Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Neurobiol Aging ; 28(8): 1206-14, 2007 Aug.
Article in English | MEDLINE | ID: mdl-16837104

ABSTRACT

Loss of Locus coeruleus (LC) noradrenergic (NA) neurons occurs in several neurodegenerative conditions including Alzheimer's disease (AD). In vitro and in vivo studies have shown that NA influences several features of AD disease including inflammation, neurodegeneration, and cognitive function. In the current study we tested if LC loss influenced beta amyloid (Abeta) plaque deposition. LC neuronal degeneration was induced in transgenic mice expressing mutant V717F human amyloid precursor protein (APP) by treatment with the selective neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine DSP4 (5mg/kg every 2 weeks beginning at age 3 months). At 9 months of age, when control mice show low amyloid load, DSP4-treated mice showed an approximately 5-fold increase in the average number of Abeta plaques. This was accompanied by an increase in the levels of APP C-terminal cleavage fragments. DSP4-treatment increased both microglial and astroglial activation. In vivo, DSP4-treatment decreased expression and activity of the Abeta degrading enzyme neprilysin, while in vitro NA increased phagocytosis of Abeta1-42 by microglia. These findings suggest that noradrenergic innervation from LC are needed to maintain adequate Abeta clearance, and therefore that LC degeneration could contribute to AD pathogenesis.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain/pathology , Norepinephrine/deficiency , Plaque, Amyloid/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Newborn , Benzylamines/toxicity , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Isoproterenol/pharmacology , Locus Coeruleus/drug effects , Locus Coeruleus/injuries , Locus Coeruleus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Neprilysin/metabolism , Neurotransmitter Uptake Inhibitors/toxicity , Norepinephrine/pharmacology , Phagocytosis/drug effects , Plaque, Amyloid/drug effects
2.
J Neurochem ; 99(5): 1389-1402, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17074064

ABSTRACT

Brain abscesses arise from a focal parenchymal infection by various pathogens, particularly Staphylococcus aureus. We have shown that astrocytes are activated upon exposure to S. aureus and may contribute to the excessive tissue damage characteristic of brain abscess. Therefore, modulating astrocyte activation may facilitate a reduction in brain abscess severity. Peroxisome proliferator activated receptor-gamma (PPAR-gamma) agonists are potent inhibitors of microglial activation; however, the effects of these compounds on S. aureus-dependent astrocyte activation have not yet been examined. Here, we demonstrate that two chemically distinct PPAR-gamma agonists, 15-deoxy-delta12,14-prostaglandin J2 (15d-PGJ2) and ciglitazone, suppress the production of several pro-inflammatory molecules in S. aureus-stimulated astrocytes including interleukin-1beta and nitric oxide (NO). Interestingly, 15d-PGJ2 attenuated Toll-like receptor 2 (TLR2) and inducible nitric oxide synthase expression, but failed to modulate macrophage inflammatory protein-2 (MIP-2/CXCL2) production, suggesting that 15d-PGJ2 is not a global inhibitor of astrocyte activation. Another novel finding of this study was the fact that both 15d-PGJ2 and ciglitazone were capable of attenuating pre-existing astrocyte activation, indicating their potential benefit in a therapeutic setting. Importantly, 15d-PGJ2 and ciglitazone were still capable of inhibiting S. aureus-induced pro-inflammatory mediator release in PPAR-gamma-deficient astrocytes, supporting PPAR-gamma-independent effects of these compounds. Collectively, these results suggest that 15d-PGJ2 and ciglitazone exert their anti-inflammatory actions on astrocytes primarily independent of the PPAR-gamma pathway.


Subject(s)
Astrocytes/drug effects , PPAR gamma/agonists , Prostaglandin D2/analogs & derivatives , Thiazolidinediones/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/microbiology , Cells, Cultured , Hypoglycemic Agents/pharmacology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/metabolism , PPAR gamma/metabolism , Prostaglandin D2/pharmacology , Staphylococcus aureus/physiology , Toll-Like Receptor 2/drug effects , Toll-Like Receptor 2/metabolism
3.
Antioxid Redox Signal ; 8(5-6): 873-83, 2006.
Article in English | MEDLINE | ID: mdl-16771677

ABSTRACT

The neurotransmitter noradrenaline (NA) exerts important antiinflammatory effects on glial cells including suppression of the inducible form of nitric oxide synthase (NOS2). The authors examined the consequences of manipulating NA in vivo by treating adult rats with the neurotoxin DSP4, which selectively lesions noradrenergic neurons of the locus ceruleus (LC), and reduces cortical NA levels. Following LC lesion, intracortical injection of aggregated amyloid beta 1-42 (Abeta1-42) caused appearance of NOS2 within neurons, and increased neuronal damage assessed by staining for nonphosphorylated neurofilament proteins with antibody SMI-32. Co-treatment with a selective alpha2-adrenergic antagonist reduced neuronal NOS2 staining as well as SMI-32 staining. Neuronal damage was dependent on NOS2 expression since injection of Abeta1-42 into DSP4-treated NOS2-deficient mice did not result in neuronal damage. These results demonstrate that decrease of NA levels in vivo can exacerbate inflammatory responses and neuronal damage due to inflammatory stimuli such as Abeta. These findings suggest that alpha2-adrenergic antagonists could provide therapeutic benefit in neurological diseases such as AD or PD where LC loss is known to occur.


Subject(s)
Adrenergic alpha-Antagonists/metabolism , Amyloid beta-Peptides/metabolism , Nitric Oxide Synthase Type II/metabolism , Norepinephrine/metabolism , Peptide Fragments/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic Agents/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , Benzylamines/pharmacology , Cerebral Cortex/metabolism , Locus Coeruleus , Male , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Rats , Rats, Sprague-Dawley
4.
Antioxid Redox Signal ; 8(5-6): 885-92, 2006.
Article in English | MEDLINE | ID: mdl-16771678

ABSTRACT

The authors previously showed that conditioned media (CM) from activated microglia increased inducible nitric oxide synthase (NOS2) in cortical neurons. Here they examined the ability of noradrenaline (NA) to reduce neuronal NOS2 or cell death. Primary mouse cortical neurons were activated using CM from microglia incubated with lipopolysaccharide (LPS). Neuronal NOS2 was assessed by increases in nitrite accumulation, and increases in NOS2 mRNA levels and fluorescence of the NO-sensitive probe DAF-2 DA. NOS2 induction was associated with an increase in neuronal LDH release. When NA was added during microglial activation, neuronal NOS2 was significantly reduced (by approximately 70%); in contrast if NA was added to the neurons along with CM, there was less reduction (about 35% decrease) in NOS2 expression. NA added to either microglia or to neurons reduced neuronal LDH release comparably. Pretreatment of CM with blocking antibody to TNFalpha, alone or with IL1-receptor antagonist, partially reduced neuronal cell death and NOS2. Incubation of neurons with NA increased IkBalpha, which could reduce NOS2. These results demonstrate that NA modulates neuronal NOS2 expression and damage, and that these effects are primarily due to inhibition of microglia released factors. Perturbations of NA could exacerbate neuronal damage by allowing for increased inflammatory responses.


Subject(s)
Adrenergic alpha-Agonists/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type II/metabolism , Norepinephrine/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Culture Media, Conditioned/chemistry , I-kappa B Proteins/metabolism , Lipopolysaccharides/pharmacology , Mice , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , NF-KappaB Inhibitor alpha , Neurons/cytology , Nitric Oxide Synthase Type II/genetics , Nitrites/metabolism , Rats , Rats, Sprague-Dawley
5.
J Neuroimmunol ; 168(1-2): 65-75, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16098614

ABSTRACT

Agonists of the peroxisome proliferator-activated receptor gamma (PPARgamma) exert anti-inflammatory and anti-proliferative effects which led to testing of these drugs in experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis. In contrast, the effect of PPARdelta (PPARdelta) agonists in EAE is not yet known. We show that oral administration of the selective PPARdelta agonist GW0742 reduced clinical symptoms in C57BL/6 mice that had been immunized with encephalitogenic myelin oligodendrocyte glycoprotein (MOG) peptide. In contrast to previous results with PPARgamma agonists, GW0742 only modestly attenuated clinical symptoms when the drug was provided simultaneously with immunization, but a greater reduction was observed if administered during disease progression. Reduced clinical symptoms were accompanied by a reduction in the appearance of new cortical lesions, however cerebellar lesion load was not reduced. Treatment of T-cells with GW0742 either in vivo or in vitro did not reduce IFNgamma production; however GW0742 reduced astroglial and microglial inflammatory activation and IL-1beta levels in EAE brain. RTPCR analysis showed that GW0742 increased expression of some myelin genes. These data demonstrate that PPARdelta agonists, like other PPAR ligands, can exert protective actions in an autoimmune model of demyelinating disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , PPAR delta/agonists , Thiazoles/administration & dosage , Animals , Brain/cytology , Brain/drug effects , Brain/immunology , Brain/metabolism , Concanavalin A/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Enzyme-Linked Immunosorbent Assay/methods , Female , Gene Expression Regulation, Enzymologic/drug effects , Glycoproteins , Immunohistochemistry/methods , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Myelin-Oligodendrocyte Glycoprotein , Neuroglia/drug effects , Neuroglia/metabolism , Peptide Fragments , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Severity of Illness Index , Time Factors
6.
J Neurochem ; 92(5): 1140-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15715664

ABSTRACT

Brain inflammation is regulated by endogenous substances, including neurotransmitters such as noradrenaline (NA), which can increase anti-inflammatory genes. To identify NA-regulated, anti-inflammatory genes, we used TOGA (total gene expression analysis) to screen rat astrocyte-derived RNA. NA-inducible cDNA clone DST11 encodes an isoform of the complement C5a receptor (C5aR), with 39% identity at the amino acid level to the rat C5aR, and 56% identity to a recently described human C5aR variant termed C5L2 (complement 5a-like receptor). Quantitative PCR confirmed that in astrocytes, DST11 mRNA expression is increased by NA, whereas in vivo depletion of cortical NA reduced DST11 levels. Western blot analysis demonstrated basal and NA-induced expression of DST11 as a 45 kDa protein in primary astrocytes cultures. Immunocytochemical staining of adult rat brain revealed DST11-immunoreactivity throughout brain, co-localized to neurons and astrocytes. In astrocytes, induction of nitric oxide synthase type 2 was increased by treatment with antisense oligonucleotides to DST11. Reducing DST11 expression also increased nuclear factor kappaB reporter gene, and decreased cAMP response element reporter gene activation. These results demonstrate that DST11 is a C5aR isoform expressed by glia and neurons, which is regulated by NA, and exerts anti-inflammatory functions. Changes in DST11 levels in diseased brain could therefore contribute to the progression of inflammatory damage.


Subject(s)
Astrocytes/metabolism , Norepinephrine/pharmacology , Protein Isoforms/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Astrocytes/drug effects , Benzylamines/toxicity , Blotting, Western/methods , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Drug Interactions , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Glioma , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Immunohistochemistry/methods , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Luciferases/metabolism , Mice , Models, Biological , NF-kappa B/metabolism , Neurotransmitter Uptake Inhibitors/toxicity , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitrites/metabolism , Oligonucleotides, Antisense/pharmacology , Phosphopyruvate Hydratase/metabolism , Protein Isoforms/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Anaphylatoxin C5a/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcriptional Activation , Transfection/methods
7.
J Neuroinflammation ; 1(1): 9, 2004 Jun 30.
Article in English | MEDLINE | ID: mdl-15285793

ABSTRACT

BACKGROUND: Under pathological conditions, microglia produce proinflammatory mediators which contribute to neurologic damage, and whose levels can be modulated by endogenous factors including neurotransmitters such as norepinephrine (NE). We investigated the ability of NE to suppress microglial activation, in particular its effects on induction and activity of the inducible form of nitric oxide synthase (NOS2) and the possible role that IL-1beta plays in that response. METHODS: Rat cortical microglia were stimulated with bacterial lipopolysaccharide (LPS) to induce NOS2 expression (assessed by nitrite and nitrate accumulation, NO production, and NOS2 mRNA levels) and IL-1beta release (assessed by ELISA). Effects of NE were examined by co-incubating cells with different concentrations of NE, adrenergic receptor agonists and antagonists, cAMP analogs, and protein kinase (PK) A and adenylate cyclase (AC) inhibitors. Effects on the NFkappaB:IkappaB pathway were examined by using selective a NFkappaB inhibitor and measuring IkappaBalpha protein levels by western blots. A role for IL-1beta in NOS2 induction was tested by examining effects of caspase-1 inhibitors and using caspase-1 deficient cells. RESULTS: LPS caused a time-dependent increase in NOS2 mRNA levels and NO production; which was blocked by a selective NFkappaB inhibitor. NE dose-dependently reduced NOS2 expression and NO generation, via activation of beta2-adrenergic receptors (beta2-ARs), and reduced loss of inhibitory IkBalpha protein. NE effects were replicated by dibutyryl-cyclic AMP. However, co-incubation with either PKA or AC inhibitors did not reverse suppressive effects of NE, but instead reduced nitrite production. A role for IL-1beta was suggested since NE potently blocked microglial IL-1beta production. However, incubation with a caspase-1 inhibitor, which reduced IL-1beta levels, had no effect on NO production; incubation with IL-receptor antagonist had biphasic effects on nitrite production; and NE inhibited nitrite production in caspase-1 deficient microglia. CONCLUSIONS: NE reduces microglial NOS2 expression and IL-1beta production, however IL-1beta does not play a critical role in NOS2 induction nor in mediating NE suppressive effects. Changes in magnitude or kinetics of cAMP may modulate NOS2 induction as well as suppression by NE. These results suggest that dysregulation of the central cathecolaminergic system may contribute to detrimental inflammatory responses and brain damage in neurological disease or trauma.

8.
Am J Physiol Heart Circ Physiol ; 287(6): H2501-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15271668

ABSTRACT

No studies have specifically addressed whether cAMP can influence nitric oxide (NO)/cGMP-induced cerebral vasodilation. In this study, we examined whether cAMP can enhance or reduce NO-induced cerebral vasodilation in vivo via interfering with cGMP efflux or through potentiating phosphodiesterase 5 (PDE5)-mediated cGMP breakdown, respectively, in cerebral vascular smooth muscle cells (CVSMCs). To that end, we evaluated, in male rats, the effects of knockdown [via antisense oligodeoxynucleotide (ODN) applications] of the cGMP efflux protein multidrug resistance protein 5 (MRP5) and PDE5 inhibition on pial arteriolar NO donor [S-nitroso-N-acetyl penicillamine (SNAP)]-induced dilations in the absence and presence of cAMP elevations via forskolin. Pial arteriolar diameter changes were measured using well-established protocols in anesthetized rats. In control (missense ODN treated) rats, forskolin elicited a leftward shift in the SNAP dose-response curves (approximately 50% reduction in SNAP EC50). However, in MRP5 knockdown rats, cAMP increases were associated with a substantial reduction in SNAP-induced vasodilations (reflected as a significant 35-50% lower maximal response). In the presence of the PDE5 inhibitor MY-5445, the repression of the NO donor response accompanying forskolin was prevented. These findings suggest that cAMP has opposing effects on NO-stimulated cGMP increases. On the one hand, cAMP limits CVSMC cGMP loss by restricting cGMP efflux. On the other, cAMP appears to enhance PDE5-mediated cGMP breakdown. However, because increased endogenous cAMP seems to potentiate NO/cGMP-induced arteriolar relaxation when MRP5 expression is normal, the effect of cAMP to reduce cGMP efflux appears to predominate over cAMP stimulation of cGMP hydrolysis.


Subject(s)
Cerebrovascular Circulation/physiology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Vasodilation/physiology , 3',5'-Cyclic-GMP Phosphodiesterases , Animals , Arterioles/physiology , Colforsin/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 5 , Enzyme Activators/pharmacology , Hydrolysis , Indazoles/pharmacology , Male , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects
9.
Am J Physiol Heart Circ Physiol ; 286(5): H2020-7, 2004 May.
Article in English | MEDLINE | ID: mdl-14715514

ABSTRACT

Multidrug resistance protein 5 (MRP5) has been linked to cGMP cellular export in peripheral vascular smooth muscle cells (VSMCs) and is widely expressed in brain vascular tissue. In the present study, we examined whether knockdown of MRP5 in pial arterioles [via antisense oligodeoxynucleotide (ODN) applications] affected nitric oxide (NO)/cGMP-induced dilations. The antisense or (as a control) missense ODN was applied to the cortical surface approximately 24 h before study via closed cranial windows. The efficacy of the antisense vs. missense ODN in eliciting selective reductions in MRP5 expression was confirmed by analysis of MRP5 mRNA in pial tissue. Unexpectedly, in initial studies, a significantly lower maximal pial arteriolar diameter increase in the presence of the NO donor S-nitrosoacetylpenicillamine (SNAP) was seen in the antisense vs. missense ODN-treated rats (35 vs. 48% diameter increase, respectively). It was suspected that this related to a reduced vascular smooth muscle cell sensitivity to cGMP due to prolonged exposure to increased intracellular cGMP levels elevated by overnight restriction of cGMP efflux. That postulate was supported by a finding of a diminished vasodilating response to the cGMP-dependent protein kinase-activating cGMP analog 8-p-chlorophenylthio-cGMP in antisense vs. missense ODN-treated rats. To prevent desensitization, additional rats were studied in the presence of chronic NOS inhibition via Nomega-nitro-L-arginine. In the NO synthase (NOS)-inhibited rats, the maximal SNAP response was much higher in the antisense (62% increase) vs. the missense ODN (40% increase) group. A similar result was obtained when monitoring responses to the soluble guanylyl cyclase-activating drugs YC-1 and BAY 41-2272. Moreover, in the presence of NOS inhibition, the normal SNAP-induced rise in periarachnoid cerebrospinal fluid cGMP levels, which reflects cGMP efflux, was absent in the antisense ODN-treated rats, a finding consistent with loss of MRP5 function. In conclusion, if one minimizes the confounding effects of basal cGMP production, a clearer picture emerges, one that indicates an important role for MRP5-mediated cGMP efflux in the regulation of NO-induced cerebral arteriolar relaxation.


Subject(s)
Cyclic GMP/metabolism , Multidrug Resistance-Associated Proteins/physiology , Muscle, Smooth, Vascular/physiology , Pia Mater/blood supply , Vasodilation/physiology , Animals , Arachnoid , Arterioles/physiology , Carbon Dioxide/pharmacology , Cyclic GMP/cerebrospinal fluid , Enzyme Inhibitors/pharmacology , Male , Multidrug Resistance-Associated Proteins/genetics , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects
10.
J Neurochem ; 86(4): 907-16, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12887689

ABSTRACT

Cerebral inflammatory events play an important part in the pathogenesis of Alzheimer's disease (AD). Agonists of the peroxisome proliferator-activated receptor gamma (PPARgamma), a nuclear hormone receptor that mediates anti-inflammatory actions of non-steroidal anti-inflammatory drugs (NSAIDs) and thiazolidinediones, have been therefore proposed as a potential treatment of AD. Experimental evidence suggests that cortical noradrenaline (NA) depletion due to degeneration of the locus ceruleus (LC) - a pathological hallmark of AD - plays a permissive role in the development of inflammation in AD. To study a possible relationship between NA depletion and PPARgamma-mediated suppression of inflammation we investigated the influence of NA on PPARgamma expression in murine primary cortical astrocytes and neurons. Incubation of astrocytes and neurons with 100 micro m NA resulted in an increase of PPARgamma mRNA as well as PPARgamma protein levels in both cell types. These effects were blocked by the beta-adrenergic antagonist propranolol but not by the alpha-adrenergic antagonist phentolamine, suggesting that they might be mediated by beta-adrenergic receptors. Our results indicate for the first time that PPARgamma expression can be modulated by the cAMP signalling pathway, and suggest that the anti-inflammatory effects of NA on brain cells may be partly mediated by increasing PPARgamma levels. Conversely, decreased NA due to LC cell death in AD may reduce endogenous PPARgamma expression and therefore potentiate neuroinflammatory processes.


Subject(s)
Astrocytes/metabolism , Gene Expression/drug effects , Neurons/metabolism , Norepinephrine/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Alzheimer Disease/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Neurons/cytology , Neurons/drug effects , RNA, Messenger/metabolism , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Time Factors , Transcription Factors/genetics
11.
J Neurochem ; 85(2): 387-98, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12675915

ABSTRACT

The inflammatory responses in many cell types are reduced by noradrenaline (NA) binding to beta-adrenergic receptors. We previously demonstrated that cortical inflammatory responses to aggregated amyloid beta (Abeta) are increased if NA levels were first depleted by lesioning locus ceruleus (LC) noradrenergic neurons, which replicates the loss of LC occurring in Alzheimer's disease. To examine the molecular basis for increased responses, we used the selective neurotoxin DSP4 to lesion the LC, and then examined levels of putative anti-inflammatory molecules. Inflammatory responses were achieved by injection of aggregated Abeta1-42 peptide and IL-1beta into frontal cortex, which induced neuronal inducible nitric oxide synthase (iNOS) and microglial IL-1beta expression. DSP4-treatment reduced basal levels of nuclear factor kappa B (NF-kappaB) inhibitory IkappaB proteins, and of heat shock protein (HSP)70. Inflammatory responses were prevented by co-injection (ibuprofen or ciglitzaone) or oral administration (pioglitazone) of peroxisome proliferator-activated receptor gamma (PPARgamma) agonists. Treatment with PPARgamma agonists restored IkappaBalpha, IkappaBbeta, and HSP70 levels to values equal or above those observed in control animals, and reduced activation of cortical NF-kappaB. These results suggest that noradrenergic depletion reduces levels of anti-inflammatory molecules which normally limit cortical responses to Abeta, and that PPARgamma agonists can reverse that effect. These findings suggest one mechanism by which PPARgamma agonists could provide benefit in neurological diseases having an inflammatory component.


Subject(s)
Brain/metabolism , Encephalitis/metabolism , HSP70 Heat-Shock Proteins/metabolism , I-kappa B Proteins/metabolism , Norepinephrine/metabolism , Amyloid beta-Peptides , Animals , Benzylamines/pharmacology , Brain/drug effects , Brain/pathology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Encephalitis/chemically induced , Encephalitis/pathology , Hypoglycemic Agents/pharmacology , Interleukin-1 , Isoenzymes/antagonists & inhibitors , Locus Coeruleus/drug effects , Male , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Neurotoxins/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Peptide Fragments , Prostaglandin-Endoperoxide Synthases , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists
12.
J Biol Chem ; 278(8): 5828-36, 2003 Feb 21.
Article in English | MEDLINE | ID: mdl-12486128

ABSTRACT

Activation of peroxisome proliferator-activated receptors (PPARs) can regulate brain physiology and provide protection in models of neurological disease; however, neither their exact targets nor mechanisms of action in brain are known. In many cells, PPAR gamma agonists increase glucose uptake and metabolism. Because astrocytes store glucose and provide lactate to neurons on demand, we tested effects of PPAR gamma agonists on astroglial glucose metabolism. Incubation of cortical astrocytes with the PPAR gamma thiazolidinedione (TZD) agonist pioglitazone (Pio) significantly increased glucose consumption in a time- and dose-dependent manner, with maximal increase of 36% observed after 4 h in 30 microm Pio. Pio increased 2-deoxy-glucose uptake because of increased flux through the type 1 glucose transporter. However, at this time point Pio did not increase type 1 glucose transporter expression, nor were its effects blocked by transcriptional or translational inhibitors. Pio also increased astrocyte lactate production as soon as 3 h after incubation. These effects were replicated by other TZDs; however, the order of efficacy (troglitazone > pioglitazone > rosiglitazone) suggests that effects were not mediated via PPAR gamma activation. TZDs increased astrocyte cAMP levels, and their glucose modifying effects were reduced by protein kinase A inhibitors. TZDs inhibited state III respiration in isolated brain mitochondria, whereas in astrocytes they caused mitochondrial membrane hyperpolarization. Pio protected astrocytes against hypoglycemia-induced cell death. Finally, glucose uptake was modified in brain sections prepared from Pio-fed rats. These results demonstrate that TZDs modify astrocyte metabolism and mitochondrial function, which could be beneficial in neurological conditions where glucose availability is reduced.


Subject(s)
Astrocytes/metabolism , Carbazoles , Cerebral Cortex/metabolism , Glucose/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Thiazoles/agonists , Thiazoles/pharmacology , Thiazolidinediones , Transcription Factors/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glucose Transporter Type 1 , Glycolysis/drug effects , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Indoles/pharmacology , Kinetics , Lactates/metabolism , Monosaccharide Transport Proteins/drug effects , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Pioglitazone , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics
13.
Neurochem Int ; 41(5): 357-65, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12176079

ABSTRACT

It is now well accepted that inflammatory events contribute to the pathogenesis of numerous neurological disorders, including multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease, and AID's dementia. Whereas inflammation in the periphery is subject to rapid down regulation by increases in anti-inflammatory molecules and the presence of scavenging soluble cytokine receptors, the presence of an intact blood-brain barrier may limit a similar autoregulation from occurring in brain. Mechanisms intrinsic to the brain may provide additional immunomodulatory functions, and whose dysregulation could contribute to increased inflammation in disease. The findings that noradrenaline (NA) reduces cytokine expression in microglial, astroglial, and brain endothelial cells in vitro, and that modification of the noradrenergic signaling system occurs in some brain diseases having an inflammatory component, suggests that NA could act as an endogenous immunomodulator in brain. Furthermore, accumulating studies indicate that modification of the noradrenergic signaling system occurs in some neurodiseases. In this article, we will briefly review the evidence that NA can modulate inflammatory gene expression in vitro, summarize data supporting a similar immunomodulatory role in brain, and present recent data implicating a role for NA in attenuating the cortical inflammatory response to beta amyloid protein.


Subject(s)
Brain Chemistry/genetics , Brain Chemistry/physiology , Inflammation/genetics , Inflammation/pathology , Norepinephrine/physiology , Animals , Denervation , Humans , Immunosuppression Therapy , Plaque, Amyloid/pathology
14.
Ann Neurol ; 51(6): 694-702, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12112074

ABSTRACT

The development of clinical symptoms in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE) involves T-cell activation and migration into the central nervous system, production of glial-derived inflammatory molecules, and demyelination and axonal damage. Ligands of the peroxisome proliferator-activated receptor (PPAR) exert anti-inflammatory effects on glial cells, reduce proliferation and activation of T cells, and induce myelin gene expression. We demonstrate in two models of EAE that orally administered PPARgamma ligand pioglitazone reduced the incidence and severity of monophasic, chronic disease in C57BL/6 mice immunized with myelin oligodendrocyte glycoprotein peptide and of relapsing disease in B10.Pl mice immunized with myelin basic protein. Pioglitazone also reduced clinical signs when it was provided after disease onset. Clinical symptoms were reduced by two other PPARgamma agonists, suggesting a role for PPARgamma activation in protective effects. The suppression of clinical signs was paralleled by decreased lymphocyte infiltration, lessened demyelination, reduced chemokine and cytokine expression, and increased inhibitor of kappa B (IkB) expression in the brain. Pioglitazone also reduced the antigen-dependent interferon-gamma production from EAE-derived T cells. These results suggest that orally administered PPARgamma agonists could provide therapeutic benefit in demyelinating disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , I-kappa B Proteins , Receptors, Cytoplasmic and Nuclear/agonists , Thiazoles/therapeutic use , Thiazolidinediones , Transcription Factors/agonists , Tyrosine/analogs & derivatives , Animals , Cerebellum/cytology , Cerebellum/physiology , Cerebral Cortex/cytology , Cerebral Cortex/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Glycoproteins/administration & dosage , Glycoproteins/immunology , Humans , Hypoglycemic Agents/therapeutic use , Ligands , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Myelin-Oligodendrocyte Glycoprotein , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Oxazoles/therapeutic use , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Pioglitazone , Receptors, Cytoplasmic and Nuclear/immunology , Remission Induction , Spinal Cord/cytology , Spinal Cord/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription Factors/immunology , Tyrosine/therapeutic use
15.
J Biol Chem ; 277(33): 29662-8, 2002 Aug 16.
Article in English | MEDLINE | ID: mdl-12050158

ABSTRACT

The neurotransmitter norepinephrine (NE) can inhibit inflammatory gene expression in glial cells; however, the mechanisms involved are not clear. In primary astrocytes, NE dose-dependently increased the expression of inhibitory I kappa B alpha protein accompanied by an increase in steady state levels of I kappa B alpha mRNA. Maximal increases were observed at 30-60 min for the mRNA and at 4 h for protein, and these effects were mediated by NE binding to beta-adrenergic receptors. NE activated a 1.3-kilobase I kappa B alpha promoter transfected into astrocytes or C6 glioma cells, and this activation was prevented by a beta-antagonist and by protein kinase A inhibitors but not by an NF kappa B inhibitor. NE increased I kappa B alpha protein in both the cytosolic and the nuclear fractions, suggesting an increase in nuclear uptake of I kappa B alpha. I kappa B alpha was detected in the frontal cortex of normal adult rats, and its levels were reduced if central NE levels were depleted by lesion of the locus ceruleus. The reduction of brain I kappa B alpha levels was paralleled by increased inflammatory responses to lipopolysaccharide. These results demonstrate that I kappa B alpha expression is regulated by NE at both transcriptional and post-transcriptional levels, which could contribute to the observed anti-inflammatory properties of NE in vitro and in vivo.


Subject(s)
Astrocytes/drug effects , DNA-Binding Proteins/metabolism , I-kappa B Proteins , Norepinephrine/pharmacology , Animals , Astrocytes/metabolism , Base Sequence , Brain/drug effects , Brain/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA Primers , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , NF-KappaB Inhibitor alpha , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
16.
J Neurosci Res ; 67(4): 461-70, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11835313

ABSTRACT

The production of nitric oxide by the inflammatory isoform of nitric oxide synthase (NOS2) in brain glial cells is thought to contribute to the causes and development of neurological diseases and trauma. We previously demonstrated that activation of a heat shock response (HSR) by hyperthermia reduced NOS2 expression in vitro, and in vivo attenuated the clinical and histological symptoms of the demyelinating disease experimental autoimmune encephalomyelitis (EAE; Heneka et al. [2001] J. Neurochem. 77:568-579). Benzoquinoid ansamycins are fungal-derived antibiotics with tyrosine kinase inhibitory properties, and which also induce a HSR by allowing activation of HS transcription factor HSF1. We now show that two members of this class of drugs (geldanamycin and 17-allylamino-17-demethoxygeldanamycin) also induce a HSR in primary rat astrocytes and rat C6 glioma cells. Both drugs dose-dependently reduced nitrite accumulation, NOS2 steady-state mRNA levels, and the cytokine-dependent activation of a rat 2.2-kB NOS2 promoter construct stably expressed in C6 cells. These inhibitory effects were partially reversed by quercetin, a bioflavonoid which prevents HSF1 binding to DNA and thus attenuates the HSR. Ansamycins increased mRNA levels of the inhibitory IkappaBalpha protein, suggesting that inhibition of NFkappaB activation could contribute to their suppressive effects. Finally, in C57BL/6 mice actively immunized to develop EAE, a single injection of geldanamycin at 3 days after immunization reduced disease onset by over 50%. These results indicate that ansamycins can exert potent anti-inflammatory effects on brain glial cells which may provide therapeutic benefit in neuroinflammatory diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Astrocytes/drug effects , Encephalitis/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Heat-Shock Response/drug effects , I-kappa B Proteins , Nitric Oxide Synthase/metabolism , Rifabutin/analogs & derivatives , Rifabutin/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Astrocytes/enzymology , Benzoquinones , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Encephalitis/enzymology , Encephalitis/physiopathology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , HSP70 Heat-Shock Proteins/drug effects , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , Interferon-gamma/pharmacology , Lactams, Macrocyclic , Lipopolysaccharides/pharmacology , NF-KappaB Inhibitor alpha , Nitric Oxide/metabolism , Nitric Oxide Synthase/drug effects , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/metabolism , Quercetin/pharmacology , Quinones/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...