Prostaglandin D2/chemistry , Prostaglandin D2/physiology , Prostaglandin D2/toxicity , Rats , Rats, Sprague-Dawley , Transduction, Genetic/methods , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics
11.
J Neuroinflammation ; 6: 18, 2009 Jul 25.
Article in English | MEDLINE | ID: mdl-19630993

ABSTRACT

BACKGROUND: Chronic neuroinflammation is implicated in Parkinson's disease (PD). Inflammation involves the activation of microglia and astrocytes that release high levels of prostaglandins. There is a profound gap in our understanding of how cyclooxygenases and their prostaglandin products redirect cellular events to promote PD neurodegeneration. The major prostaglandin in the mammalian brain is prostaglandin D2, which readily undergoes spontaneous dehydration to generate the bioactive cyclopentenone prostaglandins of the J2 series. These J2 prostaglandins are highly reactive and neurotoxic products of inflammation shown in cellular models to impair the ubiquitin/proteasome pathway and cause the accumulation of ubiquitinated proteins. PD is a disorder that exhibits accumulation of ubiquitinated proteins in neuronal inclusions (Lewy bodies). The role of J2 prostaglandins in promoting PD neurodegeneration has not been investigated under in vivo conditions. METHODS: We addressed the neurodegenerative and behavioral effects of the administration of prostaglandin J2 (PGJ2) simultaneously into the substantia nigra/striatum of adult male FVB mice by subchronic microinjections. One group received unilateral injections of DMSO (vehicle, n = 6) and three groups received PGJ2 [3.4 microg or 6.7 microg (n = 6 per group) or 16.7 microg (n = 5)] per injection. Immunohistochemical and behavioral analyses were applied to assess the effects of the subchronic PGJ2 microinfusions. RESULTS: Immunohistochemical analysis demonstrated a PGJ2 dose-dependent significant and selective loss of dopaminergic neurons in the substantia nigra while the GABAergic neurons were spared. PGJ2 also triggered formation of aggregates immunoreactive for ubiquitin and alpha-synuclein in the spared dopaminergic neurons. Moreover, PGJ2 infusion caused a massive microglia and astrocyte activation that could initiate a deleterious cascade leading to self-sustained progressive neurodegeneration. The PGJ2-treated mice also exhibited locomotor and posture impairment. CONCLUSION: Our studies establish the first model of inflammation in which administration of an endogenous highly reactive product of inflammation, PGJ2, recapitulates key aspects of PD. Our novel PGJ2-induced PD model strongly supports the view that localized and chronic production of highly reactive and neurotoxic prostaglandins, such as PGJ2, in the CNS could be an integral component of inflammation triggered by insults evoked by physical, chemical or microbial stimuli and thus establishes a link between neuroinflammation and PD neurodegeneration.


Subject(s)
Encephalitis/chemically induced , Inflammation Mediators/toxicity , Parkinsonian Disorders/chemically induced , Prostaglandin D2/analogs & derivatives , Substantia Nigra/drug effects , Animals , Cell Death/drug effects , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Encephalitis/pathology , Encephalitis/physiopathology , Gliosis/chemically induced , Gliosis/pathology , Gliosis/physiopathology , Immunohistochemistry , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Male , Mice , Microinjections , Movement Disorders/etiology , Movement Disorders/pathology , Movement Disorders/physiopathology , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Prostaglandin D2/metabolism , Prostaglandin D2/toxicity , Substantia Nigra/pathology , Substantia Nigra/physiopathology , alpha-Synuclein/drug effects , alpha-Synuclein/metabolism
12.
J Neurochem ; 110(1): 328-42, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19457109

ABSTRACT

Neurofibrillary tangles (NFT) are a hallmark of Alzheimer's disease. The major neurofibrillary tangle component is tau that is truncated at Asp421 (Deltatau), hyperphosphorylated and aggregates into insoluble paired helical filaments. Alzheimer's disease brains also exhibit signs of inflammation manifested by activated astrocytes and microglia, which produce cytotoxic agents among them prostaglandins. We show that prostaglandin (PG) J2, an endogenous product of inflammation, induces caspase-mediated cleavage of tau, generating Deltatau, an aggregation prone form known to seed tau aggregation prior to neurofibrillary tangle formation. The initial event observed upon PGJ2-treatment of human neuroblastoma SK-N-SH cells was the build-up of ubiquitinated (Ub) proteins indicating an early disruption of the ubiquitin-proteasome pathway. Apoptosis kicked in later, manifested by caspase activation and caspase-mediated cleavage of tau at Asp421 and poly (ADP-ribose) polymerase. Furthermore, cathepsin inhibition stabilized Deltatau suggesting its lysosomal clearance. Upon PGJ2-treatment tau accumulated in a large perinuclear aggregate. In rat E18 cortical neuronal cultures PGJ2-treatment also generated Deltatau detected in dystrophic neurites. Levels of Deltatau were diminished by caspase 3 knockdown using siRNA. PGD2, the precursor of PGJ2, produced some Deltatau. PGE2 generated none. Our data suggest a potential sequence of events triggered by the neurotoxic product of inflammation PGJ2 leading to tau pathology. The accumulation of Ub proteins is an early response. If cells fail to overcome the toxic effects induced by PGJ2, including accumulation of Ub proteins, apoptosis kicks in triggering caspase activation and tau cleavage, the clearance of which by cathepsins could be compromised culminating in tau pathology. Our studies are the first to provide a mechanistic link between inflammation and tau pathology.


Subject(s)
Caspases/metabolism , Cathepsins/metabolism , Neurofibrillary Tangles/metabolism , Prostaglandin D2/analogs & derivatives , Proteasome Endopeptidase Complex/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspases/drug effects , Cathepsins/drug effects , Cell Line, Tumor , Cells, Cultured , Encephalitis/metabolism , Encephalitis/physiopathology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Neurofibrillary Tangles/drug effects , Prostaglandin D2/metabolism , Prostaglandin D2/toxicity , Proteasome Endopeptidase Complex/drug effects , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Tauopathies/metabolism , Tauopathies/physiopathology , Ubiquitination/drug effects , Ubiquitination/physiology , tau Proteins/drug effects
13.
Toxicology ; 248(2-3): 121-9, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18450357

ABSTRACT

The cyclopentenone prostaglandin 15-deoxy-delta 12,14-prostaglandin J2 (15d-PGJ2) induces apoptosis in various cell types. However, the underlying mechanism of 15d-PGJ2-induced apoptosis is not fully understood. The present study was undertaken to determine the molecular mechanism by which 15d-PGJ2 induces apoptosis in MC3T3-E1 mouse osteoblastic cells. 15d-PGJ2 caused a concentration- and time-dependent apoptotic cell death. 15d-PGJ2 induced a transient activation of ERK1/2 and sustained activation of JNK. 15d-PGJ2-induced cell death was prevented by the JNK inhibitor SP6001, but not by inhibitors of ERK1/2 and p38. JNK activation by 15d-PGJ2 was blocked by antioxidants N-acetylcysteine (NAC) and GSH. 15d-PGJ2 caused ROS generation and 15d-PGJ2-induced cell death was prevented by antioxidants, suggesting involvement of ROS generation in 15d-PGJ2-induced cell death. 15d-PGJ2 triggered the mitochondrial apoptotic pathway indicated by enhanced Bax expression, loss of mitochondrial membrane potential, cytochrome c release, and caspase-3 activation. The JNK inhibitor blocked these events induced by 15d-PGJ2. Taken together, these results suggest that the 15d-PGJ2 induces cell death through the mitochondrial apoptotic pathway dependent of ROS and JNK activation in osteoblastic cells.


Subject(s)
Apoptosis/drug effects , Immunologic Factors/toxicity , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/drug effects , Osteoblasts/drug effects , Prostaglandin D2/analogs & derivatives , Animals , Cell Line , Cell Survival/drug effects , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , PPAR gamma/metabolism , Prostaglandin D2/toxicity , Reactive Oxygen Species/metabolism , Signal Transduction
14.
Chem Res Toxicol ; 21(1): 138-44, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18052108

ABSTRACT

Electrophilic molecules are endogenously generated and are causally involved in many pathophysiological effects. Prostaglandin D (20 (PGD (2)), a major cyclooxygenase product in a variety of tissues, readily undergoes dehydration to yield the cyclopentenone-type PGs of the J (2)-series such as 15-deoxy-Delta (12,14)-PGJ (2) (15d-PGJ (2)). 15d-PGJ (2) is an electrophile, which can covalently react via the Michael addition reaction with nucleophiles, such as the free sulfhydryls of glutathione and cysteine residues in cellular proteins that play an important role in the control of the redox cell-signaling pathways. Covalent binding of 15d-PGJ (2) to cellular proteins may be one of the mechanisms by which 15d-PGJ (2) induces a cellular response involved in most of the pathophysiological effects associated with inflammation. In the present perspective, we provide a comprehensive summary of 15d-PGJ (2) as an electrophilic mediator of cellular responses.


Subject(s)
Immunologic Factors/toxicity , Prostaglandin D2/analogs & derivatives , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cell Differentiation/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Humans , Immunologic Factors/biosynthesis , Immunologic Factors/chemistry , Immunologic Factors/metabolism , Inflammation/metabolism , Inflammation/pathology , Neurons/drug effects , Oxidation-Reduction , Prostaglandin D2/biosynthesis , Prostaglandin D2/chemistry , Prostaglandin D2/metabolism , Prostaglandin D2/toxicity
15.
J Neuroinflammation ; 4: 18, 2007 Jul 16.
Article in English | MEDLINE | ID: mdl-17634127

ABSTRACT

BACKGROUND: Prostaglandin (PG) production is associated with inflammation, a major feature in multiple sclerosis (MS) that is characterized by the loss of myelinating oligodendrocytes in the CNS. While PGs have been shown to have relevance in MS, it has not been determined whether PGs have a direct effect on cells within the oligodendrocyte lineage. METHODS: Undifferentiated or differentiated mouse oligodendrocyte precursor (mOP) cells were treated with PGE2, PGF2alpha, PGD2 or 15-deoxy-Delta12,14-PGJ2 (15d-PGJ2). Cell growth and survival following treatment were examined using cytotoxicity assays and apoptosis criteria. The membrane receptors for PGD2 and the nuclear receptor peroxisome proliferator-activated receptor (PPAR)gamma, as well as reactive oxygen species (ROS) in the death mechanism were examined. RESULTS: PGE2 and PGF2alpha had minimal effects on the growth and survival of mOP cells. In contrast, PGD2 and 15d-PGJ2 induced apoptosis of undifferentiated mOP cells at relatively low micromolar concentrations. 15d-PGJ2 was less toxic to differentiated mOP cells. Apoptosis was independent of membrane receptors for PGD2 and the nuclear receptor PPARgamma. The cytotoxicity of 15d-PGJ2 was associated with the production of ROS and was inversely related to intracellular glutathione (GSH) levels. However, the cytotoxicity of 15d-PGJ2 was not decreased by the free radical scavengers ascorbic acid or alpha-tocopherol. CONCLUSION: Taken together, these results demonstrated that 15d-PGJ2 is toxic to early stage OP cells, suggesting that 15d-PGJ2 may represent a deleterious factor in the natural remyelination process in MS.


Subject(s)
Apoptosis/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Prostaglandin D2/analogs & derivatives , Stem Cells/cytology , Stem Cells/physiology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Mice , Oligodendroglia/drug effects , Prostaglandin D2/physiology , Prostaglandin D2/toxicity , Rats , Stem Cells/drug effects
16.
Mol Cell Neurosci ; 29(2): 222-31, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15911346

ABSTRACT

The mechanisms implicated in the aggregation of ubiquitinated proteins detected in neurodegenerative disorders remain elusive. We report that prostaglandin J2 (PGJ2), an endogenous product of inflammation, up-regulates sequestosome 1/p62 in a time- and dose-dependent manner in human neuroblastoma SK-N-SH cells. We previously demonstrated that prostaglandins of the J2 series inhibit ubiquitin hydrolases, such as UCH-L1. Herein, we show that sequestosome 1/p62 is co-localized with ubiquitinated proteins and the ubiquitin hydrolase UCH-L1 in cytoplasmic aggregates induced by PGJ2. Preventing sequestosome 1/p62 up-regulation by RNA interference abolishes the aggregation but not the accumulation of ubiquitinated proteins or PGJ2 cytotoxicity. Sequestosome 1/p62 is known to bind poly-ubiquitinated proteins through its ubiquitin-associated domain. Our data support the notion that sequestosome 1/p62 up-regulation under stress conditions contributes to the "sequestration" of poly-ubiquitinated proteins into aggregates. However, the overwhelming accumulation of ubiquitinated proteins, rather than their aggregation, is likely to be an important contributor to PGJ2 cytotoxicity.


Subject(s)
Nerve Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Prostaglandin D2/analogs & derivatives , Proteins/metabolism , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing , Cell Death/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Encephalitis/metabolism , Encephalitis/physiopathology , Humans , Inclusion Bodies/metabolism , Nerve Degeneration/physiopathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/drug effects , Prostaglandin D2/antagonists & inhibitors , Prostaglandin D2/toxicity , Protein Binding/drug effects , Protein Binding/physiology , Proteins/drug effects , Proteins/genetics , RNA Interference , Sequestosome-1 Protein , Stress, Physiological/metabolism , Stress, Physiological/physiopathology , Time Factors , Ubiquitin Thiolesterase/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
17.
J Neurosci Res ; 78(6): 824-36, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15523637

ABSTRACT

The role of the proinflammatory and inducible form of cyclooxygenases (COX-2) in neurodegeneration is not well defined. Some of its metabolic products, such as prostaglandins (PG) of the J2 series, are known to be neurotoxic. Here we demonstrate that PGJ2 enhances COX-2 gene expression without elevating COX-1 levels in neuronal cells. PGJ2 also increased PGE2 production, establishing that the de novo synthesized COX-2 is enzymatically active. PGJ2 derivatives, such as 15d-PGJ2, are known activators of PPARgamma, a nuclear receptor that activates gene expression. However, the selective PPARgamma agonist ciglitazone failed to up-regulate COX-2, indicating that the PGJ2 effect on COX-2 is PPARgamma independent. Furthermore, PGJ2 stabilized IkappaBalpha levels, indicating that NFkappaB is not active under these conditions. The blocking of neuronal NFkappaB activity by PGJ2 may be an important contributor to its neurotoxicity, insofar as NFkappaB transactivation seems to be required for neuronal survival in the CNS. Interleukin-1 (IL1) is a proinflammatory cytokine known to stimulate the expression of genes associated with inflammation, including COX-2. Notably, IL1 mRNA levels in the neuronal cells were increased by PGJ2 treatment. The proinflammatory cytokine may mediate COX-2 up-regulation by PGJ2 through p38MAPK and not JNK activation, in that only an inhibitor of the former prevented the COX-2 increase. Thiol-reducing agents, such as N-acetylcysteine, protected the neuronal cells from the deleterious effects of PGJ2, whereas ascorbic acid did not. Collectively, our findings suggest that proinflammatory conditions that lead to COX-2 up-regulation and the concomitant production of PGJ2 initiate a mechanism of self-destruction through an autotoxic loop between PGJ2 and COX-2 that may exacerbate neurodegeneration beyond a point of no return. Thiol-reducing antioxidants may offer an optimal strategy for halting this neurodegenerative process.


Subject(s)
Neurons/enzymology , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/toxicity , Prostaglandin-Endoperoxide Synthases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Base Sequence , Cell Line , Cyclooxygenase 2 , DNA Primers , Gene Expression Regulation, Enzymologic/drug effects , Hippocampus/enzymology , Mice , Neurons/drug effects , Neurotoxins , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem J ; 382(Pt 1): 83-91, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15171682

ABSTRACT

PPARgamma (peroxisome proliferator-activated receptor gamma) is a ligand-activated transcription factor that responds to 15dPGJ2 (15-deoxy-Delta12,14-prostglandin J2). 15dPGJ2, in vitro, halts neuroblastoma cell growth, but reported mechanisms vary. Here we evaluated the modulatory effects of endogenous serum lipid mitogens upon the extent of 15dPGJ2-induced growth inhibition and on the precise cellular responses of neuroblastoma cells to PPARgamma activation. We show that 15dPGJ2 specifically inhibited cell growth in both complete and delipidated media. 15dPGJ2-induced growth inhibition was accompanied by decreased cell viability, although the effect was far more marked in delipidated medium than in complete medium. Incubation with 15dPGJ2 in complete medium resulted in cytoplasmic changes characteristic of type II programmed cell death (autophagy), while prior serum lipid removal resulted in cell death via an apoptotic mechanism. These distinct, serum lipid-dependent cellular responses to 15dPGJ2 were accompanied by increases in the expression of a reporter gene construct containing a PPAR response element of 2.3-fold in complete medium, but of 4.8-fold in delipidated medium. Restoration of the serum lysolipid LPA (lysophosphatidic acid) to cells in delipidated medium reduced 15dPGJ2-mediated PPARgamma activation, growth inhibition and cell death; following addition of S1P (sphingosine 1-phosphate), decreases were apparent but more marginal. Further, while the effects of LPA in delipidated medium were mediated through a G(i)/phosphoinositide 3-kinase/MAPK (mitogen-activated protein kinase) pathway, those of S1P did not involve the MAPK component. These data suggest that the serum lysolipid LPA modulates the degree of PPARgamma activation and the precise cellular response to 15dPGJ2 via activation of a G(i)/phosphoinositide 3-kinase/MAPK pathway.


Subject(s)
Lysophospholipids/pharmacology , Neuroblastoma/metabolism , PPAR gamma/physiology , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/antagonists & inhibitors , Prostaglandin D2/toxicity , Cell Death/drug effects , Cell Death/physiology , Cell Growth Processes/drug effects , Cell Line, Tumor , Humans , Ligands , Lipid Metabolism , Lipids/blood , Neuroblastoma/pathology , PPAR gamma/metabolism
19.
Biochemistry ; 43(8): 2345-52, 2004 Mar 02.
Article in English | MEDLINE | ID: mdl-14979731

ABSTRACT

15-Deoxy-Delta(12,14)prostaglandin J(2) (15-d-PGJ(2)), a terminal metabolite of the J-series cyclopentenone prostaglandins, influences a variety of cellular processes including gene expression, differentiation, growth, and apoptosis. As a ligand of peroxisomal proliferator-activated receptor gamma (PPAR gamma), 15-d-PGJ(2) can transactivate PPAR gamma-responsive promoters. Previously, we showed that multidrug resistance proteins MRP1 and MRP3 attenuate cytotoxic and transactivating activities of 15-d-PGJ(2) in MCF7 breast cancer cells. Attenuation was glutathione-dependent and was associated with formation of the glutathione conjugate of 15-d-PGJ(2), 15-d-PGJ(2)-SG, and its active efflux by MRP. Here we have investigated whether the glutathione S-transferases (GST) can influence biological activities of 15-d-PGJ(2). MCF7 cells were stably transduced with human cytosolic GST isozymes M1a, A1, or P1a. These GSTs had no effect on 15-d-PGJ(2) cytotoxicity when expressed either alone or in combination with MRP1. However, expression of any of the three GSTs significantly inhibited 15-d-PGJ(2)-dependent transactivation of a PPAR gamma-responsive reporter gene. The degree of inhibition correlated with the level of GST expressed. Under physiologic conditions, the nonenzymatic rate of 15-d-PGJ(2) conjugation with glutathione was significant. Of the three GST isozymes, only GSTM1a-1a further stimulated the rate of 15-d-PGJ(2)-SG formation. Moreover, GSTM1a-1a rate enhancement was only a transient burst that was complete within 15 s. Hence, catalysis plays little, if any, role in GST inhibition of 15-d-PGJ(2)-dependent transactivation. In contrast, inhibition of transactivation was associated with strong GST/15-d-PGJ(2) interactions. Potent inhibition by 15-d-PGJ(2) and 15-d-PGJ(2)-SG of GST activity was observed with K(i) in the 0.15-2.0 microM range for the three GST isozymes, results suggesting avid associations between GST and 15-d-PGJ(2) or 15-d-PGJ(2)-SG. Electrospray ionization mass spectrometry (ESI/MS) studies revealed no stable adducts of GST and 15-d-PGJ(2) indicating that GST/15-d-PGJ(2) interactions are primarily noncovalent. These results are consistent with a mechanism of GST-mediated inhibition of transactivation in which GST binds 15-d-PGJ(2) and 15-d-PGJ(2)-SG thereby sequestering the ligands in the cytosol away from their nuclear target, PPAR gamma.


Subject(s)
Glutathione Transferase/chemistry , Prostaglandin D2/antagonists & inhibitors , Prostaglandin D2/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/chemistry , Transcription Factors/metabolism , Transcriptional Activation , Catalysis , Cell Death/drug effects , Cell Line, Tumor , Genetic Vectors , Glutathione/chemistry , Glutathione/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Growth Inhibitors/antagonists & inhibitors , Growth Inhibitors/chemistry , Growth Inhibitors/metabolism , Growth Inhibitors/toxicity , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Prostaglandin D2/toxicity , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/chemistry , Trans-Activators/metabolism , Trans-Activators/toxicity , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Transduction, Genetic
20.
Toxicol Sci ; 77(2): 290-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14657518

ABSTRACT

Allyl alcohol causes hepatotoxicity that is potentiated by small doses of bacterial lipopolysaccharide (LPS) through a cyclooxygenase-2 (COX-2)-dependent mechanism. The COX-2 product prostaglandin D(2) (PGD(2)) increases hepatocyte killing by allyl alcohol in vitro. In the present study the ability of the nonenzymatic product of PGD(2), 15-deoxy-Delta12,14-prostaglandin J(2) (15d-PGJ(2)), to increase the cytotoxicity of allyl alcohol was evaluated. In a concentration-dependent manner, 15d-PGJ(2) significantly augmented cell death caused by allyl alcohol in isolated rat hepatocytes. 15d-PGJ(2) also increased the cytotoxicity of acrolein, the active metabolite of allyl alcohol. An agonist for the PGD(2) receptor neither reproduced the increase in allyl alcohol-mediated cytotoxicity nor altered the response to 15d-PGJ(2). Similarly, these responses were not affected by either an agonist or an antagonist for the peroxisome proliferator-activated receptor-gamma. The enhancement by 15d-PGJ(2) of allyl alcohol-mediated cell killing was unaffected by augmentation or inhibition of cAMP. Protein synthesis was markedly decreased by 15d-PGJ(2), but inhibition of protein synthesis alone with cycloheximide did not increase allyl alcohol-mediated cell killing. Allyl alcohol at subtoxic concentrations increased translocation of nuclear factor kappa B (NF-kappaB), whereas at cytotoxic concentrations no translocation occurred. 15d-PGJ(2) inhibited translocation of NF-kappaB from the cytosol to the nucleus both in the presence and absence of allyl alcohol. Like 15d-PGJ(2), MG132, an inhibitor of NF-kappaB activation, enhanced allyl alcohol-induced hepatocyte death. Together these results indicate that 15d-PGJ(2) augments hepatocyte killing by allyl alcohol, and the mechanism may be related to the inhibition of NF-kappaB activation.


Subject(s)
Hepatocytes/drug effects , Propanols/toxicity , Prostaglandin D2/toxicity , Acrolein/toxicity , Alanine Transaminase/analysis , Animals , Drug Synergism , Electrophoresis, Polyacrylamide Gel , Electrophoretic Mobility Shift Assay , Hepatocytes/metabolism , Male , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Prostaglandin D2/analogs & derivatives , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...