Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Eat Weight Disord ; 26(8): 2453-2461, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33426629

ABSTRACT

BACKGROUND/AIMS: Whey proteins (WP), obtained from milk after casein precipitation, represent a heterogeneous group of proteins. WP are reported to inhibit food intake in diet-induced experimental obesity; WP have been proposed as adjuvant therapy in oxidative stress-correlated pathologies. This work evaluates the effects of WP in comparison with casein, as a source of alimentary proteins, on food intake, weight growth and some indexes of oxidative equilibrium in Zucker Rats, genetically prone to obesity. METHODS: We monitored food intake and weight of Zucker Rats during the experiment, and some markers of oxidative equilibrium. RESULTS: WP induced significant decrease of food intake in comparison to casein (WP 80.41 ± 1.069 ml/day; CAS: 88.95 ± 1.084 ml/day; p < 0.0005). Body weight growth was slightly reduced, and the difference was just significant (WP 128.2 ± 6.56 g/day; CAS 145.2 ± 3.29 g/day; p = 0.049), while plasma HNE level was significantly lower in WP than in CAS (WP 41.2 ± 6.3 vs CAS 69.61 ± 4.69 pmol/ml, p = 0.007). Mild amelioration of oxidative equilibrium was indicated by a slight increase of total glutathione both in the liver and in the blood and a significant decrease of plasma 4-hydroxynonenal in the group receiving WP. CONCLUSIONS: The effect of WP on food intake and weight growth in Zucker Rats is particularly noteworthy since the nature of their predisposition to obesity is genetic; the possible parallel amelioration of the oxidative balance may constitute a further advantage of WP since oxidative stress is believed to be interwoven to obesity, metabolic syndrome and their complications.


Subject(s)
Obesity , Oxidative Stress , Animals , Eating , Humans , Obesity/drug therapy , Rats , Rats, Zucker , Whey Proteins/pharmacology
2.
Oxid Med Cell Longev ; 2016: 9348651, 2016.
Article in English | MEDLINE | ID: mdl-27313835

ABSTRACT

RAGE is a multiligand receptor able to bind advanced glycation end-products (AGEs), amphoterin, calgranulins, and amyloid-beta peptides, identified in many tissues and cells, including neurons. RAGE stimulation induces the generation of reactive oxygen species (ROS) mainly through the activity of NADPH oxidases. In neuronal cells, RAGE-induced ROS generation is able to favor cell survival and differentiation or to induce death through the imbalance of redox state. The dual nature of RAGE signaling in neurons depends not only on the intensity of RAGE activation but also on the ability of RAGE-bearing cells to adapt to ROS generation. In this review we highlight these aspects of RAGE signaling regulation in neuronal cells.


Subject(s)
Neurons/metabolism , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products/metabolism , Animals , Cell Differentiation , Glycation End Products, Advanced/metabolism , Humans , NADPH Oxidases/metabolism , Neurons/cytology , Receptor for Advanced Glycation End Products/chemistry , Signal Transduction
3.
PLoS One ; 11(3): e0152465, 2016.
Article in English | MEDLINE | ID: mdl-27023064

ABSTRACT

The activation of Nrf2 has been demonstrated to play a crucial role in cancer cell resistance to different anticancer therapies. The inhibition of proteasome activity has been proposed as a chemosensitizing therapy but the activation of Nrf2 could reduce its efficacy. Using the highly chemoresistant neuroblastoma cells HTLA-230, here we show that the strong reduction in proteasome activity, obtained by using low concentration of bortezomib (BTZ, 2.5 nM), fails in reducing cell viability. BTZ treatment favours the binding of Nrf2 to the ARE sequences in the promoter regions of target genes such as heme oxygenase 1 (HO-1), the modulatory subunit of γ-glutamylcysteine ligase (GCLM) and the transporter for cysteine (x-CT), enabling their transcription. GSH level is also increased after BTZ treatment. The up-regulation of Nrf2 target genes is responsible for cell resistance since HO-1 silencing and GSH depletion synergistically decrease BTZ-treated cell viability. Moreover, cell exposure to all-trans-Retinoic acid (ATRA, 3 µM) reduces the binding of Nrf2 to the ARE sequences, decreases HO-1 induction and lowers GSH level increasing the efficacy of bortezomib. These data suggest the role of Nrf2, HO-1 and GSH as molecular targets to improve the efficacy of low doses of bortezomib in the treatment of malignant neuroblastoma.


Subject(s)
Bortezomib/pharmacology , Drug Resistance, Neoplasm/drug effects , Glutathione/metabolism , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Neuroblastoma/metabolism , Amino Acid Transport System y+/metabolism , Antioxidant Response Elements/genetics , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/genetics , Humans , Neuroblastoma/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Up-Regulation/drug effects
4.
Oxid Med Cell Longev ; 2016: 1958174, 2016.
Article in English | MEDLINE | ID: mdl-26697129

ABSTRACT

The transcription factor, nuclear factor erythroid 2 p45-related factor 2 (Nrf2), acts as a sensor of oxidative or electrophilic stresses and plays a pivotal role in redox homeostasis. Oxidative or electrophilic agents cause a conformational change in the Nrf2 inhibitory protein Keap1 inducing the nuclear translocation of the transcription factor which, through its binding to the antioxidant/electrophilic response element (ARE/EpRE), regulates the expression of antioxidant and detoxifying genes such as heme oxygenase 1 (HO-1). Nrf2 and HO-1 are frequently upregulated in different types of tumours and correlate with tumour progression, aggressiveness, resistance to therapy, and poor prognosis. This review focuses on the Nrf2/HO-1 stress response mechanism as a promising target for anticancer treatment which is able to overcome resistance to therapies.


Subject(s)
Drug Resistance, Neoplasm , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction , Animals , Humans
5.
Cell Death Dis ; 4: e589, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23579276

ABSTRACT

Neuroblastoma (NB) is the second most common solid pediatric tumor and is characterized by clinical and biological heterogeneity, and stage-IV of the disease represents 50% of all cases. Considering the limited success of present chemotherapy treatment, it has become necessary to find new and effective therapies. In this context, our approach consists of identifying and targeting key molecular pathways associated with NB chemoresistance. This study has been carried out on three stage-IV NB cell lines with different status of MYCN amplification. Cells were exposed to a standard chemotherapy agent, namely etoposide, either alone or in combination with particular drugs, which target intracellular signaling pathways. Etoposide alone induced a concentration-dependent reduction of cell viability and, at very high doses, totally counteracted cell tumorigenicity and neurosphere formation. In addition, etoposide activated p38 mitogen-activated protein kinase (MAPK), AKT and c-Jun N-terminal kinase. Pre-treatment with SB203580, a p38MAPK inhibitor, dramatically sensibilized NB cells to etoposide, strongly reducing the dosage needed to inhibit tumorigenicity and neurosphere formation. Importantly, SB203580-etoposide cotreatment also reduced cell migration and invasion by affecting cyclooxygenase-2, intercellular adhesion molecule-1, C-X-C chemokine receptor-4 and matrix metalloprotease-9. Collectively, our results suggest that p38MAPK inhibition, in combination with standard chemotherapy, could represent an effective strategy to counteract NB resistance in stage-IV patients.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Imidazoles/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Apoptosis/genetics , Cell Differentiation , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Synergism , Humans , JNK Mitogen-Activated Protein Kinases/economics , JNK Mitogen-Activated Protein Kinases/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Staging , Nervous System Neoplasms/drug therapy , Neuroblastoma/drug therapy , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Biochem Biophys Res Commun ; 430(3): 957-62, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23261460

ABSTRACT

Molecular mechanisms relating interferon-alpha (IFN-alpha) to brain damage have recently been identified in a microarray analysis of cerebrospinal fluid lymphocytes from patients with Aicardi-Goutières Syndrome (AGS). These findings demonstrate that the inhibition of angiogenesis and the activation of neurotoxic lymphocytes are the major pathogenic mechanisms involved in the brain damage consequent to elevated interferon-alpha levels. Our previous study demonstrated that cathepsin D, a lysosomal aspartyl endopeptidase, is the primary mediator of the neurotoxicity exerted by AGS lymphocytes. Cathepsin D is a potent pro-apoptotic, neurotoxic, and demyelinating protease if it is not properly inhibited by the activities of leukocystatins. In central nervous system white matter, demyelination results from cathepsin over-expression when not balanced by the expression of its inhibitors. In the present study, we used RNA interference to inhibit cathepsin D expression in AGS lymphocytes with the aim of decreasing the neurotoxicity of these cells. Peripheral blood lymphocytes collected from an AGS patient were immortalized and co-cultured with astrocytes in the presence of interferon alpha with or without cathepsin D RNA interference probes. Cathepsin D expression was measured by qPCR, and neurotoxicity was evaluated by microscopy. RNA interference inhibited cathepsin D over-production by 2.6-fold (P<0.01) in AGS lymphocytes cultured in the presence of interferon alpha. AGS lymphocytes treated using RNA interference exhibited a decreased ability to induce neurotoxicity in astrocytes. Such neurotoxicity results in the inhibition of astrocyte growth and the inhibition of the ability of astrocytes to construct web-like aggregates. These results suggest a new strategy for repairing AGS lymphocytes in vitro by inhibiting their ability to induce astrocyte damage and leukodystrophy.


Subject(s)
Astrocytes/pathology , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/pathology , Cathepsin D/antagonists & inhibitors , Lymphocytes/immunology , Nerve Tissue Proteins/antagonists & inhibitors , Nervous System Malformations/immunology , Nervous System Malformations/pathology , Astrocytes/immunology , Cathepsin D/genetics , Cell Line, Tumor , Humans , Interferon-alpha/immunology , Nerve Tissue Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics
7.
Oncol Rep ; 27(5): 1689-94, 2012 May.
Article in English | MEDLINE | ID: mdl-22367235

ABSTRACT

T lymphocytes play a major role in counteracting cancer occurrence and development. Immune therapies against cancer are focused on eliciting a cytotoxic T cell response. This anticancer activity is related to a variety of mechanisms including the activation of cytokines and proapoptotic mediators. Interferon α is an established inhibitor of cancer cell growth. A clinical situation involving the coexistence of high interferon α levels and lymphocyte activation is the Aicardi-Goutières syndrome, a progressive encephalopathy arising usually during the first year of life characterized by intracranial basal ganglia calcifications, leukodystrophy and microcephaly. Aicardi-Goutières syndrome 1 mutation silences the TREX1 gene, a major endogenous nuclease. The in vitro study presented herein evaluates the efficacy of the TREX1 mutation in potentiating the anticancer properties of T cells. A TREX1-mutated lymphocyte cell line was derived from an Aicardi-Goutières syndrome patient and co-cultured with neuroblastoma cells and vascular endothelial cells in the presence of interferon α. TREX1-mutated lymphocytes exerted marked inhibitory action on neuroblastoma cell growth. Cathepsin D was recognized by qPCR as the main mediator produced by TREX1-mutated lymphocytes involved in the inhibition of neuroblastoma cell growth. These effects were enhanced in the presence of interferon α. Similar inhibitory effects in cell growth were exerted by TREX1-mutated lymphocytes towards vascular endothelial cell angiogenesis as evaluated on Matrigel. The results obtained provide evidence that mutations of the TREX1 gene increase the capability of T-lymphocytes to inhibit growth of neoplastic neuronal cells and related angiogenesis.


Subject(s)
Exodeoxyribonucleases/genetics , Lymphocytes/metabolism , Mutation , Neuroblastoma/genetics , Phosphoproteins/genetics , Cathepsin D/genetics , Cell Line , Cell Proliferation , Gene Expression/drug effects , Humans , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Lymphocytes/drug effects , Lymphocytes/immunology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neuroblastoma/immunology
8.
Br J Pharmacol ; 164(8): 2054-63, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21649644

ABSTRACT

BACKGROUND AND PURPOSE: Strategies designed to enhance cerebral cAMP have been proposed as symptomatic treatments to counteract cognitive deficits. However, pharmacological therapies aimed at reducing PDE4, the main class of cAMP catabolizing enzymes in the brain, produce severe emetic side effects. We have recently synthesized a 3-cyclopentyloxy-4-methoxybenzaldehyde derivative, structurally related to rolipram, and endowed with selective PDE4D inhibitory activity. The aim of the present study was to investigate the effect of the new drug, namely GEBR-7b, on memory performance, nausea, hippocampal cAMP and amyloid-ß (Aß) levels. EXPERIMENTAL APPROACH: To measure memory performance, we performed object recognition tests on rats and mice treated with GEBR-7b or rolipram. The emetic potential of the drug, again compared with rolipram, was evaluated in rats using the taste reactivity test and in mice using the xylazine/ketamine anaesthesia test. Extracellular hippocampal cAMP was evaluated by intracerebral microdialysis in freely moving rats. Levels of soluble Aß peptides were measured in hippocampal tissues and cultured N2a cells by elisa. KEY RESULTS: GEBR-7b increased hippocampal cAMP, did not influence Aß levels and improved spatial, as well as object memory performance in the object recognition tests. The effect of GEBR-7b on memory was 3 to 10 times more potent than that of rolipram, and its effective doses had no effect on surrogate measures of emesis in rodents. CONCLUSION AND IMPLICATIONS: Our results demonstrate that GEBR-7b enhances memory functions at doses that do not cause emesis-like behaviour in rodents, thus offering a promising pharmacological perspective for the treatment of memory impairment.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Imines/pharmacology , Memory/drug effects , Morpholines/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Animals , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Hippocampus/drug effects , Hippocampus/metabolism , Ketamine/administration & dosage , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Rats, Wistar , Xylazine/administration & dosage
9.
Cell Death Differ ; 17(6): 1059-68, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20057501

ABSTRACT

In liver ischemic preconditioning (IP), stimulation of adenosine A2a receptors (A2aR) prevents ischemia/reperfusion injury by promoting diacylglycerol-mediated activation of protein kinase C (PKC). By concerting diacylglycerol to phosphatidic acid, diacylglycerol kinases (DGKs) act as terminator of diacylglycerol signalling. This study investigates the role of DGK in the development of hepatocyte IP. DGK activity and cell viability were evaluated in isolated rat hepatocytes preconditioned by 10 min hypoxia followed by 10 min re-oxygenation or by the treatment with the A2aR agonist, CGS21680, and subsequently exposed to prolonged hypoxia. We observed that after IP or A2aR activation, a decrease in DGK activity was associated with the onset of hepatocyte tolerance to hypoxia. CGS21680-induced stimulation of A2aR specifically inhibited DGK isoform theta by activating RhoA-GTPase. Consistently, both siRNA-mediated downregulation of DGK theta and hepatocyte pretreatment with the DGK inhibitor R59949 induced cell tolerance to hypoxia. The pharmacological inhibition of DGK was associated with the diacylglycerol-dependent activation of PKC delta and epsilon and of their downstream target p38 MAPK. In conclusion, we unveil a novel signalling pathway contributing to the onset of hepatocyte preconditioning, which through RhoA-GTPase, couples A2aR to the downregulation of DGK. Such an inhibition is essential for the sustained accumulation of diacylglycerol required for triggering PKC-mediated survival signals.


Subject(s)
Adenosine/pharmacology , Diacylglycerol Kinase/metabolism , Hepatocytes/enzymology , Animals , Cell Death , Cell Hypoxia , Cells, Cultured , Diacylglycerol Kinase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Male , Piperidines/pharmacology , Quinazolinones/pharmacology , Rats , Rats, Wistar , Receptor, Adenosine A2A/metabolism , Receptors, Purinergic P1/metabolism , rhoA GTP-Binding Protein/metabolism
10.
Biochim Biophys Acta ; 1737(2-3): 83-93, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16311069

ABSTRACT

4-Hydroxynonenal, a significant aldehyde end product of membrane lipid peroxidation with numerous biochemical activities, has consistently been detected in various human diseases. Concentrations actually detectable in vivo (0.1-5 microM) have been shown to up-regulate different genes and modulate various enzyme activities. In connection with the latter aspect, we show here that, in isolated rat hepatocytes, 1 microM 4-hydroxynonenal selectively activates protein kinase C-delta, involved in apoptosis of many cell types; it also induces very early activation of Jun N-terminal kinase, in parallel increasing activator protein-1 DNA-binding activity in a time-dependent manner and triggering apoptosis after only 120 min treatment. These phenomena are likely protein kinase C-delta-dependent, being significantly reduced or annulled by cell co-treatment with rottlerin, a selective inhibitor of protein kinase C-delta. We suggest that 4-hydroxynonenal may induce apoptosis through activation of protein kinase C-delta and of Jun N-terminal kinase, and consequent up-regulation of activator protein-1 DNA binding.


Subject(s)
Aldehydes/pharmacology , Apoptosis/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Protein Kinase C-delta/metabolism , Aldehydes/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Enzyme Activation/drug effects , Glutathione/metabolism , Hepatocytes/metabolism , In Vitro Techniques , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Rats , Rats, Wistar , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism
11.
Biochem Biophys Res Commun ; 294(3): 547-52, 2002 Jun 14.
Article in English | MEDLINE | ID: mdl-12056801

ABSTRACT

4-Hydroxynonenal (HNE) in the concentration range detectable in many pathophysiologic conditions is able to modulate signal transduction cascades and gene expression. Here, we report the stimulating effect of 1 microM HNE on the release of the monocyte chemotactic protein-1 (MCP-1) by murine macrophages. MCP-1-increased export following 1-h cell treatment with HNE proved to be comparable to that exerted by standard amounts of bacterial lipopolysaccharide (LPS). However, the key molecular event in HNE-induced secretion of MCP-1 appeared to be the increased activity of beta-PKC isoforms, which are recognized as playing a role in the regulation of cell protein transport and secretion. On the other hand, in LPS-stimulated cells, the delta isoform was seen to be involved and was probably related to LPS-mediated effects on MCP-1 expression and synthesis. In conclusion, HNE might interact with other pro-inflammatory stimuli, like LPS, in a concerted amplification of MCP-1 production and secretion.


Subject(s)
Chemokine CCL2/metabolism , Isoenzymes/metabolism , Macrophages/metabolism , Protein Kinase C/metabolism , Aldehydes/pharmacology , Animals , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation/drug effects , Mice , Protein Kinase C beta , Signal Transduction/drug effects
12.
Hepatology ; 33(1): 131-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11124829

ABSTRACT

Ischemic preconditioning improves liver resistance to hypoxia and reduces reperfusion injury following transplantation. However, the intracellular signals that mediate the development of liver hypoxic preconditioning are largely unknown. We have investigated the signal pathway leading to preconditioning in freshly isolated rat hepatocytes. Hepatocytes were preconditioned by 10-minute incubation under hypoxic conditions followed by 10 minutes of reoxygenation and subsequently exposed to 90 minutes of hypoxia. Preconditioning reduced hepatocyte killing by hypoxia by about 35%. A similar protection was also obtained by preincubation with chloro-adenosine or with A(2A)-adenosine receptor agonist CGS21680, whereas A(1)-adenosine receptor agonist N-phenyl-isopropyladenosine (R-PIA) was inactive. Conversely, the development of preconditioning was blocked by A(2)-receptor antagonist 3,7-dimethyl-1-propargylxanthine (DMPX), but not by A(1)-receptor antagonist 8-cyclopenthyl-1, 3-dipropylxanthine (DPCPX). In either preconditioned or CGS21680-treated hepatocytes a selective activation of delta and epsilon protein kinase C (PKC) isoforms was also evident. Inhibition of heterotrimeric G(i) protein or of phospholypase C by, respectively, pertussis toxin or U73122, prevented PKC activation as well as the development of preconditioning. MEK inhibitor PD98509 did not interfere with preconditioning that was instead blocked by p38 MAP kinase inhibitor SB203580. The direct activation of p38 MAPK by anisomycin A mimicked the protection against hypoxic injury given by preconditioning. Consistently, an increased phosphorylation of p38 MAPK was observed in preconditioned or CGS21680-treated hepatocytes, and this effect was abolished by PKC-blocker, chelerythrine. We propose that a signal pathway involving A(2A)-adenosine receptors, G(i)-proteins, phospholypase C, delta- and epsilon-PKCs, and p38 MAPK, is responsible for the development of liver ischemic preconditioning.


Subject(s)
Hepatocytes/physiology , Ischemic Preconditioning , Signal Transduction/physiology , Animals , Isoenzymes/physiology , Male , Protein Kinase C/physiology , Protein Kinases/physiology , Rats , Rats, Wistar , Receptors, Purinergic P1/physiology
13.
Free Radic Biol Med ; 29(12): 1280-90, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11118818

ABSTRACT

Treatment of isolated rat hepatocytes with the glutathione depleting agents L-buthionine-S,R-sulfoximine or diethylmaleate reproduced various cellular conditions of glutathione depletion, from moderate to severe, similar to those occurring in a wide spectrum of human liver diseases. To evaluate molecular changes and possible cellular dysfunction and damage consequent to a pathophysiologic level of GSH depletion, the effects of this condition on protein kinase C (PKC) isoforms were investigated, since these are involved in the intracellular specific regulatory processes and are potentially sensitive to redox changes. Moreover, a moderate perturbation of cellular redox state was found to activate novel PKC isoforms, and a clear relationship was shown between novel kinase activation and nuclear binding of the redox-sensitive transcription factor, activator protein-1 (AP-1). Apoptotic death of a significant number of cells, confirmed in terms of internucleosomal DNA fragmentation was a possible effect of these molecular reactions, and was triggered by a condition of glutathione depletion usually detected in human liver diseases. Finally, the inhibition of novel PKC enzymatic activity in cells co-treated with rottlerin, a selective novel kinase inhibitor, prevented glutathione-dependent novel PKC up-regulation, markedly moderated AP-1 activation, and protected cells against apoptotic death. Taken together, these findings indicate the existence of an apoptotic pathway dependent on glutathione depletion, which occurs through the up-regulation of novel PKCs and AP-1.


Subject(s)
Apoptosis/physiology , Buthionine Sulfoximine/pharmacology , Cell Nucleus/metabolism , Glutathione/metabolism , Hepatocytes/cytology , Hepatocytes/physiology , Maleates/pharmacology , Protein Kinase C/metabolism , Transcription Factor AP-1/metabolism , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Hepatocytes/drug effects , Humans , Isoenzymes/metabolism , Kinetics , Liver/cytology , Male , Rats , Rats, Wistar
14.
Biochem Biophys Res Commun ; 268(2): 642-6, 2000 Feb 16.
Article in English | MEDLINE | ID: mdl-10679257

ABSTRACT

Amyloid beta-protein (Abeta) aggregation produces an oxidative stress in neuronal cells that, in turn, may induce an amyloidogenic shift of neuronal metabolism. To investigate this hypothesis, we analyzed intra- and extracellular Abeta content in NT2 differentiated cells incubated with 4-hydroxy-2,3-nonenal (HNE), a major product of lipid peroxidation. In parallel, we evaluated protein kinase C (PKC) isoenzymes activity, a signaling system suspected to modulate amyloid precursor protein (APP) processing. Low HNE concentrations (0.1-1 microM) induced a 2-6 fold increase of intracellular Abeta production that was concomitant with selective activation of betaI and betaII PKC isoforms, without affecting either cell viability or APP full-length expression. Selective activation of the same PKC isoforms was observed following NT2 differentiation. Our findings suggest that PKC beta isoenzymes are part of cellular mechanisms that regulate production of the intracellular Abeta pool. Moreover, they indicate that lipid peroxidation fosters intracellular Abeta accumulation, creating a vicious neurodegenerative loop.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Isoenzymes/metabolism , Oxidative Stress , Protein Kinase C/metabolism , Enzyme Activation , Humans , Neurons/enzymology , Neurons/metabolism , Protein Kinase C beta , Tumor Cells, Cultured
15.
Toxicology ; 143(3): 283-92, 2000 Mar 07.
Article in English | MEDLINE | ID: mdl-10755714

ABSTRACT

Previous investigations have demonstrated that 1,2-dichloroethane (DCE) poisoning affects dolichol (Dol) concentration in rat liver. Dol, a long-chain polyprenol, is considered an important membrane component: as dolichyl phosphate, it is rate limiting for the synthesis of glycoprotein; as free or fatty acid, it is highly concentrated in the Golgi apparatus (GA) where it can increase membrane fluidity and permeability, required glycoprotein maturation and secretion. DCE biotransformation may stimulate pro-oxidant events through hepatocellular glutathione depletion. Since the molecules of Dol are susceptible to oxidative degradation, the aim of this investigation is to verify whether vitamin E (vit. E) supplementation in rats is able to prevent Dol breakdown during acute DCE treatment. Before acute DCE administration (628 mg/kg body weight), a group of male Wistar rats were pretreated with vit. E (33 mg/kg body weight) for 3 days. High-performance liquid chromatography analysis has shown that within 5-60 min after DCE administration, the Dol concentration decreased in liver homogenate, cytosol, microsomes and GA. Particularly, 60 min after the treatment, Dol levels in the trans Golgi fraction were 71% lower than in controls. Rat pre-treatment with vit. E prevented the DCE-induced decrease in Dol concentrations of all liver fractions considered, in particular the reduction of total-Dol observed in the trans Golgi fraction 60 min after treatment was only 40%. These data suggest that hepatic metabolism of DCE is able to promote peroxidative attacks which lead to the degradation of Dol molecules. The pre-treatment of rats with vit. E results in a good, although not complete, prevention of total-Dol depletion after DCE poisoning.


Subject(s)
Dolichols/metabolism , Ethylene Dichlorides/poisoning , Vitamin E/pharmacology , Animals , Ascorbic Acid/pharmacology , Body Weight/drug effects , Chromatography, High Pressure Liquid , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Iron/pharmacology , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Male , Oxidants/poisoning , Rats , Rats, Wistar , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
17.
Hepatology ; 29(5): 1565-72, 1999 May.
Article in English | MEDLINE | ID: mdl-10216144

ABSTRACT

A major aldehydic end product of the peroxidation of arachidonic acid, 4-hydroxy-2,3-nonenal (HNE), has recently been considered for its potential involvement in a variety of cell functions. Here we report on the differential regulation of rat hepatocyte protein kinase C (PKC) isoforms by concentrations of HNE actually detectable in specific biological fluids or tissues. PKC betaI and, to a much greater extent, PKC betaII activities were markedly increased by 0.1 micromol/L HNE (final concentration in cell medium) whereas they were unaffected or even inhibited by 1 to 10 micromol/L HNE. On the contrary, the calcium independent PKC delta activity was inhibited by 0.1 micromol/L and increased by 1 and 10 micromol/L. Further, we show here that HNE-induced stimulation of PKC betaI and betaII activities, both in cytosolic and in membrane fractions, is paralleled by a marked stimulation of the anterograde transport of a lysosomal enzyme within the central vacuolar system. In fact, the treatment with 0.1 micromol/L HNE accelerated the PKC-dependent transport of lysosomal procathepsin D from the trans-Golgi network to the endosomal-lysosomal compartment and, in addition, increased the exocytosis of mature cathepsin D (CD) from these compartments. On the other hand, hepatocyte cotreatment with a selective inhibitor of classic PKCs prevented the aldehyde-induced activation of CD transport. These results support the possible involvement of HNE in the PKC-dependent regulation of the traffic of secretory glycoproteins, and point to remarkable implications of this aldehyde in the pathophysiology of various exocytic processes including hepatocyte lipoprotein secretion.


Subject(s)
Aldehydes/metabolism , Isoenzymes/metabolism , Lipid Peroxides/metabolism , Liver/enzymology , Protein Kinase C/metabolism , Aldehydes/pharmacology , Animals , Biological Transport/drug effects , Cathepsin D/metabolism , Cell Survival/drug effects , Glutathione/metabolism , Glycoproteins/metabolism , Liver/cytology , Liver/drug effects , Lysosomes/metabolism , Male , Phosphotransferases/drug effects , Precipitin Tests , Protein Kinase C beta , Rats , Rats, Wistar , Signal Transduction/physiology
18.
Free Radic Biol Med ; 25(4-5): 529-35, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9741589

ABSTRACT

Acute ethanol exposure of rat isolated hepatocytes leads to a significant decrease (-30%) in cytosolic enzymatic activity of classic protein kinase C (PKC) isoforms, while immunoreactive protein level measured by Western Blot remains unaffected. The inactivation of classic cytosolic isoforms appears dependent on the modification of the enzyme function, probably due to ethanol metabolism. In fact, pretreatment with 4-methylpyrazole (4MP), an inhibitor of alcohol dehydrogenase, fully prevented such damage. After ethanol treatment, a decrease of about 40% in both enzymatic activity and immunoreactive protein level of novel PKC isoforms was evident both in the soluble and particulate fractions. Even if 4MP cell pre-treatment afforded protection in this case too, the inhibitory action of ethanol on novel PKC hepatocyte isoforms involves a proteolytic mechanism as shown by Western Blot analysis. The reproduction of PKC inactivation by ethanol in hepatocyte lysate excluded a role of peroxisomal hydrogen peroxide in the pathogenesis of the damage investigated. This damage was not reduced by addition of catalase to the lysate model system.


Subject(s)
Ethanol/pharmacology , Isoenzymes/antagonists & inhibitors , Liver/enzymology , Protein Kinase C/antagonists & inhibitors , Animals , Antidotes/pharmacology , Blotting, Western , Cytosol/enzymology , Enzyme Activation , Ethanol/administration & dosage , Fomepizole , Glutathione/metabolism , Isoenzymes/analysis , Male , Malondialdehyde/metabolism , Protein Kinase C/analysis , Pyrazoles/pharmacology , Rats , Rats, Wistar
19.
Chem Biol Interact ; 114(1-2): 33-43, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9744554

ABSTRACT

Acute ethanol administration induces significant modifications both in secretive and formative membranes of rat liver Golgi apparatus. The decrease in glycolipoprotein secretion and their retention into the hepatocyte contribute to the pathogenesis of alcohol-induced fatty liver. Molecular and cellular mechanisms behind the ethanol-induced injury of the liver secretory pathway are not yet completely defined. In this study on intact livers from ethanol-treated rats, the involvement of the Golgi compartment in the impairment of hepatic glycolipoprotein secretion has been correlated with changes in the expression level, subcellular distribution and enzymatic activity of protein kinase C (PKC) isoforms. Acute ethanol exposure determined a translocation of classic PKCs and delta isoform from the cytosol to cis and trans Golgi membranes, the site of glycolipoprotein retention in the hepatic cell. A marked stimulation of cytosolic epsilon PKC activity was observed throughout the period of treatment. The presence of activated PKC isozymes at the Golgi compartment of alcohol-treated rat livers may play a role in hepatic secretion and protein accumulation. Direct and indirect effects of ethanol consumption on PKC isozymes and Golgi function are discussed.


Subject(s)
Ethanol/pharmacology , Golgi Apparatus/drug effects , Isoenzymes/biosynthesis , Liver/drug effects , Protein Kinase C/biosynthesis , Animals , Blotting, Western , Cytosol/drug effects , Cytosol/enzymology , Glycoproteins/metabolism , Golgi Apparatus/enzymology , Intracellular Membranes/drug effects , Lipoproteins/metabolism , Liver/enzymology , Liver/ultrastructure , Male , Protein Kinase C-delta , Protein Kinase C-epsilon , Rats , Rats, Wistar , Time Factors
20.
J Toxicol Environ Health A ; 54(2): 133-44, 1998 May 22.
Article in English | MEDLINE | ID: mdl-9652549

ABSTRACT

Dolichols are long-chain polyprenols containing 14-22 isoprene units, present in mammalian tissues as free dolichol (Free-Dol), fatty acyl dolichyl esters (Dol-FA), and dolichyl phosphate (Dol-P). The hepatic level of Dol-P seems to be a rate-limiting factor for glycosylation processes. Previous studies from our laboratory demonstrated the susceptibility of the dolichol molecule to undergo radical attacks. Since the toxicity of 1,1,2,2-tetrachloroethane (TTCE)is dependent on the free-radical production during hepatic biotrasformation, it was of interest to determine whether this haloalkane might affect glycosylation mechanisms by changing dolichol levels and distribution in rat liver microsomes and Golgi apparatus (GA). Male Sprague-Dawley rats received a single dose of TTCE (574 mg/kg body weight) and were then sacrificed at different times (5, 15, 30, or 60 min). In the TTCE-treated rats both serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) activities and hepatic triglycerides (TG) were significantly higher than control, while microsomal glucose 6-phosphatase (G6Pase) activity was decreased. In total microsomes Dol-P levels considered rate-limiting for the biosynthesis of the N-glycosylated proteins were significantly lower than in the control group 15 min after TTCE treatment. In normal rat liver, F1 secretory fraction of CA is 60-fold enriched in total dolichol content with respect to microsomes. In this compartment the total dolichol content, essential for the increase in membrane fluidity and permeability required for glycoprotein maturation and secretion, decreased significantly 5 min after TTCE treatment. Our results suggest that TTCE may affect dolichol functions in rat liver.


Subject(s)
Dolichols/metabolism , Ethane/analogs & derivatives , Golgi Apparatus/drug effects , Hydrocarbons, Chlorinated/toxicity , Microsomes, Liver/drug effects , Animals , Ethane/pharmacology , Ethane/toxicity , Free Radicals , Glycosylation , Golgi Apparatus/metabolism , Hydrocarbons, Chlorinated/pharmacology , Male , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...