Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Calcium ; 49(6): 365-75, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21536328

ABSTRACT

In vitro studies show that microglia, the resident immune cells of the brain, express neurotransmitter and neuropeptide receptors which are linked to Ca(2+) signaling. Here we describe an approach to obtain Ca(2+) recordings from microglia in situ. We injected a retrovirus encoding a calcium sensor into the cortex of mice 2 days after stimulation of microglial proliferation by a stab wound injury. Microglial cells were identified with tomato lectin in acute slices prepared 3, 6, 21 and 42 days after the injury. The membrane current profile and the ameboid morphology indicated that microglial cells were activated at day 6 while at day 42 they resembled resting microglia. We recorded transient Ca(2+) responses to application of ATP, endothelin-1, substance P, histamine and serotonin. The fluorescence amplitude of ATP was increased only at day 6 compared to other time points, while responses to all other ligands did not vary. Only half of the microglial cells that responded to ATP also responded to endothelin-1, serotonin and histamine. Substance P, in contrast, showed a complete overlap with the ATP responding microglial population at day 6, at day 42 this population was reduced to 55%. Cultured cells were less responsive to these ligands. This study shows that in situ microglia consist of heterogeneous populations with respect to their sensitivity to neuropeptides and -transmitters.


Subject(s)
Brain/cytology , Calcium/metabolism , Macrophages/metabolism , Microglia/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Endothelin-1/pharmacology , Histamine/pharmacology , Membrane Potentials , Mice , Microglia/physiology , Patch-Clamp Techniques , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Retroviridae/genetics , Serotonin/pharmacology , Substance P/pharmacology , Transduction, Genetic
2.
Glia ; 59(1): 119-31, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20967887

ABSTRACT

Microglia activation is central to the neuroinflammation associated with neurological and neurodegenerative diseases, particularly because activated microglia are often a source of proinflammatory cytokines. Despite decade-long research, the molecular cascade of proinflammatory transformation of microglia in vivo remains largely elusive. Here, we report increased ß-catenin expression, a central intracellular component of WNT signaling, in microglia undergoing a proinflammatory morphogenic transformation under pathogenic conditions associated with neuroinflammation such as Alzheimer's disease. We substantiate disease-associated ß-catenin signaling in microglia in vivo by showing age-dependent ß-catenin accumulation in mice with Alzheimer's-like pathology (APdE9). In cultured mouse microglia expressing the WNT receptors Frizzled FZD(4,5,7,8) and LDL receptor-related protein 5/6 (LRP5/6), we find that WNT-3A can stabilize ß-catenin. WNT-3A dose dependently induces LRP6 phosphorylation with downstream activation of disheveled, ß-catenin stabilization, and nuclear import. Gene-expression profiling reveals that WNT-3A stimulation specifically increases the expression of proinflammatory immune response genes in microglia and exacerbates the release of de novo IL-6, IL-12, and tumor necrosis factor α. In summary, our data suggest that the WNT family of lipoglycoproteins can instruct proinflammatory microglia transformation and emphasize the pathogenic significance of ß-catenin-signaling networks in this cell type.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Inflammation/metabolism , Microglia/metabolism , Signal Transduction/physiology , Wnt Proteins/metabolism , Analysis of Variance , Animals , Blotting, Western , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Gene Expression Profiling , Humans , Immunohistochemistry , Inflammation/genetics , Mice , Microglia/drug effects , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Wnt Proteins/genetics , Wnt Proteins/pharmacology , beta Catenin/genetics , beta Catenin/metabolism
3.
J Neurochem ; 112(5): 1261-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20002522

ABSTRACT

Intense neuronal activity in the sensory retina is associated with a volume increase of neuronal cells (Uckermann et al., J. Neurosci. 2004, 24:10149) and a decrease in the osmolarity of the extracellular space fluid (Dmitriev et al., Vis. Neurosci. 1999, 16:1157). Here, we show the existence of an endogenous purinergic mechanism that prevents hypoosmotic swelling of retinal glial (Müller) cells in mice. In contrast to the cells from wild-type mice, hypoosmotic stress induced rapid swelling of glial cell somata in retinal slices from mice deficient in P2Y(1), adenosine A(1) receptors, or ecto-5'-nucleotidase (CD73). Consistently, glial cell bodies in retinal slices from wild-type mice displayed osmotic swelling when P2Y(1) or A(1) receptors, or CD73, were pharmacologically blocked. Exogenous ATP, UTP, and UDP inhibited glial swelling in retinal slices, while the swelling of isolated glial cells was prevented by ATP but not by UTP or UDP, suggesting that uracil nucleotides indirectly regulate the glial cell volume via activation of neuronal P2Y(4/6) and neuron-to-glia signaling. It is suggested that autocrine/paracrine activation of purinergic receptors and enzymes is crucially involved in the regulation of the glial cell volume.


Subject(s)
Cell Size , Neuroglia/cytology , Osmosis , Receptors, Purinergic/metabolism , Retina/cytology , Signal Transduction/physiology , 5'-Nucleotidase/deficiency , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Barium Compounds/metabolism , Calcium/metabolism , Chlorides/metabolism , Cyclic AMP/metabolism , Drug Combinations , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate Receptors/deficiency , Mice , Mice, Knockout , Neuroglia/drug effects , Neurons/drug effects , Neurons/metabolism , Osmolar Concentration , Potassium Channel Blockers/pharmacology , Purinergic Agonists , Purinergic Antagonists , Pyrimidine Nucleotides/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, Purinergic/deficiency , Signal Transduction/drug effects , Signal Transduction/genetics , Thionucleotides/pharmacology , Time Factors , Valerates/pharmacology , Xanthines/pharmacology
4.
Mol Vis ; 15: 1858-67, 2009 Sep 12.
Article in English | MEDLINE | ID: mdl-19756184

ABSTRACT

PURPOSE: Osmotic swelling of Müller glial cells has been suggested to contribute to retinal edema. We determined the role of adenosine signaling in the inhibition of Müller cell swelling in the murine retina. METHODS: The size of Müller cell somata was recorded before and during perfusion of retinal sections and isolated Müller cells with a hypoosmolar solution. Retinal tissues were freshly isolated from wild-type mice and mice deficient in A(1) adenosine receptors (A(1)AR(-/-)), or cultured as whole-mounts for three days. The potassium conductance of Müller cells was recorded in isolated cells, and retinal slices were immunostained against Kir4.1. RESULTS: Hypotonic exposure for 4 min induced a swelling of Müller cell bodies in retinal slices from A(1)AR(-/-) mice but not wild-type mice. Pharmacological inhibition of A(1) receptors or of the ecto-5'-nucleotidase induced hypoosmotic swelling of Müller cells from wild-type mice. Exogenous adenosine prevented the swelling of Müller cells from wild-type but not A(1)AR(-/-) mice. The antiinflammatory corticosteroid, triamcinolone acetonide, inhibited the swelling of Müller cells from wild-type mice; this effect was blocked by an antagonist of A(1) receptors. The potassium conductance of Müller cells and the Kir4.1 immunolabeling of retinal slices were not different between A(1)AR(-/-) and wild-type mice, both in freshly isolated tissues and retinal organ cultures. CONCLUSIONS: The data suggest that autocrine activation of A(1) receptors by extracellularly generated adenosine mediates the volume homeostasis of Müller cells in the murine retina. The swelling-inhibitory effect of triamcinolone is mediated by enhancement of endogenous adenosine signaling.


Subject(s)
Neuroglia/metabolism , Osmosis , Receptors, Purinergic P1/metabolism , Retina/cytology , Adenosine/metabolism , Adenosine/pharmacology , Animals , Immunohistochemistry , Ion Channel Gating/drug effects , Mice , Models, Biological , Neuroglia/drug effects , Organ Culture Techniques , Osmosis/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Purinergic P1/deficiency , Signal Transduction/drug effects
5.
Exp Neurol ; 216(2): 321-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19146854

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists (thiazolidinediones) have anti-inflammatory effects and improve endothelium function. Here, we analyzed the effects of pioglitazone on short- and longer-term outcome after mild transient brain ischemia. 129/SV mice were subjected to 30 min filamentous middle cerebral artery occlusion (MCAo), followed by reperfusion. Post event, animals were treated with daily intraperitoneal (i.p.) pioglitazone (20 mg/kg body weight) or vehicle. Pioglitazone given acutely after transient brain ischemia/reperfusion reduced lesion size and the number of Iba1-expressing microglia in the ischemic striatum at three days. In vitro, pioglitazone attenuated migration and proliferation of primary mouse microglia. However, analysis at 6 weeks after MCAo/reperfusion no longer yielded an effect of pioglitazone on either lesion size or Iba1+ cell counts. Regarding functional longer-term outcome, we also did not detect a beneficial effect of pioglitazone on motor function measured either on the pole test or the wire hanging test or on learning and memory in the Morris water maze. Our study thus underscores the importance of extending experimental stroke studies to an analysis of longer-term outcome.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Thiazolidinediones/therapeutic use , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Calcium-Binding Proteins/metabolism , Cell Count/methods , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Corpus Striatum/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Maze Learning/drug effects , Mice , Microfilament Proteins , Microglia/drug effects , Motor Activity/drug effects , Neurons/metabolism , Pioglitazone , Reaction Time/drug effects , Reperfusion Injury/prevention & control , Time Factors
6.
J Neurosci Res ; 87(3): 644-52, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18831010

ABSTRACT

Microglia, central nervous system (CNS) resident phagocytic cells, persistently police the integrity of CNS tissue and respond to any kind of damage or pathophysiological changes. These cells sense and rapidly respond to danger and inflammatory signals by changing their cell morphology; by release of cytokines, chemokines, or nitric oxide; and by changing their MHC expression profile. We have shown previously that microglial biosynthesis of the complement subcomponent C1q may serve as a reliable marker of microglial activation ranging from undetectable levels of C1q biosynthesis in resting microglia to abundant C1q expression in activated, nonramified microglia. In this study, we demonstrate that cultured microglial cells respond to extrinsic C1q with a marked intracellular Ca(2+) increase. A shift toward proinflammatory microglial activation is indicated by the release of interleukin-6, tumor necrosis factor-alpha, and nitric oxide and the oxidative burst in rat primary microglial cells, an activation and differentiation process similar to the proinflammatory response of microglia to exposure to lipopolysaccharide. Our findings indicate 1) that extrinsic plasma C1q is involved in the initiation of microglial activation in the course of CNS diseases with blood-brain barrier impairment and 2) that C1q synthesized and released by activated microglia is likely to contribute in an autocrine/paracrine way to maintain and balance microglial activation in the diseased CNS tissue.


Subject(s)
Complement C1q/pharmacology , Microglia/metabolism , Animals , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Interleukin-6/metabolism , Mannose-Binding Lectin/pharmacology , Microglia/drug effects , Nitric Oxide/metabolism , Phagocytosis/drug effects , Rats , Rats, Wistar , Respiratory Burst/drug effects , Tumor Necrosis Factor-alpha/metabolism
7.
Mol Cell Neurosci ; 39(4): 579-85, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18804537

ABSTRACT

Integrins are heterodimeric transmembrane proteins, which mediate cell-cell and cell-extracellular matrix (ECM) interaction. We show, that an inhibitor of alpha5 beta1 integrin (alpha5beta1), JSM6427, attenuated glioma growth and decreased the density of microglia at the tumor border. 21 days after glioma cell injection into an experimental mouse model, the tumor volume was significantly smaller after treating animals for 14 days with JSM6427 as compared to controls. We could demonstrate the expression of integrin alpha5beta1 on both microglia and glioma cells using flow cytometry. In a slice culture we could compare glioma growth in the presence and absence of microglia. Slices injected with glioma cells were treated with the integrin inhibitor JSM6427 and showed a significant reduction in tumor size as compared to control. Depleting microglial cells from the slice culture by treatment with clodronate liposomes abrogated the effect of JSM6427 on glioma invasion indicating that the presence of microglia is required. We show further, that microglial migration, and proliferation was attenuated dose-dependently by JSM6427.


Subject(s)
Glioma , Integrin alpha5beta1/antagonists & inhibitors , Aminopyridines/metabolism , Animals , Cell Movement/physiology , Cells, Cultured , Cytokines/metabolism , Glioma/metabolism , Glioma/pathology , Integrin alpha5beta1/metabolism , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/metabolism , Neoplasm Transplantation , Propionates/metabolism , Tissue Culture Techniques
8.
Glia ; 56(3): 331-41, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18098126

ABSTRACT

Microglia is activated by brain injury. They migrate in response to ATP and although adenosine alone has no effect on wild type microglial migration, we show that inhibition of adenosine receptors impedes ATP triggered migration. CD39 is the dominant cellular ectonucleotidase that degrades nucleotides to nucleosides, including adenosine. Importantly, ATP fails to stimulate P2 receptor mediated migration in cd39(-/-) microglia. However, the effects of ATP on migration in cd39(-/-) microglia can be restored by co-stimulation with adenosine or by addition of a soluble ectonucleotidase. We also tested the impact of cd39-deletion in a model of ischemia, in an entorhinal cortex lesion and in the facial nucleus after facial nerve lesion. The accumulation of microglia at the pathological sites was markedly decreased in cd39(-/-) animals. We conclude that the co-stimulation of purinergic and adenosine receptors is a requirement for microglial migration and that the expression of cd39 controls the ATP/adenosine balance.


Subject(s)
Antigens, CD/physiology , Apyrase/physiology , Cell Movement/genetics , Microglia/physiology , Receptors, Purinergic P2/physiology , Adenine Nucleotides/pharmacology , Animals , Animals, Newborn , Apyrase/deficiency , Apyrase/pharmacology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Movement/drug effects , Cells, Cultured , Cytokines/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Entorhinal Cortex/pathology , Facial Nerve Diseases/metabolism , Facial Nerve Diseases/pathology , Facial Nerve Diseases/physiopathology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Microglia/ultrastructure , Patch-Clamp Techniques/methods , Receptors, Purinergic P1/deficiency
9.
J Neurosci ; 27(48): 13065-73, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-18045900

ABSTRACT

Bradykinin (BK) is produced and acts at the site of injury and inflammation. In the CNS, migration of microglia toward the lesion site plays an important role pathologically. In the present study, we investigated the effect of BK on microglial migration. Increased motility of cultured microglia was mimicked by B1 receptor agonists and markedly inhibited by a B1 antagonist but not by a B2 receptor antagonist. BK induced chemotaxis in microglia isolated from wild-type and B2-knock-out mice but not from B1-knock-out mice. BK-induced motility was not blocked by pertussis toxin but was blocked by chelating intracellular Ca2+ or by low extracellular Ca2+, implying that Ca2+ influx is prerequisite. Blocking the reverse mode of Na+/Ca2+ exchanger (NCX) completely inhibited BK-induced migration. The involvement of NCX was further confirmed by using NCX+/- mice; B1-agonist-induced motility and chemotaxis was decreased compared with that in NCX+/+ mice. Activation of NCX seemed to be dependent on protein kinase C and phosphoinositide 3-kinase, and resultant activation of intermediate-conductance (IK-type) Ca2+-dependent K+ currents (I(K(Ca))) was activated. Despite these effects, BK did not activate microglia, as judged from OX6 staining. Using in vivo lesion models and pharmacological injection to the brain, it was shown that microglial accumulation around the lesion was also dependent on B1 receptors and I(K(Ca)). These observations support the view that BK functions as a chemoattractant by using the distinct signal pathways in the brain and, thus, attracts microglia to the lesion site in vivo.


Subject(s)
Bradykinin/pharmacology , Calcium/metabolism , Cell Movement/drug effects , Microglia/drug effects , Receptor, Bradykinin B1/physiology , Sodium-Calcium Exchanger/physiology , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Bradykinin/analogs & derivatives , Bradykinin B1 Receptor Antagonists , Bradykinin B2 Receptor Antagonists , Cells, Cultured , Cerebellar Cortex/cytology , Chemotaxis/drug effects , Dose-Response Relationship, Drug , Mice , Mice, Knockout , Rats , Rats, Wistar , Receptor, Bradykinin B1/agonists , Receptor, Bradykinin B1/deficiency , Receptor, Bradykinin B2/agonists , Receptor, Bradykinin B2/deficiency , Signal Transduction/drug effects , Time Factors
10.
Mol Cell Neurosci ; 33(4): 401-11, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17055293

ABSTRACT

Activation of microglia by LPS leads to an induction of cytokine and NO release, reduced proliferation and increased outward K(+) conductance, the latter involving the activation of Kv1.5 and Kv1.3 channels. We studied the role of these channels for microglial function using two strategies to interfere with channel expression, a Kv1.5 knockout (Kv1.5(-/-)) mouse and an antisense oligonucleotide (AO) approach. The LPS-induced NO release was reduced by AO Kv1.5 and completely absent in the Kv1.5(-/-) animal; the AO Kv1.3 had no effect. In contrast, proliferation was augmented with both, loss of Kv1.3 or Kv1.5 channel expression. After facial nerve lesion, proliferation rate was higher in Kv1.5(-/-) animals as compared to wild type. Patch clamp experiments confirmed the reduction of the LPS-induced outward current amplitude in Kv1.5(-/-) microglia as well as in Kv1.5- or Kv1.3 AO-treated cells. Our study indicates that induction of K(+) channel expression is a prerequisite for the full functional spectrum of microglial activation.


Subject(s)
Kv1.3 Potassium Channel/physiology , Kv1.5 Potassium Channel/physiology , Microglia/physiology , Animals , Brain/cytology , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Embryo, Mammalian , Facial Nerve Diseases/metabolism , Facial Nerve Diseases/pathology , Immunohistochemistry/methods , Kv1.3 Potassium Channel/chemistry , Kv1.5 Potassium Channel/chemistry , Kv1.5 Potassium Channel/genetics , Lipopolysaccharides/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Nitric Oxide/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Patch-Clamp Techniques/methods , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
11.
Cancer Res ; 66(17): 8550-7, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951167

ABSTRACT

We report that experimental glioblastoma grow more vigorously in A(1) adenosine receptor (A(1)AR)-deficient mice associated with a strong accumulation of microglial cells at and around the tumors. A(1)ARs were prominently expressed in microglia associated with tumor cells as revealed with immunocytochemistry but low in microglia in the unaffected brain tissue. The A(1)AR could also be detected on microglia from human glioblastoma resections. To study functional interactions between tumor and host cells, we studied glioblastoma growth in organotypical brain slice cultures. A(1)AR agonists suppressed tumor growth. When, however, microglial cells were depleted from the slices, the agonists even stimulated tumor growth. Thus, adenosine attenuates glioblastoma growth acting via A(1)AR in microglia.


Subject(s)
Brain Neoplasms/pathology , Cell Communication/physiology , Glioblastoma/pathology , Microglia/cytology , Receptor, Adenosine A1/physiology , Adenosine/pharmacology , Animals , Cell Communication/drug effects , Cell Division , Cells, Cultured , Genes, Reporter , Genotype , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/physiology , Receptor, Adenosine A1/deficiency , Receptor, Adenosine A1/genetics
12.
Glia ; 54(7): 656-665, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17006894

ABSTRACT

In this review we summarize mechanisms of Ca(2+) signaling in microglial cells and the impact of Ca(2+) signaling and Ca(2+) levels on microglial function. So far, Ca(2+) signaling has been only characterized in cultured microglia and thus these data refer rather to activated microglia as observed in pathology when compared with the resting form found under physiological conditions. Purinergic receptors are the most prominently expressed ligand-gated Ca(2+)-permeable channels in microglia and control several microglial functions such as cytokine release in a Ca(2+)-dependent fashion. A large variety of metabotropic receptors are linked to Ca(2+) release from intracellular stores. Depletion of these intracellular stores triggers a capacitative Ca(2+) entry. While microglia are already in an activated state in culture, they can be further activated, for example, by exposure to bacterial endotoxin. This activation leads to a chronic increase of [Ca(2+)](i) and this Ca(2+) increase is a prerequisite for the release of nitric oxide and cytokines. Moreover, several factors (TNFalpha, IL-1beta, and IFN-gamma) regulate resting [Ca(2+)](i) levels.


Subject(s)
Calcium Signaling/physiology , Cell Communication/physiology , Central Nervous System/metabolism , Microglia/metabolism , Animals , Calcium Channels/metabolism , Central Nervous System/cytology , Cytokines/metabolism , Gliosis/immunology , Gliosis/metabolism , Microglia/cytology , Receptors, Purinergic/metabolism , Second Messenger Systems/physiology
13.
Pflugers Arch ; 452(5): 615-21, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16791619

ABSTRACT

Microglial cells are considered as the pathologic sensors of the brain. In this paper, we review mechanisms of purinergic signaling in microglia. As ATP is not only considered as a physiological signaling substance but is also elevated in pathology, it is not surprising that microglia express a variety of P2X, P2Y and adenosin receptors. As a rapid physiological event, ATP triggers a cationic conductance, increases the potassium conductance and also elicits a calcium response. As a long-term effect, purinergic receptor activation is linked to the movement of microglial processes and, in the context of pathology, to chemotaxis. The purinoreceptors also modulate the release of substances from microglia, such as cytokines, nitric oxide, or superoxide, which are important in the context of a pathologic response.


Subject(s)
Adenosine Triphosphate/metabolism , Brain/physiology , Calcium/metabolism , Ion Channel Gating/physiology , Microglia/physiology , Receptors, Purinergic P2/metabolism , Signal Transduction/physiology , Animals , Humans , Microglia/cytology , Models, Neurological
14.
Mol Cell Neurosci ; 29(1): 128-38, 2005 May.
Article in English | MEDLINE | ID: mdl-15866053

ABSTRACT

Microglial cells are the immune-competent elements of the brain. They not only express receptors for chemokines and cytokines but also for neurotransmitters such as GABA [Charles et al., Mol. Cell Neurosci. 24 (2003) 214], glutamate [Noda et al., J. Neurosci. 20 (2000) 251], and adrenaline [Mori et al., Neuropharmacology 43 (2002) 1026]. Here we report the functional expression of dopamine receptors in mouse and rat microglia, in culture and brain slices. Using the patch clamp technique as the functional assay we identified D1- and D2-like dopamine receptors using subtype-specific ligands. They triggered the inhibition of the constitutive potassium inward rectifier and activated potassium outward currents in a subpopulation of microglia. Chronic dopamine receptor stimulation enhanced migratory activity and attenuated the lipopolysaccharide (LPS)-induced nitric oxide (NO) release similar as by stimulation of adrenergic receptors. While, however, noradrenaline attenuated the LPS-induced release of TNF-alpha and IL-6, dopamine was ineffective in modulating this response. We conclude that microglia express dopamine receptors which are distinct in function from adrenergic receptors.


Subject(s)
Dopamine/pharmacology , Microglia/drug effects , Microglia/physiology , Norepinephrine/pharmacology , Sympathomimetics/pharmacology , Adrenergic Agonists/pharmacology , Animals , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Membrane Potentials/drug effects , Mice , Mice, Inbred Strains , Microglia/cytology , Nitric Oxide/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Potassium/metabolism , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/physiology
15.
Brain Res Brain Res Rev ; 48(2): 133-43, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15850652

ABSTRACT

Microglial cells in culture and in situ express a defined pattern of K(+) channels, which is distinct from that of other glial cells and neurons. This pattern undergoes defined changes with microglial activation. As expected for a cell with immunological properties, microglia express a variety of cytokine and chemokine receptors, which are linked to the mobilization of Ca(2+) (cytosolic free calcium) from internal stores. Microglial cells also have the capacity to respond to neuronal activity: they express receptors for the major excitatory receptor glutamate and the main inhibitory receptor GABA (gamma-amino butyric acid). By expressing purinergic receptors, microglia can sense astrocyte activity in the form of Ca(2+) waves. Activation of transmitter receptors can affect cytokine release which is a potential means as to how brain activity can affect immune function.


Subject(s)
Brain/cytology , Microglia/physiology , Animals , Calcium/metabolism , Cytokines/metabolism , Humans , Immune System/physiology , Neuroglia , Neurons/physiology , Potassium Channels/physiology , Receptors, GABA/metabolism , Receptors, Glutamate/metabolism , Receptors, Purinergic/metabolism
16.
Mol Cell Neurosci ; 25(2): 312-22, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15019947

ABSTRACT

gamma-Aminobutyric acid (GABA) can act as a neuroprotective agent besides its well-established role as the main inhibitory neurotransmitter in the CNS. Here we report that microglial cells express GABA(B) receptors indicating that these prominent immunocompetent cells in the brain are a target for GABA. Agonists of GABA(B) receptors triggered the induction of K(+) conductance in microglial cells from acute brain slices and in culture. Both subunits of GABA(B) receptors were identified in cultured microglia by Western blot analysis and immunocytochemistry, and were detected on a subpopulation of microglia in situ by immunohistochemistry. In response to facial nerve axotomy, we observed an increase in GABA(B) receptor expressing microglial cells in the facial nucleus. We activated microglial cells in culture with lipopolysaccharide (LPS) to induce the release of interleukin-6 and interleukin-12p40. This release activity was attenuated by simultaneous activation of the GABA(B) receptors indicating that GABA can modulate the microglial immune response.


Subject(s)
Central Nervous System/metabolism , Interleukins/metabolism , Microglia/metabolism , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Axotomy , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/immunology , Facial Nerve Injuries/immunology , Facial Nerve Injuries/pathology , Facial Nerve Injuries/physiopathology , GABA Agonists/pharmacology , GABA-B Receptor Agonists , Gliosis/immunology , Gliosis/metabolism , Gliosis/physiopathology , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-12 Subunit p40 , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukins/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Potassium Channels/drug effects , Potassium Channels/metabolism , Protein Subunits/immunology , Protein Subunits/metabolism , Rats , Rats, Wistar
17.
Eur J Neurosci ; 17(11): 2267-76, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12814360

ABSTRACT

Microglial cells are the pathologic sensors in the brain. ATP released from damaged cells is a candidate for signalling neural injury to microglia. Moreover, ATP is an extracellular messenger for propagating astrocyte activity in the form of Ca2+ waves. To test for the functional expression of purinoreceptors in microglial cells we employed the patch-clamp technique in acute slices of adult mouse brain. ATP triggered a nonselective cationic and a K+ current. Pharmacological screening with purinergic ligands indicated the presence of P2Y1 and P2Y2/4 receptors linked to the activation of a K+ current and P2X receptors, including P2X7, linked to the activation of a nonselective cationic current. These findings suggest that microglial cells in situ express different purinergic receptors with distinct sensitivity and functional coupling. To test for the involvement of purinoreceptors in microglial activation, we stimulated cultured microglial cells with lipopolysaccharide and measured the release of tumour necrosis factor alpha, interleukin-6, interleukin-12 and macrophage inflammatory protein 1alpha, induction of K+ outward currents and nitric oxide release. All these parameters were reduced in the presence of purinergic ligands, indicating that purinergic receptor activation attenuated indicators of microglial activation.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Brain/physiology , Microglia/physiology , Receptors, Purinergic/physiology , Adenosine Triphosphate/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Chloride Channels/drug effects , Dose-Response Relationship, Drug , Drug Interactions , In Vitro Techniques , Lipopolysaccharides/pharmacology , Magnesium/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Neural Conduction/drug effects , Nitric Oxide/metabolism , Patch-Clamp Techniques/methods , Potassium Channels/drug effects , Receptors, Purinergic/classification , Receptors, Purinergic/genetics , Time Factors , Uridine Diphosphate/pharmacology , Uridine Triphosphate/pharmacology , Vasodilator Agents/pharmacology
18.
Phytochemistry ; 62(3): 491-500, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12620362

ABSTRACT

The biosynthesis of benzophenanthridine alkaloids, phytoalexins of Eschscholzia californica, in cultured cells can be induced by a glycoprotein preparation from yeast, methyljasmonate, artificial acidification with permeant acids, or mild osmotic stress. Each of these stimuli strongly attenuated the subsequent response to the same stimulus (homologous desensitization). Elicitor contact and artificial acidification mutually desensitized the cells for either signal. In contrast, elicitor-treated cells maintained their responsiveness to methyljasmonate or hyperosmolarity (sorbitol). Elicitor concentrations that nearly saturated the alkaloid response did not cause a detectable increase of jasmonate content. Transient acidification of the cytoplasm is a necessary step of signaling by low elicitor concentrations but was not detectable after jasmonate treatment. Seen together, the data indicate the existence of a jasmonate-dependent and jasmonate-independent (Delta pH controlled) signal pathway towards the expression of benzophenanthridine biosynthesis. Selective desensitization allows either stimulus to activate a distinct share of the biosynthetic capacity of the cell and limits the accumulation of toxic defense metabolites.


Subject(s)
Acetates/pharmacology , Alkaloids/biosynthesis , Cells, Cultured/drug effects , Cells, Cultured/physiology , Cyclopentanes/pharmacology , Eschscholzia/physiology , Phenanthridines/metabolism , Plant Growth Regulators/pharmacology , Cells, Cultured/cytology , Dose-Response Relationship, Drug , Eschscholzia/cytology , Eschscholzia/drug effects , Eschscholzia/metabolism , Gene Expression Regulation, Plant/drug effects , Gene Expression Regulation, Plant/physiology , Hydrogen-Ion Concentration , Inflammation Mediators/pharmacology , Microscopy, Confocal/methods , Oxylipins , Pentanoic Acids/pharmacology , Phenanthridines/chemistry , Saccharomyces cerevisiae/chemistry , Signal Transduction/drug effects , Signal Transduction/physiology , Sorbitol/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...