Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Mol Med (Berl) ; 94(9): 1039-51, 2016 09.
Article in English | MEDLINE | ID: mdl-27038180

ABSTRACT

UNLABELLED: ∆(9)-Tetrahydrocannabinol (THC) is one of the major bioactive cannabinoids derived from the Cannabis sativa plant and is known for its anti-inflammatory properties. Delayed-type hypersensitivity (DTH) is driven by proinflammatory T helper cells including the classic inflammatory Th1 lineage as well as the more recently discovered Th17 lineage. In the current study, we investigated whether THC can alter the induction of Th1/Th17 cells involved in mBSA-induced DTH response. THC treatment (20 mg/kg) of C57BL/6 mice with DTH caused decreased swelling and infiltration of immune cells at the site of antigen rechallenge. Additionally, THC treatment decreased lymphocyte activation as well as Th1/Th17 lineage commitment, including reduced lineage-specific transcription factors and cytokines. Interestingly, while DTH caused an overexpression of miR-21, which increases Th17 differentiation via SMAD7 inhibition, and downregulation of miR-29b, an IFN-γ inhibitor, THC treatment reversed this microRNA (miR) dysregulation. Furthermore, when we transfected primary cells from DTH mice with miR-21 inhibitor or miR-29b mimic, as seen with THC treatment, the expression of target gene message was directly impacted increasing SMAD7 and decreasing IFN-γ expression, respectively. In summary, the current study suggests that THC treatment during DTH response can simultaneously inhibit Th1/Th17 activation via regulation of microRNA (miRNA) expression. KEY MESSAGES: • THC treatment inhibits simultaneous Th1/Th17 driven inflammation. • THC treatment corrects DTH-mediated microRNA dysregulation. • THC treatment regulates proinflammatory cytokines and transcription factors.


Subject(s)
Dronabinol/pharmacology , Hypersensitivity, Delayed/genetics , Hypersensitivity, Delayed/immunology , MicroRNAs/genetics , Th1 Cells/drug effects , Th1 Cells/physiology , Th17 Cells/drug effects , Th17 Cells/physiology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hypersensitivity, Delayed/drug therapy , Hypersensitivity, Delayed/pathology , Inflammation Mediators/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , RNA Interference , Transcription Factors/genetics
2.
Drug Metab Dispos ; 42(12): 2097-108, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25301936

ABSTRACT

Methamphetamine (MA), which remains one of the widely used drugs of abuse, is metabolized by the cytochrome P450 (P450) family of enzymes in humans. However, metabolism of methamphetamine in macaques is poorly understood. Therefore, we first developed and validated a very sensitive liquid chromatography with tandem mass spectrometry (LC-MS/MS) method using solid phase extraction of rhesus plasma with a lower limit of quantitation at 1.09 ng/ml for MA and its metabolites, 4-hydroxy methamphetamine (4-OH MA), amphetamine (AM), 4-OH amphetamine (4-OH AM), and norephedrine. We then analyzed plasma samples of MA-treated rhesus, which showed >10-fold higher concentrations of AM (∼29 ng/ml) and 4-OH AM (∼28 ng/ml) than MA (∼2 ng/ml). Because the plasma levels of MA metabolites in rhesus were much higher than in human samples, we examined MA metabolism in human and rhesus microsomes. Interestingly, the results showed that AM and 4-OH AM were formed more rapidly and that the catalytic efficiency (Vmax/Km) for the formation of AM was ∼8-fold higher in rhesus than in human microsomes. We further examined the differences in these kinetic characteristics using three selective inhibitors of each human CYP2D6 and CYP3A4 enzymes. The results showed that each of these inhibitors inhibited both d- and l-MA metabolism by 20%-60% in human microsomes but not in rhesus microsomes. The differences between human and rhesus CYP2D6 and CYP3A4 enzymes were further assessed by docking studies for both d and l-MA. In conclusion, our results demonstrated an enhanced MA metabolism in rhesus compared with humans, which is likely to be caused by differences in MA-metabolizing P450 enzymes between these species.


Subject(s)
Amphetamine/blood , Amphetamine/metabolism , Methamphetamine/blood , Methamphetamine/metabolism , Phenylpropanolamine/blood , Phenylpropanolamine/metabolism , Animals , Chromatography, Liquid/methods , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/metabolism , Humans , Kinetics , Macaca mulatta/metabolism , Microsomes, Liver/metabolism , Tandem Mass Spectrometry/methods
3.
PLoS One ; 9(10): e109603, 2014.
Article in English | MEDLINE | ID: mdl-25290377

ABSTRACT

Methamphetamine (MA) is a potent psychostimulant with a high addictive capacity, which induces many deleterious effects on the brain. Chronic MA abuse leads to cognitive dysfunction and motor impairment. MA affects many cells in the brain, but the effects on astrocytes of repeated MA exposure is not well understood. In this report, we used Gene chip array to analyze the changes in the gene expression profile of primary human astrocytes treated with MA for 3 days. Range of genes were found to be differentially regulated, with a large number of genes significantly downregulated, including NEK2, TTK, TOP2A, and CCNE2. Gene ontology and pathway analysis showed a highly significant clustering of genes involved in cell cycle progression and DNA replication. Further pathway analysis showed that the genes downregulated by multiple MA treatment were critical for G2/M phase progression and G1/S transition. Cell cycle analysis of SVG astrocytes showed a significant reduction in the percentage of cell in the G2/M phase with a concomitant increase in G1 percentage. This was consistent with the gene array and validation data, which showed that repeated MA treatment downregulated the genes associated with cell cycle regulation. This is a novel finding, which explains the effect of MA treatment on astrocytes and has clear implication in neuroinflammation among the drug abusers.


Subject(s)
Astrocytes/drug effects , Cell Cycle Checkpoints/drug effects , DNA Replication/drug effects , Methamphetamine/pharmacology , Sympathomimetics/pharmacology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclins/antagonists & inhibitors , Cyclins/genetics , Cyclins/metabolism , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Molecular Sequence Annotation , Multigene Family , NIMA-Related Kinases , Oligonucleotide Array Sequence Analysis , Poly-ADP-Ribose Binding Proteins , Primary Cell Culture , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Signal Transduction
4.
PLoS One ; 9(4): e93954, 2014.
Article in English | MEDLINE | ID: mdl-24699635

ABSTRACT

Endogenous cannabinoids [endocannabinoids] are lipid signaling molecules that have been shown to modulate immune functions. However, their role in the regulation of Th17 cells has not been studied previously. In the current study, we used methylated Bovine Serum Albumin [mBSA]-induced delayed type hypersensitivity [DTH] response in C57BL/6 mice, mediated by Th17 cells, as a model to test the anti-inflammatory effects of endocannabinoids. Administration of anandamide [AEA], a member of the endocannabinoid family, into mice resulted in significant mitigation of mBSA-induced inflammation, including foot pad swelling, cell infiltration, and cell proliferation in the draining lymph nodes [LN]. AEA treatment significantly reduced IL-17 and IFN-γ production, as well as decreased RORγt expression while causing significant induction of IL-10 in the draining LNs. IL-10 was critical for the AEA-induced mitigation of DTH response inasmuch as neutralization of IL-10 reversed the effects of AEA. We next analyzed miRNA from the LN cells and found that 100 out of 609 miRNA species were differentially regulated in AEA-treated mice when compared to controls. Several of these miRNAs targeted proinflammatory mediators. Interestingly, many of these miRNA were also upregulated upon in vitro treatment of LN cells with IL-10. Together, the current study demonstrates that AEA may suppress Th-17 cell-mediated DTH response by inducing IL-10 which in turn triggers miRNA that target proinflammatory pathways.


Subject(s)
Arachidonic Acids/pharmacology , Endocannabinoids/pharmacology , Hypersensitivity, Delayed/drug therapy , Interleukin-10/metabolism , MicroRNAs/metabolism , Polyunsaturated Alkamides/pharmacology , Th17 Cells/drug effects , Animals , Arachidonic Acids/therapeutic use , Cytokines/metabolism , Endocannabinoids/therapeutic use , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/metabolism , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/pharmacology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Polyunsaturated Alkamides/therapeutic use , Th17 Cells/metabolism
5.
J Leukoc Biol ; 95(4): 609-19, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24319288

ABSTRACT

Endocannabinoids are lipid-signaling molecules found in the nervous system; however, their precise role in the periphery is unclear. In the current study, we observed that a single i.p. administration of AEA caused rapid induction of MDSCs. The MDSCs contained a mixture of granulocytic and monocytic subtypes and expressed Arg-1 and iNOS. The MDSCs suppressed T cell proliferation in vitro and used iNOS to mediate their effect. Moreover, adoptive transfer of MDSCs led to suppression of mBSA-induced DTH. Through the use of pharmacological inhibition, as well as genetic knockout mice, we found that the induction of MDSCs by AEA was CB1-dependent. The induction of MDSCs by AEA was reduced significantly in mast cell-deficient mice, while maintained in LPS-insensitive mice, showing that the induction of MDSCs by AEA was dependent, at least in part, on mast cells and independent of TLR4. Chemokine analysis of AEA- treated WT mice showed an early spike of MCP-1, which was decreased in Kit(W/W-sh) mice, showing a role of mast cells in the secretion of MCP-1 in response to AEA. Also, use of antibodies against MCP-1 or mice deficient in MCP-1 confirmed the role played by MCP-1. Interestingly, MCP-1 played a significant role in the induction of monocytic but not granulocytic MDSCs. Our studies demonstrate for the first time that endocannaboinids activate CB1 on mast cells to induce MCP-1, which facilitates recruitment of monocytic MDSCs.


Subject(s)
Arachidonic Acids/pharmacology , Chemokine CCL2/physiology , Endocannabinoids/pharmacology , Mast Cells/physiology , Myeloid Cells/drug effects , Polyunsaturated Alkamides/pharmacology , Animals , CD11b Antigen/analysis , Female , Mice , Mice, Inbred C57BL , Myeloid Cells/physiology , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/physiology , Receptors, Chemokine/analysis
6.
J Biol Chem ; 288(52): 36810-26, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24202177

ABSTRACT

Δ(9)-Tetrahydrocannabinol (THC), the major bioactive component of marijuana, has been shown to induce functional myeloid-derived suppressor cells (MDSCs) in vivo. Here, we studied the involvement of microRNA (miRNA) in this process. CD11b(+)Gr-1(+) MDSCs were purified from peritoneal exudates of mice administered with THC and used for genome-wide miRNA profiling. Expression of CD31 and Ki-67 confirmed that the THC-MDSCs were immature and proliferating. THC-induced MDSCs exhibited distinct miRNA expression signature relative to various myeloid cells and BM precursors. We identified 13 differentially expressed (>2-fold) miRNA in THC-MDSCs relative to control BM precursors. In silico target prediction for these miRNA and pathway analysis using multiple bioinformatics tools revealed significant overrepresentation of Gene Ontology clusters within hematopoiesis, myeloid cell differentiation, and regulation categories. Insulin-like growth factor 1 signaling involved in cell growth and proliferation, and myeloid differentiation pathways were among the most significantly enriched canonical pathways. Among the differentially expressed, miRNA-690 was highly overexpressed in THC-MDSCs (∼16-fold). Transcription factor CCAAT/enhancer-binding protein α (C/EBPα) was identified as a potential functional target of miR-690. Supporting this, C/EBPα expression was attenuated in THC-MDSCs as compared with BM precursors and exhibited an inverse relation with miR-690. miR-690 knockdown using peptide nucleic acid-antagomiR was able to unblock and significantly increase C/EBPα expression establishing the functional link. Further, CD11b(+)Ly6G(+)Ly6C(+) and CD11b(+)Ly6G(-)Ly6C(+) purified subtypes showed high levels of miR-690 with attenuated C/EBPα expression. Moreover, EL-4 tumor-elicited MDSCs showed increased miR-690 expression. In conclusion, miRNA are significantly altered during the generation of functional MDSC from BM. Select miRNA such as miR-690 targeting genes involved in myeloid expansion and differentiation likely play crucial roles in this process and therefore in cannabinoid-induced immunosuppression.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , CCAAT-Enhancer-Binding Proteins/biosynthesis , Dronabinol/pharmacology , Gene Expression Regulation/drug effects , Immune Tolerance/drug effects , Myeloid Cells/metabolism , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Cells, Cultured , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Immune Tolerance/genetics , Mice , Myeloid Cells/cytology , Myeloid Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...