Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 334(2): 530-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20472667

ABSTRACT

Hyperlipidemia associated with the HIV protease inhibitor (PI), the major component of highly active antiretroviral treatment (HAART) for HIV infection, has stimulated interest in developing new agents that minimize these side effects in the clinic. HIV integrase inhibitor is a new class of anti-HIV agents. Raltegravir is a first-in-its-class oral integrase inhibitor and has potent inhibitory activity against HIV-1 strains that are resistant to other antiretroviral regimens. Our previous studies have demonstrated that HIV PI-induced endoplasmic reticulum (ER) stress links to dysregulation of lipid metabolism. However, little information is available as to whether raltegravir would have similar effects as the HIV PIs. In this study, we examined the effect of raltegravir on lipid metabolism both in primary rat hepatocytes and in in vivo mouse models, and we further determined whether the combination of raltegravir with existing HIV PIs would potentially exacerbate or prevent the previously observed development of dyslipidemia. The results indicated that raltegravir did not induce ER stress or disrupt lipid metabolism either in vitro or in vivo. However, HIV PI-induced ER stress and lipid accumulation were significantly inhibited by raltegravir both in in vitro primary rat hepatocytes and in in vivo mouse liver. High-performance liquid chromatography analysis further demonstrated that raltegravir did not affect the uptake and metabolism of HIV PIs in hepatocytes. Thus, raltegravir has less hepatic toxicity and could prevent HIV PI-induced dysregulation of lipid metabolism by inhibiting ER stress. These results suggest that incorporation of this HIV integrase inhibitor may reduce the side effects associated with current HAART.


Subject(s)
Endoplasmic Reticulum/physiology , HIV Integrase Inhibitors/pharmacology , HIV Protease Inhibitors/adverse effects , Lipid Metabolism/drug effects , Liver/drug effects , Pyrrolidinones/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Drug Antagonism , HIV Integrase Inhibitors/adverse effects , HIV Integrase Inhibitors/pharmacokinetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Hyperlipidemias/prevention & control , Lipid Metabolism/genetics , Liver/metabolism , Lopinavir , Male , Mice , Mice, Inbred C57BL , Pyrimidinones/adverse effects , Pyrrolidinones/adverse effects , Pyrrolidinones/pharmacokinetics , Raltegravir Potassium , Rats , Ritonavir/adverse effects , Signal Transduction
2.
Mol Pharm ; 7(3): 844-53, 2010 Jun 07.
Article in English | MEDLINE | ID: mdl-20349948

ABSTRACT

The development of HIV protease inhibitors (PIs) has been one of the most significant advances of the past decade in controlling HIV infection. Unfortunately, the benefits of HIV PIs are compromised by serious side effects. One of the most frequent and deleterious side effects of HIV PIs is severe gastrointestinal (GI) disorders including mucosal erosions, epithelial barrier dysfunction, and leak-flux diarrhea, which occurs in 16-62% of patients on HIV PIs. Although the underlying mechanisms behind HIV PI-associated serious adverse side effects remain to be identified, our recent studies have shown that activation of endoplasmic reticulum (ER) stress response plays a critical role in HIV PI-induced GI complications. The objective of this study was to develop a novel self-microemulsifying drug delivery system (SMEDDS) using various antioxidants as surfactants and cosurfactants to reduce the GI side effects of the most commonly used HIV PI, ritonavir. The biological activities of this SMSDDS of ritonavir were compared with that of Norvir, which is currently used in the clinic. Rat normal intestinal epithelial cells (IEC-6) and mouse Raw 264.7 macrophages were used to examine the effect of new SMEDDS of ritonavir on activation of ER stress and oxidative stress. Sprague-Dawley rats and C57/BL6 mice were used for pharmacokinetic studies and in vivo studies. The intracellular and plasma drug concentrations were determined by HPLC analysis. Activation of ER stress was detected by Western blot analysis and secreted alkaline phosphatase (SEAP) reporter assay. Reactive oxygen species (ROS) was measured using dichlorodihydrofluorescein diacetate as a probe. Cell viability was determined by Roche's cell proliferation reagent WST-1. Protein levels of inflammatory cytokines (TNF-alpha and IL-6) were determined by enzyme-linked immunosorbent assays (ELISA). The intestinal permeability was assessed by luminal enteral administration of fluorescein isothiocyanate conjugated dextran (FITC-dextran, 4 kDa). The pathologic changes in intestine were determined by histological examination. The results indicated that incorporation of antioxidants in this new SMEDDS not only significantly reduced ritonavir-induced ER stress activation, ROS production and apoptosis in intestinal epithelial cells and macrophages, but also improved the solubility, stability and bioavailability of ritonavir, and significantly reduced ritonavir-induced disruption of intestinal barrier function in vivo. In conclusion, this new SMEDDS of ritonavir has less GI side effects compared to Norvir. This new SMEDDS can be used for other HIV PIs and any insoluble antiviral drug with serious GI side effects.


Subject(s)
Drug Delivery Systems , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/adverse effects , Intestinal Mucosa/drug effects , Ritonavir/administration & dosage , Ritonavir/adverse effects , Alkaline Phosphatase/metabolism , Animals , Ascorbic Acid/administration & dosage , Ascorbic Acid/chemistry , Ascorbic Acid/therapeutic use , Blotting, Western , Cell Line , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , HIV Protease Inhibitors/chemistry , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Linoleic Acids/administration & dosage , Linoleic Acids/chemistry , Linoleic Acids/therapeutic use , Male , Mice , Mice, Inbred C57BL , Oleic Acid/administration & dosage , Oleic Acid/chemistry , Oleic Acid/therapeutic use , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Ritonavir/chemistry , Tumor Necrosis Factor-alpha/metabolism
3.
PLoS One ; 5(2): e9069, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20161729

ABSTRACT

BACKGROUND: HIV protease inhibitor (PI)-induced inflammatory response plays an important role in HIV PI-associated dyslipidemia and cardiovascular complications. This study examined the effect of berberine, a traditional herb medicine, on HIV PI-induced inflammatory response and further investigated the underlying cellular/molecular mechanisms in macrophages. METHODOLOGY AND PRINCIPAL FINDINGS: Cultured mouse J774A.1 macrophages and primary mouse macrophages were used in this study. The expression of TNF-alpha and IL-6 were detected by real-time RT-PCR and ELISA. Activations of ER stress and ERK signaling pathways were determined by Western blot analysis. Immunofluorescent staining was used to determine the intracellular localization of RNA binding protein HuR. RNA-pull down assay was used to determine the association of HuR with endogenous TNF-alpha and IL-6. Berberine significantly inhibited HIV PI-induced TNF-alpha and IL-6 expression by modulating ER stress signaling pathways and subsequent ERK activation, in turn preventing the accumulation of the RNA binding protein HuR in cytosol and inhibiting the binding of HuR to the 3'-UTRs of TNF-alpha and IL-6 in macrophages. CONCLUSIONS AND SIGNIFICANCE: Inhibition of ER stress represents a key mechanism by which berberine prevents HIV PI-induced inflammatory response. Our findings provide a new insight into the molecular mechanisms of berberine and show the potential application of berberine as a complimentary therapeutic agent for HIV infection.


Subject(s)
Berberine/pharmacology , Endoplasmic Reticulum/metabolism , HIV Protease Inhibitors/toxicity , Inflammation Mediators/metabolism , Macrophages/drug effects , Signal Transduction/drug effects , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Atazanavir Sulfate , Blotting, Western , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , ELAV Proteins , ELAV-Like Protein 1 , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lopinavir , Macrophages/cytology , Macrophages/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Oligopeptides/toxicity , Pyridines/toxicity , Pyrimidinones/toxicity , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ritonavir/toxicity , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
Biochem Pharmacol ; 78(1): 70-7, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19447225

ABSTRACT

HIV protease inhibitor (PI)-associated cardiovascular risk, especially atherosclerosis, has become a major concern in the clinic. Macrophages are key players in the inflammatory response and atherosclerosis formation. We have previously shown that HIV PIs induce endoplasmic reticulum (ER) stress, activate the unfolded protein response (UPR), and increase the synthesis of the inflammatory cytokines, TNF-alpha and IL-6, by regulating the intracellular translocation of RNA binding protein HuR in macrophages. However, the underlying signaling mechanisms remain unclear. We show here that the HIV PI lopinavir significantly activated the extracellular-signal regulated protein kinase (ERK), but not c-Jun N-terminal kinase (JNK) and p38 MAPK. Lopinavir-induced cytosolic translocation of HuR and TNF-alpha and IL-6 synthesis was attenuated by specific chemical inhibitor of MEK (PD98058) or over-expression of dominant negative mutant of MEK1. In addition, we demonstrated that lopinavir-induced ERK activation and TNF-alpha and IL-6 expression were completely inhibited in macrophages from CHOP null mice. Taken together, these results indicate activation of the UPR plays an essential role in HIV PI-induced inflammatory cytokine synthesis and release by activating ERK, which increases the cytosolic translocation of HuR and subsequent binding to the 3'UTR of TNF-alpha and IL-6 mRNAs in macrophages.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , HIV Protease Inhibitors/pharmacology , Interleukin-6/genetics , Macrophages/physiology , Pyrimidinones/pharmacology , Tumor Necrosis Factor-alpha/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Leukemia, Monocytic, Acute , Lopinavir , MAP Kinase Signaling System , Macrophages/drug effects , Mice , Mice, Knockout , NF-kappa B/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Denaturation , Signal Transduction/drug effects , Signal Transduction/physiology
5.
J Biol Chem ; 282(34): 24607-14, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17603092

ABSTRACT

Bile acids are required for intestinal absorption and biliary solubilization of cholesterol and lipids. In addition, bile acids play a crucial role in cholesterol homeostasis. One of the key enzymes in the bile acid biosynthetic pathways is cholesterol 7alpha-hydroxylase/cytochrome P450 7alpha-hydroxylase (7alpha-hydroxylase), which is the rate-limiting and regulatory step of the "classic" pathway. Transcription of the 7alpha-hydroxylase gene is highly regulated. Two nuclear receptors, hepatocyte nuclear factor 4alpha (HNF-4alpha) and alpha(1)-fetoprotein transcription factor, are required for both transcription and regulation by different physiological events. It has been shown that some mitogen-activated protein kinases, such as the c-Jun N-terminal kinase and the ERK, play important roles in the regulation of 7alpha-hydroxylase transcription. In this study, we show evidence that the p38 kinase pathway plays an important role in 7alpha-hydroxylase expression and hence in bile acid synthesis. Inhibition of p38 kinase activity in primary hepatocytes results in approximately 5-10-fold reduction of 7alpha-hydroxylase mRNA. This suppression is mediated, at least in part, through HNF-4alpha. Inhibition of p38 kinase activity diminishes HNF-4alpha nuclear protein levels and its phosphorylation in vivo and in vitro, and it renders a less stable protein. Induction of the p38 kinase pathway by insulin results in an increase in HNF-4alpha protein and a concomitant induction of 7alpha-hydroxylase expression that is blocked by inhibiting the p38 pathway. These studies show a functional link between the p38 signaling pathway, HNF-4alpha, and bile acid synthesis.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol 7-alpha-Hydroxylase/biosynthesis , Gene Expression Regulation, Enzymologic , Hepatocyte Nuclear Factor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Hepatocytes/metabolism , Models, Biological , Phosphorylation , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
6.
J Biol Chem ; 279(7): 5821-8, 2004 Feb 13.
Article in English | MEDLINE | ID: mdl-14660582

ABSTRACT

We have shown previously that bile acids can activate the JNK pathway and down-regulate cholesterol 7alpha-hydroxylase (CYP7A1), the rate-limiting enzyme in the neutral pathway of bile acid biosynthesis. In this study, the mechanism(s) by which deoxycholic acid (DCA) activates the JNK pathway were examined. FAS receptor (FAS-R) and acidic sphingomyelinase (ASM)-deficient hepatocytes were resistant to DCA-induced activation of the JNK pathway. Activation of the JNK pathway (2-3-fold) in response to tumor necrosis factor-alpha was similar in both wild-type and FAS-R(-/-) hepatocytes. In wild-type and FAS-R(-/-) hepatocytes, ceramide elevation was detected as early as 2 min and peaked at 10 min after DCA treatment. In contrast, ASM(-/-) hepatocytes were defective in DCA-induced ceramide generation. Treatment with DCA resulted in movement of FAS-R to the cell surface, which was blocked upon treatment with brefeldin A. However, brefeldin A failed to block DCA-mediated JNK activation in wild-type hepatocytes. DCA-induced JNK activation was independent of either the epidermal growth factor receptor activation or free radical generation. Addition of ASM to rat hepatocytes activated JNK and down-regulated CYP7A1 mRNA levels. In conclusion, these results show that DCA activates JNK and represses CYP7A1 mRNA levels in primary hepatocytes via an ASM/FAS-R-dependent mechanism that is independent of either the epidermal growth factor receptor or free radical generation.


Subject(s)
Deoxycholic Acid/metabolism , Hepatocytes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Sphingomyelin Phosphodiesterase/metabolism , fas Receptor/metabolism , Alleles , Animals , Bile Acids and Salts/metabolism , Brefeldin A/pharmacology , Cells, Cultured , Ceramides/metabolism , Cholesterol 7-alpha-Hydroxylase/biosynthesis , Cholesterol 7-alpha-Hydroxylase/metabolism , Down-Regulation , Enzyme Activation , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Free Radicals , JNK Mitogen-Activated Protein Kinases , Ligands , MAP Kinase Kinase 4 , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Time Factors , Tumor Necrosis Factor-alpha/metabolism
7.
Hepatology ; 35(2): 307-14, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11826403

ABSTRACT

Bile acids have been reported to activate several different cell signaling cascades in rat hepatocytes. However, the mechanism(s) of activation of these pathways have not been determined. This study aims to determine which bile acids activate the Raf-1/MEK/ERK cascade and the mechanism of activation of this pathway. Taurodeoxycholic acid (TDCA) stimulated (+235%) the phosphorylation of p(74) Raf-1 in a time (5 to 20 minutes) and concentration-dependent (10 to 100 micromol/L) manner. Raf-1 and ERK activities were both significantly increased by most bile acids tested. Deoxycholic acid (DCA) was the best activator of ERK (3.6-fold). A dominant negative Ras (N17) construct expressed in primary hepatocytes prevented the activation of ERK by DCA. The epidermal growth factor receptor (EGFR)-specific inhibitor (AG1478) significantly inhibited (approximately 81%) the activation of ERK by DCA. DCA rapidly (30 to 60 seconds) increased phosphorylation of the EGFR (approximately 2-fold) and Shc (approximately 4-fold). A dominant negative mutant of the EGFR (CD533) blocked the ability of DCA to activate ERK. In conclusion, these results show that DCA activates the Raf-1/MEK/ERK signaling cascade in primary hepatocytes primarily via an EGFR/Ras-dependent mechanism.


Subject(s)
Deoxycholic Acid/pharmacology , Detergents/pharmacology , Hepatocytes/drug effects , Hepatocytes/enzymology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Taurodeoxycholic Acid/pharmacology , Animals , Cells, Cultured , Enzyme Activation/drug effects , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Genes, Dominant , Hepatocytes/metabolism , Rats , ras Proteins/genetics , ras Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...