Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Neuroimmunol ; 332: 37-48, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30933849

ABSTRACT

Fundamentally, microglia have two activation states, a pro-inflammatory neurotoxic (M1) and an anti-inflammatory neuroprotective (M2) phenotype, and their conversion from M1-like to M2-like microglia may provide therapeutic benefits to prevent neuronal loss in neurodegenerative diseases such as Parkinson's disease (PD). Previously, we showed that Salmeterol, a long-acting ß2-adrenergic receptor (ß2-AR) agonist, has neuroprotective effects in PD models in vitro and in vivo through the ß-arrestin2-dependent inhibition of pro-inflammatory M1-type mediator production. In the present study, we explored whether Salmeterol can mediate phenotypic conversion in LPS-activated murine microglial BV2 cells from the neurotoxic M1-like to a neuroprotective M2-like phenotype. Salmeterol inhibited the production of LPS-induced mediators of the pro-inflammatory M1 phenotype such as tumor necrosis factor-α (TNF-α), IL-(interleukin) 18, IL-6, chemokines (CCL2, CCL3, CCL4) and reactive oxygen species from BV2 cells. Conversely, treatment with Salmeterol and other ß2-AR agonists robustly enhances the production of the M2 cytokine IL-10 from LPS-activated microglia. In addition, Salmeterol upregulates the expression of arginase-1 and CXCL14. Furthermore, using siRNA approach we found that silencing of the transcription factor Creb abrogates the Salmeterol-mediated production of IL-10 in LPS-activated BV2 cells, but silencing of ß-arrestin2 with Arrb2 siRNA did not. In addition, our data shows conversion from an M1- to M2-like phenotype in LPS-activated microglia by ß2-AR agonists involves activation of the classical cAMP/PKA/CREB as well as the PI3K and p38 MAPK signaling pathways, and provides a novel therapeutic approach targeting microglial cell activation and inducing their phenotypic conversion in the treatment of neuroinflammatory diseases such as PD.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Microglia/drug effects , Salmeterol Xinafoate/pharmacology , Signal Transduction/drug effects , Animals , Biomarkers , Cell Line , Cyclic AMP Response Element-Binding Protein/physiology , Cytokines/biosynthesis , Endotoxins/pharmacology , Gene Expression Regulation/drug effects , Inflammation , Interleukin-10/biosynthesis , Interleukin-10/physiology , MAP Kinase Signaling System/drug effects , Mice , Phenotype , Phosphatidylinositol 3-Kinases/physiology , RNA Interference , Reactive Oxygen Species/metabolism
2.
J Neuroimmunol ; 325: 10-19, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30352316

ABSTRACT

Microglial activation contributes to chronic inflammation and neuronal loss in progressive neurodegenerative disorders such as Parkinson's disease (PD). Thus, treatments suppressing microglial activation may have therapeutic benefits to prevent neuronal loss in neurodegenerative diseases. Our previous findings show that Salmeterol, a long-acting ß2-adrenergic receptor (ß2-AR) agonist, is neuroprotective in two distinct animal models of PD, including where lipopolysaccharide (LPS) from E. coli was used to initiate chronic neurodegeneration. Salmeterol was found to be a potent inhibitor of dopaminergic neurodegeneration by regulating the production of pro-inflammatory mediators from activated microglial cells. In the present study, we investigated the molecular basis of the anti-inflammatory effects of Salmeterol on LPS-activated murine microglial BV2 cells. BV2 cells were pretreated with Salmeterol and followed by stimulation with LPS. Salmeterol inhibited LPS-induced release of the pro-inflammatory mediators such as tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and nitric oxide from BV2 cells. Additionally, Salmeterol suppressed nuclear translocation of nuclear factor kappa-B (NF-κB) p65 by inhibiting the IκB-α degradation and TAK1 (transforming growth factor-beta-activated kinase1) phosphorylation. We have also found that Salmeterol increases the expression of ß-arrestin2 and enhances the interaction between ß-arrestin2 and TAB1 (TAK1-binding protein), reduced TAK1/TAB1 mediated activation of NFκB and expression of pro-inflammatory genes. Furthermore, silencing of ß-arrestin2 abrogates the anti-inflammatory effects of Salmeterol in LPS-stimulated BV2 cells. Our findings suggest that the anti-inflammatory properties of Salmeterol is ß-arrestin2 dependent and also offers novel therapeutics targeting inflammatory pathways to prevent microglial cell activation and neuronal loss in neuroinflammatory diseases like PD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Microglia/metabolism , Salmeterol Xinafoate/pharmacology , beta-Arrestin 2/physiology , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Cell Line , Inflammation Mediators/antagonists & inhibitors , Mice , Microglia/drug effects
3.
J Periodontol ; 88(7): 681-692, 2017 07.
Article in English | MEDLINE | ID: mdl-28398147

ABSTRACT

BACKGROUND: Salmeterol is a long-acting ß2-adrenergic receptor agonist used to treat chronic obstructive pulmonary disease. The authors of the current study previously showed that preincubation of primary microglial-enriched cells with salmeterol could inhibit the inflammatory response induced by Escherichia coli lipopolysaccharide (LPS), a Toll-like receptor (TLR)-4 agonist. In this study, the authors sought to determine if salmeterol had a similar inhibitory effect on the inflammatory response of the murine macrophage cell line RAW264.7 and human monocyte THP-1 to LPS from Porphyromonas gingivalis (PgLPS), an oral microbe implicated in the pathogenesis of periodontal disease. METHODS: RAW264.7 and THP-1 cells were pretreated with salmeterol, followed by PgLPS, and monitored for production of inflammatory mediators by enzyme-linked immunosorbent assay. The nitric oxide concentration and nuclear factor-kappa B (NF-κB) activity were measured by Griess method and secretory alkaline phosphatase reporter activity assay, respectively. Reverse-transcriptase polymerase chain reaction and immunoblot analysis were used to measure messenger RNA and protein levels. Nuclear translocation of NF-κB was detected by immunofluorescence. RESULTS: Pretreatment with salmeterol significantly inhibited production of proinflammatory mediators by RAW264.7 and THP-1 cells. Salmeterol downregulated PgLPS-mediated phosphorylation of the extracellular signal-regulated kinase 1/2 and c-Jun N-terminal kinase but not p38 mitogen-activated protein kinases (MAPKs). Salmeterol also attenuated activation of NF-κB via inhibition of nuclear translocation of p65-NFκB, the transcriptional activity of NF-κB and IκBα phosphorylation. CONCLUSION: Salmeterol can significantly inhibit activation of macrophage-mediated inflammation by PgLPS, suggesting that use of salmeterol may be an effective treatment in inhibiting or lessening the inflammatory response mediated through TLR pathway activation.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Macrophage Activation/drug effects , Salmeterol Xinafoate/pharmacology , Adrenergic beta-2 Receptor Agonists/therapeutic use , Animals , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Mice , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Phosphorylation , Porphyromonas gingivalis , RAW 264.7 Cells , Salmeterol Xinafoate/therapeutic use , THP-1 Cells , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Mediators Inflamm ; 2012: 401264, 2012.
Article in English | MEDLINE | ID: mdl-22544998

ABSTRACT

Significant evidence has now been accumulated that microglial cells play a central role in the degeneration of DA neurons in animal models of PD. The oxidative stress response by microglial cells, most notably the activity of the enzyme NADPH oxidase, appears to play a central role in the pathology of PD. This oxidative stress response occurs in microglia through the activation of the ERK signaling pathway by proinflammatory stimuli, leading to the phosphorylation and translocation of the p47(phox) and p67(phox) cytosolic subunits, the activation of membrane-bound PHOX, and the production of ROS. Therapeutic anti-inflammatories which prevent DA neurodegeneration in PD, including anti-inflammatory cytokines, morphinan compounds, NADPH oxidase inhibitors, NF-κB inhibitors, and ß2-AR agonists, all function to inhibit the activation of the PHOX in microglial cells. These observations suggest a central role for the oxidative stress response in microglial cells as a mediator or regulator of DA neurodegeneration in PD.


Subject(s)
Microglia/metabolism , Oxidative Stress , Parkinson Disease/metabolism , Animals , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation/metabolism , Models, Biological , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Neurons/metabolism , Oxidation-Reduction , Phosphorylation , Reactive Oxygen Species , Signal Transduction
5.
Parkinsons Dis ; 2011: 216298, 2011.
Article in English | MEDLINE | ID: mdl-21603248

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative condition characterized by chronic inflammation. Nuclear factor κB (NF-κB) is a family of inducible transcription factors that are expressed in a wide variety of cells and tissues, including microglia, astrocytes, and neurons, and the classical NF-κB pathway plays a key role in the activation and regulation of inflammatory mediator production during inflammation. Activation of the classical NF-κB pathway is mediated through the activity of the IKK kinase complex, which consists of a heterotrimer of IKKα, IKKß, and IKKγ subunits. Targeting NF-κB has been proposed as an approach to the treatment of acute and chronic inflammatory conditions, and the use of inhibitors specific for either IKKß or IKKγ has now been found to inhibit neurodegeneration of TH+ DA-producing neurons in murine and primate models of Parkinson's disease. These studies suggest that targeting the classical pathway of NF-κB through the inhibition of the IKK complex can serve as a useful therapeutic approach to the treatment of PD.

6.
J Immunol ; 186(7): 4443-54, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21335487

ABSTRACT

The role of the ß2 adrenergic receptor (ß2AR) in the regulation of chronic neurodegenerative inflammation within the CNS is poorly understood. The purpose of this study was to determine neuroprotective effects of long-acting ß2AR agonists such as salmeterol in rodent models of Parkinson's disease. Results showed salmeterol exerted potent neuroprotection against both LPS and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/1-methyl-4-phenylpyridinium-induced dopaminergic neurotoxicity both in primary neuron-glia cultures (at subnanomolar concentrations) and in mice (1-10 µg/kg/day doses). Further studies demonstrated that salmeterol-mediated neuroprotection is not a direct effect on neurons; instead, it is mediated through the inhibition of LPS-induced microglial activation. Salmeterol significantly inhibited LPS-induced production of microglial proinflammatory neurotoxic mediators, such as TNF-α, superoxide, and NO, as well as the inhibition of TAK1-mediated phosphorylation of MAPK and p65 NF-κB. The anti-inflammatory effects of salmeterol required ß2AR expression in microglia but were not mediated through the conventional G protein-coupled receptor/cAMP pathway. Rather, salmeterol failed to induce microglial cAMP production, could not be reversed by either protein kinase A inhibitors or an exchange protein directly activated by cAMP agonist, and was dependent on ß-arrestin2 expression. Taken together, our results demonstrate that administration of extremely low doses of salmeterol exhibit potent neuroprotective effects by inhibiting microglial cell activation through a ß2AR/ß-arrestin2-dependent but cAMP/protein kinase A-independent pathway.


Subject(s)
Adrenergic beta-2 Receptor Agonists/therapeutic use , Dopamine/toxicity , Microglia/immunology , Neural Inhibition/immunology , Neuroprotective Agents/therapeutic use , Signal Transduction/immunology , Adrenergic beta-2 Receptor Agonists/metabolism , Animals , Cells, Cultured , Coculture Techniques , Dopamine/biosynthesis , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/therapeutic use , Inflammation Mediators/toxicity , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/pathology , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neuroprotective Agents/metabolism , Parkinson Disease/immunology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rats , Rats, Inbred F344 , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Craniofac Growth Ser ; 48: 377-391, 2011.
Article in English | MEDLINE | ID: mdl-26549917

ABSTRACT

Current methods to assess outcomes and change in orthodontics are comparison of photographs, cephalometric measurements and superimpositions, and comparisons/measurements on dental casts. Digital models are a relatively new records modality in orthodontics. They offer numerous advantages in terms of storage space, spatial registration and superimposition. The purpose of this chapter is to determine the reproducibility of: 1) establishing occlusion of independently scanned digital models; and 2) registering digital models obtained after treatment on their homologous digital model setups produced before treatment. Reliability of both procedures was assessed with two random samples of five patient's models. In both experiments, three replicate positionings of the models per patient were created and variability in position was evaluated by the maximum surface difference between replicates, and the standard deviation of the surface distances between replicates respectively. Based on the data obtained, we concluded that it is reliable to register independently scanned models to a scanned surface of the models in occlusion. Surface-to-surface registration of final orthodontic digital models to planned setup models also is reproducible.

8.
J Neural Transm (Vienna) ; 117(8): 971-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20571837

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative movement disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra and depletion of dopamine in the striatum, which lead to pathological and clinical abnormalities. Increasing evidence has demonstrated that inflammation is the fundamental process contributing to neuron death in PD. Neuroinflammation, which is characterized by activated microglia and infiltrating T cells at sites of neuronal injury, is a prominent contributor to the pathogenesis of progressive PD. Microglia play a critical role in forming a self-propelling cycle leading to sustained chronic neuroinflammation and driving the progressive neurodegeneration in PD. This activation depends heavily on the respiratory burst within the microglia, which in turn regulates a number of downstream pro-inflammatory activities. On the other hand, the adaptive immune responses, most notably T cells, are now emerging as important components of the inflammatory response that contribute to the pathogenesis of PD. This review paper focus on the understanding of the inflammatory etiology of PD, as well as the molecular signaling involved in this inflammatory response, with the aim to provide more effective treatments to slow down or halt the progression of chronic inflammation-induced CNS disorders, such as PD.


Subject(s)
Brain/immunology , Inflammation , Oxidative Stress/immunology , Parkinson Disease , Animals , Brain/metabolism , Brain/pathology , Humans , Inflammation/complications , Inflammation/pathology , Inflammation/therapy , Microglia/physiology , Parkinson Disease/etiology , Parkinson Disease/immunology , Parkinson Disease/therapy , T-Lymphocytes/physiology
9.
J Pharmacol Exp Ther ; 333(3): 822-33, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20190013

ABSTRACT

Parkinson's disease (PD) is a progressive neurological disorder characterized by a selective loss of dopamine (DA) neurons in the substantia nigra (SN). Although current therapy can control symptoms of this disorder, there is no effective therapy available to halt its progression. Recently, neuroinflammation has been recognized as an important contributor to the pathogenesis of PD, and nuclear factor-kappaB (NF-kappaB) plays a key role in regulating neuroinflammation. Hence, the modulation of NF-kappaB pathway may have therapeutic potential for PD. Activation of NF-kappaB depends on the phosphorylation of its inhibitor, IkappaB, by the specific IkappaB kinase (IKK) subunit IKK-beta. Compound A (7-[2-(cyclopropylmethoxy)-6-hydroxyphenyl]-5-[(3S)-3-piperidinyl]-1, 4-dihydro-2H-pyrido[2,3-d][1,3]oxazin-2-one hydrochloride), a potent and selective inhibitor of IKK-beta, has recently been reported to provide cardioprotection through specific suppression of NF-kappaB signaling. The present study, for the first time, elucidates neuroprotective effects of compound A. Daily subcutaneous injection of compound A (1 mg/kg) for 7 days inhibited the activation of microglia induced by nigral stereotaxic injection of lipopolysaccharide (LPS) and significantly attenuated LPS-induced loss of DA neurons in the SN. In vitro mechanistic studies revealed that neuroprotective effects of compound A were mediated by 1) suppressing the activity of microglial NADPH oxidase and decreasing the production of reactive oxygen species, and 2) inhibiting NF-kappaB-mediated gene transcription of various proinflammatory mediators in microglia via IKK-beta suppression. These findings indicate that compound A afforded potent neuroprotection against LPS-induced neurodegeneration through selective inhibition of NF-kappaB activation and may be of potential benefit in the treatment of PD.


Subject(s)
Dopamine/physiology , Enzyme Inhibitors/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Neurons/drug effects , Neuroprotective Agents , Neurotoxins/antagonists & inhibitors , Oxazines/pharmacology , Pyridines/pharmacology , Animals , Astrocytes/drug effects , Blotting, Western , Cells, Cultured , Dopamine/metabolism , Immunohistochemistry , Indicators and Reagents , Interleukin-1beta/metabolism , Male , Microglia/drug effects , NADPH Oxidases/antagonists & inhibitors , Neuroglia/drug effects , Nitrites/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Superoxides/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Mol Immunol ; 46(11-12): 2256-66, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19477016

ABSTRACT

The beta(3)-adrenergic receptor (beta(3)AR) is an essential regulator of metabolic and endocrine functions. A major cellular and clinically significant consequence of beta(3)AR activation is the substantial elevation in interleukin-6 (IL-6) levels. Although the beta(3)AR-dependent regulation of IL-6 expression is well established, the cellular pathways underlying this regulation have not been characterized. Using a novel method of homogenous reporters, we assessed the pattern of activation of 43 transcription factors in response to the specific beta(3)AR agonist CL316243 in adipocytes, cells that exhibit the highest expression of beta(3)ARs. We observed a unique and robust activation of the CRE-response element, suggesting that IL-6 transcription is regulated via the G(s)-protein/cAMP/protein kinase A (PKA) but not nuclear factor kappa B (NF-kappaB) pathway. However, pretreatment of adipocytes with pharmacologic inhibitors of PKA pathway failed to block beta(3)AR-mediated IL-6 up-regulation. Additionally, stimulation of adipocytes with the exchange protein directly activated by cAMP (Epac) agonist did not induce IL-6 expression. Instead, the beta(3)AR-mediated transcription of IL-6 required activation of both the p38 and PKC pathways. Western blot analysis further showed that transcription factors CREB and ATF-2 but not ATF-1 were activated in a p38- and PKC-dependent manner. Collectively, our results suggest that while stimulation of the beta(3)AR leads to a specific activation of CRE-dependent transcription, there are several independent cellular pathways that converge at the level of CRE-response element activation, and in the case of IL-6 this activation is mediated by p38 and PKC but not PKA pathways.


Subject(s)
Adipocytes, White/metabolism , Interleukin-6/biosynthesis , Receptors, Adrenergic, beta-3/physiology , Activating Transcription Factor 1/metabolism , Adipocytes, White/cytology , Adrenergic beta-3 Receptor Agonists , Animals , Cell Differentiation/physiology , Cell Line , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Dioxoles/pharmacology , GTP-Binding Protein alpha Subunits, Gs/physiology , Gene Expression Regulation , Humans , Mice , NF-kappa B/physiology , Protein Kinase C/physiology , Response Elements , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Glia ; 57(15): 1600-9, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19330844

ABSTRACT

Activation of the beta2 adrenergic receptor (beta2AR) on immune cells has been reported to possess anti-inflammatory properties, however, the pro-inflammatory properties of beta2AR activation remain unclear. In this study, using rat primary mesencephalic neuron-glia cultures, we report that salmeterol, a long-acting beta2AR agonist, selectively induces dopaminergic (DA) neurotoxicity through its ability to activate microglia. Salmeterol selectively increased the production of reactive oxygen species (ROS) by NADPH oxidase (PHOX), the major superoxide-producing enzyme in microglia. A key role of PHOX in mediating salmeterol-induced neurotoxicity was demonstrated by the inhibition of DA neurotoxicity in cultures pretreated with diphenylene-iodonium (DPI), an inhibitor of PHOX activity. Mechanistic studies revealed the activation of microglia by salmeterol results in the selective phosphorylation of ERK, a signaling pathway required for the translocation of the PHOX cytosolic subunit p47(phox) to the cell membrane. Furthermore, we found ERK inhibition, but not protein kinase A (PKA) inhibition, significantly abolished salmeterol-induced superoxide production, p47(phox) translocation, and its ability to mediate neurotoxicity. Together, these findings indicate that beta2AR activation induces microglial PHOX activation and DA neurotoxicity through an ERK-dependent/PKA-independent pathway.


Subject(s)
Dopamine/metabolism , Microglia/drug effects , Microglia/enzymology , NADPH Oxidases/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/physiology , Adrenergic beta-Agonists/pharmacology , Albuterol/analogs & derivatives , Albuterol/pharmacology , Analysis of Variance , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Mesencephalon/cytology , Neurons/drug effects , Neurons/physiology , Pregnancy , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Salmeterol Xinafoate , Signal Transduction/drug effects , Superoxides/metabolism , Tritium/metabolism , Tyrosine 3-Monooxygenase/metabolism
12.
J Immunol ; 181(1): 660-8, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18566433

ABSTRACT

TGF-beta1 is one of the most potent endogenous immune modulators of inflammation. The molecular mechanism of its anti-inflammatory effect on the activation of the transcription factor NF-kappaB has been well-studied; however, the potential effects of TGF-beta1 on other proinflammatory signaling pathways is less clear. In this study, using the well-established LPS and the 1-methyl-4-phenylpyridinium-mediated models of Parkinson's disease, we demonstrate that TGF-beta1 exerts significant neuroprotection in both models via its anti-inflammatory properties. The neuroprotective effects of TGF-beta1 are mainly attributed to its ability to inhibit the production of reactive oxygen species from microglia during their activation or reactivation. Moreover, we demonstrate that TGF-beta1 inhibited LPS-induced NADPH oxidase (PHOX) subunit p47phox translocation from the cytosol to the membrane in microglia within 10 min. Mechanistic studies show that TGF-beta1 fails to protect dopaminergic neurons in cultures from PHOX knockout mice, and significantly reduced LPS-induced translocation of the PHOX cytosolic subunit p47phox to the cell membrane. In addition, LPS-induced ERK phosphorylation and subsequent Ser345 phosphorylation on p47phox were significantly inhibited by TGF-beta1 pretreatment. Taken together, our results show that TGF-beta1 exerted potent anti-inflammatory and neuroprotective properties, either through the prevention of the direct activation of microglia by LPS, or indirectly through the inhibition of reactive microgliosis elicited by 1-methyl-4-phenylpyridinium. The molecular mechanisms of TGF-beta1-mediated anti-inflammatory properties is through the inhibition of PHOX activity by preventing the ERK-dependent phosphorylation of Ser345 on p47phox in microglia to reduce oxidase activities induced by LPS.


Subject(s)
Membrane Glycoproteins/metabolism , Microglia/drug effects , Microglia/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NADPH Oxidases/metabolism , Phosphoserine/metabolism , Transforming Growth Factor beta1/pharmacology , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Cell Membrane/metabolism , Cytoprotection/drug effects , Cytosol/metabolism , Dopamine/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport , Rats , Reactive Oxygen Species/metabolism , Tissue Culture Techniques
13.
Environ Health Perspect ; 116(5): 593-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18470306

ABSTRACT

BACKGROUND: Parkinson disease (PD), a chronic neurodegenerative disease, has been proposed to be a multifactorial disorder resulting from a combination of environmental mechanisms (chemical, infectious, and traumatic), aging, and genetic deficits. Microglial activation is important in the pathogenesis of PD. OBJECTIVES: We investigated dopaminergic (DA) neurotoxicity and the underlying mechanisms of formyl-methionyl-leucyl-phenylalanine (fMLP), a bacteria-derived peptide, in relation to PD. METHODS: We measured DA neurotoxicity using a DA uptake assay and immunocytochemical staining (ICC) in primary mesencephalic cultures from rodents. Microglial activation was observed via ICC, flow cytometry, and superoxide measurement. RESULTS: fMLP can cause selective DA neuronal loss at concentrations as low as 10(-13) M. Further, fMLP (10(-13) M) led to a significant reduction in DA uptake capacity in neuron/glia (N/G) cultures, but not in microglia-depleted cultures, indicating an indispensable role of microglia in fMLP-induced neurotoxicity. Using ICC of a specific microglial marker, OX42, we observed morphologic changes in activated microglia after fMLP treatment. Microglial activation after fMLP treatment was confirmed by flow cytometry analysis of major histocompatibility antigen class II expression on a microglia HAPI cell line. Mechanistic studies revealed that fMLP (10(-13) M)-induced increase in the production of extracellular superoxide from microglia is critical in mediating fMLP-elicited neurotoxicity. Pharmacologic inhibition of NADPH oxidase (PHOX) with diphenylene-iodonium or apocynin abolished the DA neurotoxicity of fMLP. N/G cultures from PHOX-deficient (gp91PHOX-/ -) mice were also insensitive to fMLP-induced DA neurotoxicity. CONCLUSION: fMLP (10(-13) M) induces DA neurotoxicity through activation of microglial PHOX and subsequent production of superoxide, suggesting a role of fMLP in the central nervous system inflammatory process.


Subject(s)
Central Nervous System Infections/etiology , Dopamine/metabolism , Microglia/drug effects , N-Formylmethionine Leucyl-Phenylalanine/toxicity , Neurodegenerative Diseases/chemically induced , Parkinson Disease/etiology , Animals , Cells, Cultured , Central Nervous System Infections/metabolism , Central Nervous System Infections/pathology , Enzyme Activation , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/enzymology , Microglia/metabolism , NADPH Oxidases/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress , Pregnancy , Rats , Rats, Inbred F344
14.
Immunol Res ; 41(3): 155-64, 2008.
Article in English | MEDLINE | ID: mdl-18512160

ABSTRACT

Chronic inflammation mediated by microglial cells is the fundamental process contributing to the death of dopamine (DA)-producing neurons in the brain. Production of inflammatory products by these microglial cells characterizes the slow destructive process in Parkinson's disease (PD). The activation of microglial cells and the generation of pro-inflammatory cytokines that characterize PD are mediated by several different signaling pathways, with the activation of the respiratory burst by microglial cells being a critical event in the ultimate toxicity of DA-neurons. The work on our lab is concerned with understanding the mechanisms of activation, response, and therapeutic targets of microglial cells, with the aim to provide more effective treatments for PD and other inflammatory diseases of the CNS.


Subject(s)
Brain/immunology , Cytokines/metabolism , Microglia/immunology , Morphinans/metabolism , Parkinson Disease/immunology , Parkinson Disease/physiopathology , Brain/metabolism , Chronic Disease , Cytokines/immunology , Dopamine/immunology , Dopamine/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Microglia/metabolism , Morphinans/immunology , Parkinson Disease/metabolism
15.
J Neuroinflammation ; 4: 23, 2007 Sep 19.
Article in English | MEDLINE | ID: mdl-17880684

ABSTRACT

BACKGROUND: The mechanisms involved in the induction and regulation of inflammation resulting in dopaminergic (DA) neurotoxicity in Parkinson's disease (PD) are complex and incompletely understood. Microglia-mediated inflammation has recently been implicated as a critical mechanism responsible for progressive neurodegeneration. METHODS: Mesencephalic neuron-glia cultures and reconstituted cultures were used to investigate the molecular mechanisms of sinomenine (SN)-mediated anti-inflammatory and neuroprotective effects in both the lipopolysaccharide (LPS)- and the 1-methyl-4-phenylpyridinium (MPP+)-mediated models of PD. RESULTS: SN showed equivalent efficacy in protecting against DA neuron death in rat midbrain neuron-glial cultures at both micro- and sub-picomolar concentrations, but no protection was seen at nanomolar concentrations. The neuroprotective effect of SN was attributed to inhibition of microglial activation, since SN significantly decreased tumor necrosis factor-alpha (TNF-alpha, prostaglandin E2 (PGE2) and reactive oxygen species (ROS) production by microglia. In addition, from the therapeutic point of view, we focused on sub-picomolar concentration of SN for further mechanistic studies. We found that 10(-14) M of SN failed to protect DA neurons against MPP+-induced toxicity in the absence of microglia. More importantly, SN failed to show a protective effect in neuron-glia cultures from mice lacking functional NADPH oxidase (PHOX), a key enzyme for extracellular superoxide production in immune cells. Furthermore, we demonstrated that SN reduced LPS-induced extracellular ROS production through the inhibition of the PHOX cytosolic subunit p47phoxtranslocation to the cell membrane. CONCLUSION: Our findings strongly suggest that the protective effects of SN are most likely mediated through the inhibition of microglial PHOX activity. These findings suggest a novel therapy to treat inflammation-mediated neurodegenerative diseases.


Subject(s)
Encephalitis/drug therapy , Microglia/drug effects , Morphinans/pharmacology , NADPH Oxidases/antagonists & inhibitors , Neurons/drug effects , Animals , Animals, Newborn , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Death/drug effects , Cell Death/physiology , Cell Line , Coculture Techniques , Dopamine/metabolism , Dose-Response Relationship, Drug , Encephalitis/enzymology , Encephalitis/physiopathology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/enzymology , Morphinans/agonists , Morphinans/chemistry , Morphinans/therapeutic use , NADPH Oxidases/metabolism , Neurons/enzymology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/enzymology , Parkinson Disease/physiopathology , Rats , Rats, Inbred F344 , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
16.
J Immunol ; 179(2): 1198-209, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617613

ABSTRACT

Recent studies have shown that morphine modulates the function of glia cells through both opioid receptor dependent and independent mechanisms. However, the mechanism by which morphine regulates neuronal disorders through the alteration of microglia activity remains unclear. In this study, using rat primary mesencephalic neuron-glia cultures, we report that both l-morphine and its synthetic stereoenantiomer, d-morphine, an ineffective opioid receptor agonist, significantly reduced LPS- or 1-methyl-4-phenylpyridinium-induced dopaminergic neurotoxicity with similar efficacy, indicating a nonopioid receptor-mediated effect. In addition, using reconstituted neuron and glia cultures, subpicomolar concentrations of morphine were found to be neuroprotective only in the presence of microglia, and significantly inhibited the production of inflammatory mediators from LPS-stimulated microglia cells. Mechanistic studies showed that both l- and d- morphine failed to protect dopaminergic neurons in cultures from NADPH oxidase (PHOX) knockout mice and significantly reduced LPS-induced PHOX cytosolic subunit p47(phox) translocation to the cell membrane by inhibiting ERK phosphorylation. Taken together, our results demonstrate that morphine, even at subpicomolar concentrations, exerts potent anti-inflammatory and neuroprotective effects either through the inhibition of direct microglial activation by LPS or through the inhibition of reactive microgliosis elicited by 1-methyl-4-phenylpyridinium. Furthermore, our study reveals that inhibition of PHOX is a novel site of action for the mu-opioid receptor-independent effect of morphine.


Subject(s)
Microglia/drug effects , Morphine/pharmacology , NADPH Oxidases/metabolism , Neuroprotective Agents/pharmacology , Receptors, Opioid/metabolism , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Blotting, Western , Cells, Cultured , Coculture Techniques , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Herbicides/toxicity , Immunohistochemistry , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Microglia/enzymology , Microglia/pathology , Microscopy, Confocal , NADPH Oxidases/drug effects , Narcotics/pharmacology , Neurons/drug effects , Rats , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stereoisomerism
17.
Cell Signal ; 19(2): 251-60, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16996249

ABSTRACT

Activation of the beta(2) adrenergic receptor (beta(2)AR) located on macrophages has been reported to possess anti-inflammatory properties, inhibiting nuclear factor kappaB (NF-kappaB) activation and cytokine production induced by pro-inflammatory stimuli. Here, we show that activation of the beta(2)AR in the absence of pro-inflammatory stimuli produced up to an 80- and 8-fold increase in IL-1beta and IL-6 transcripts, respectively, in the RAW 264.7 murine macrophage cell line. This increase in mRNA expression was accompanied by a significant increase in IL-1beta and IL-6 protein production. Pre-treatment of RAW cells with pharmacological inhibitors of protein kinase A (PKA) or NF-kappaB pathway failed to block this cytokine increase. Instead, the beta(2)AR-mediated increase in cytokines required activation of both the B-raf-ERK1/2 and p38 pathways. Treatment of RAW cells with the exchange protein directly activated by cAMP (EPAC) agonist also resulted in the up-regulation of IL-1beta and IL-6 transcripts. Examination of the main transcription factors downstream of the ERK1/2 and p38 signaling revealed that beta(2)AR activation resulted in the stimulation of CRE-, but not C/EBPbeta-, ETS-, or NF-kappaB-dependent transcription. Western blot analysis further showed that among the transcription factors which recognize the CRE-binding site, ATF-1 and ATF-2 but not CREB proteins were phosphorylated in an ERK1/2- and p38-dependent manner. Collectively, these results demonstrate that beta(2)ARs possess pro-inflammatory properties and that their activation leads to IL-1beta and IL-6 production through ERK1/2- and p38-dependent activation of ATF-1 and ATF-2 transcription factors.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Receptors, Adrenergic, beta-2/metabolism , Activating Transcription Factors/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Cell Line , Cytokines/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Proto-Oncogene Proteins B-raf/metabolism , RNA, Messenger/metabolism , Signal Transduction , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Parkinsonism Relat Disord ; 13 Suppl 3: S316-20, 2007.
Article in English | MEDLINE | ID: mdl-18267257

ABSTRACT

In this paper we report that diphenyliodonium (DPI), a NADPH oxidase inhibitor, shows potent anti-inflammatory and neuroprotective effects at femtomolar concentrations (10(-13) to 10(-14) M) in primary midbrain cultures. Mechanistic studies revealed that DPI-elicited effects were mediated by the inhibition of LPS-induced microglial ROS production and the subsequent release of pro-inflammatory cytokine TNFa, and the production of nitric oxide. Further studies showed that 10(-14) M DPI significantly reduced LPS-induced ERK phosphorylation. Taken together, our results demonstrate that femtomolar concentrations of DPI exert potent anti-inflammatory and neuroprotective effects by inhibiting microglial activation through the inhibition of ERK-regulated PHOX activity.


Subject(s)
Biphenyl Compounds/pharmacology , Microglia/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Onium Compounds/pharmacology , Analysis of Variance , Animals , Brain/cytology , Cells, Cultured , Dopamine/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Microglia/physiology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neurons/cytology , Neurons/metabolism , Nitrites/metabolism , Pregnancy , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Biomaterials ; 27(30): 5170-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16808973

ABSTRACT

A role for monocyte/macrophage modulation of wound healing at endosseous implants is proposed. The modification of the endosseous implant surface topography can alter cell adhesion and resultant cell behavior. The aim of this study was to define the effect of increased cpTitanium surface topography on adherent J744A.1 macrophage phenotype in culture. The J744A.1 cells were cultured on 20mm diameter cpTitanium disks prepared with smooth and grit-blasted/acid rough surface topographies for 24-72 h. Following culture in growth media with or without lipopolysaccharide (LPS), total RNA was isolated and real-time polymerase chain reaction (PCR) was used to measure the steady-state levels of the pro-inflammatory cytokines interleukin 1-beta (IL-1beta) and interleukin 6 (IL-6) and the anti-inflammatory cytokine interleukin-10 (IL-10). Additional evidence of pro-inflammatory signaling was sought by measurement of cellular nitric oxide (NO) production. In the absence of LPS, IL-1beta levels were increased on grit-blasted/acid rough surfaces during the first 48 h. In contrast, IL-6 levels were reduced on the grit-blasted/acid rough surfaces. When cultures were treated with LPS, high levels of IL-1beta and IL-6 expression were measured, irrespective of surface topography. The responses of J744A.1 cells to surface and superimposed LPS stimulation suggest only modest effects of the modeled endosseous implant surface on adherent cell pro-inflammatory cytokine expression and NO signaling.


Subject(s)
Cytokines/metabolism , Macrophages/drug effects , Nitric Oxide/metabolism , Titanium/pharmacology , Wound Healing/drug effects , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Cell Adhesion , Cells, Cultured , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Surface Properties , Transforming Growth Factor beta/metabolism
20.
J Pharmacol Exp Ther ; 319(1): 44-52, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16807359

ABSTRACT

The role of anti-inflammatory cytokines in Parkinson's disease is not completely understood. In this study, using mesencephalic neuron-glia cultures, we report that both pretreatment and post-treatment of rat mesencephalic neuron-glia cultures with interleukin (IL)-10, a natural immune modulator, reduced lipopolysaccharide (LPS)-induced DA neurotoxicity. The main purpose of this study was to elucidate the molecular mechanism underlying IL-10-elicited neuroprotection. IL-10 significantly inhibited LPS-induced production of tumor necrosis factor-alpha, nitric oxide, and extracellular superoxide in microglia cells. In addition, using reconstituted neuron and glia cell cultures, IL-10 was shown to be neuroprotective only in the presence of microglia. More importantly, IL-10 failed to protect DA neurons in cultures from mice lacking NADPH oxidase (PHOX), a key enzyme for extracellular superoxide production in immune cells, suggesting the critical role of PHOX in IL-10 neuroprotection. This conclusion was further supported by the finding that IL-10 inhibited LPS-induced translocation of the cytosolic subunit of NADPH oxidase p47(phox) to the membrane. When the Janus tyrosine kinase (JAK) 1 signaling pathway was blocked, IL-10 failed to attenuate LPS-induced superoxide production, indicating that the JAK1 signaling cascade mediates the inhibitory effect of IL-10. Together, our results suggest that IL-10 inhibits LPS-induced DA neurotoxicity through the inhibition of PHOX activity in a JAK1-dependent mechanism.


Subject(s)
Interleukin-10/pharmacology , Lipopolysaccharides/toxicity , Mesencephalon/drug effects , NADPH Oxidases/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Animals , Cells, Cultured , Janus Kinase 1 , Mice , Mice, Inbred C57BL , NADPH Oxidases/physiology , Protein Transport , Protein-Tyrosine Kinases/physiology , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Superoxides/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...