Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 7: 30, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20482831

ABSTRACT

BACKGROUND: Brain inflammation plays a central role in multiple sclerosis (MS). Dimethylfumarate (DMF), the main ingredient of an oral formulation of fumaric acid esters with proven therapeutic efficacy in psoriasis, has recently been found to ameliorate the course of relapsing-remitting MS. Glial cells are the effector cells of neuroinflammation; however, little is known of the effect of DMF on microglia and astrocytes. The purpose of this study was to use an established in vitro model of brain inflammation to determine if DMF modulates the release of neurotoxic molecules from microglia and astrocytes, thus inhibiting glial inflammation. METHODS: Primary microglial and astrocytic cell cultures were prepared from cerebral cortices of neonatal rats. The control cells were treated with LPS, an accepted inducer of pro-inflammatory properties in glial cells, and the experimental groups with LPS and DMF in different concentrations. After stimulation/incubation, the generation of nitric oxide (NO) in the cell culture supernatants was determined by measuring nitrite accumulation in the medium using Griess reagent. After 6 hours of treatment RT-PCR was used to determine transcription levels of iNOS, IL-1beta, IL-6 and TNF-alpha mRNA in microglial and astrocytic cell cultures initially treated with DMF, followed after 30 min by LPS treatment. Moreover, we investigated possible involvement of the ERK and Nrf-2 transduction pathway in microglia using western blot analysis. RESULTS: Pretreatment with DMF decreased synthesis of the proinflammatory mediators iNOS, TNF-alpha, IL-1beta and IL-6 at the RNA level in activated microglia and astrocytes in vitro, associated with a decrease in ERK phosphorylation in microglia. CONCLUSIONS: Collectively, these results suggest that the neuroprotective effects of DMF may be in part functionally attributable to the compound's ability to inhibit expression of multiple neuroinflammatory mediators in brain of MS patients.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Astrocytes , Encephalomyelitis, Autoimmune, Experimental , Fumarates , Inflammation , Microglia , Animals , Rats , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Blotting, Western , Brain/pathology , Dimethyl Fumarate , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fumarates/pharmacology , Inflammation/prevention & control , Interleukin-1beta/antagonists & inhibitors , Interleukin-6/antagonists & inhibitors , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitrites/metabolism , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics
2.
J Neurosci Res ; 88(1): 16-23, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19681171

ABSTRACT

Previous studies have shown that activin A, a neuroprotective cytokine and dimeric polypeptide composed of two betaA subunits, is elevated in the cerebrospinal fluid of patients suffering from bacterial meningitis. In this study, to elucidate further the functional significance and pathophysiological implications of these findings, we demonstrated that microglial cells are not only the source but also the target cells of activin A in the central nervous system: immunohistochemistry and RT-PCR revealed expression of activin subunit betaA mRNA as well as activin receptor type I and type II mRNA in rat microglia in vitro. Further studies showed that activin enhances microglial proliferation and decreases the gamma-interferon-induced synthesis of nitric oxide, one of several microglial mediators involved in the inflammatory response in microglia activation. Furthermore, quantitative RT-PCR, Western blotting, and ELISA showed an inhibitory effect of activin on inducible nitric oxide synthase, tumor necrosis factor-alpha, interleukin-6, and interleukin-1beta gene and protein levels after lipopolysaccharide treatment. We suggest that the increased synthesis of activin A is directly involved, via influence on microglia cell functions, in the modulation of the inflammatory response in bacterial meningitis.


Subject(s)
Activins/biosynthesis , Meningitis, Bacterial/metabolism , Microglia/metabolism , Activin Receptors/genetics , Activin Receptors/metabolism , Activins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Analysis of Variance , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Male , Meningitis, Bacterial/genetics , Meningitis, Viral/genetics , Meningitis, Viral/metabolism , Microglia/drug effects , Middle Aged , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
3.
J Neuroimmunol ; 212(1-2): 106-11, 2009 Jul 25.
Article in English | MEDLINE | ID: mdl-19464738

ABSTRACT

Erythropoietin is a hematopoietic cytokine which is also produced in the brain under hypoxia. Since this pathology is associated with glial cell activation and release of cytotoxic molecules, we investigated the expression of EPO receptors (EPO-R) and effects of erythropoietin on microglial cell functions in vitro using RT-PCR, Western immunoblotting, nitric oxide measurement, tumor necrosis factor-alpha-(TNF-alpha)-ELISA and gel shift assay analyses. Furthermore, we examined if erythropoietin could modulate proliferation of microglia. As shown by reverse transcription-polymerase chain reaction and immunocytochemistry, rat microglial cells and the murine microglia cell line BV-2 express the EPO-R. However, EPO showed no effect on the release of the proinflammatory mediators' nitric oxide and TNF-alpha. Moreover, EPO was not able to reduce the LPS (lipopolysaccharide) stimulated translocation of the proinflammatory transcription factor NF-kappaB into the nucleus of murine microglia, but induced (3)H-thymidine incorporation into DNA of microglial cells. These results show that microglia are target cells for erythropoietin which possesses mitogenic, but not anti-inflammatory effects on microglia. Therefore, the well-documented neuroprotective effects of erythropoietin could not be ascribed to an anti-inflammatory effect on microglia.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/biosynthesis , Erythropoietin/pharmacology , Microglia/drug effects , Neuroprotective Agents/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Mice , Microglia/immunology , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Rats , Rats, Wistar , Receptors, Erythropoietin/analysis , Tumor Necrosis Factor-alpha/biosynthesis
4.
Acta Neuropathol ; 116(1): 47-55, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18343932

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by a selective loss of dopaminergic neurons in the substantia nigra (SN). It has been suggested that microglial inflammation augments the progression of PD. Neuromelanin (NM), a complex polymer pigment found in catecholaminergic neurons, has sparked interest because of the suggestion that NM is involved in cell death in Parkinson's disease, possibly via microglia activation. To further investigate the possible role of NM in the pathogenesis of PD, we conducted in vivo experiments to find out whether microglial cells become activated after injection of human neuromelanin (NM) into (1) the cerebral cortex or (2) the substantia nigra to monitor in this PD-relevant model both microglial activation and possible neurodegeneration. In this study, adult male Wistar rats received an intracerebral injection of either NM, bacterial lipopolysaccharide (LPS, positive control), phosphate-buffered saline (PBS, negative control) or colloidal gold suspension (negative particular control). After different survival times (1, 8 or 12 weeks), brain slices from the cerebral cortex or substantia nigra (SN, 1 week) were stained with Iba-1 and/or GFAP antibody to monitor microglial and astrocytic reaction, and with tyrosine hydroxylase (TH) to monitor dopaminergic cell survival (SN group only). The injection of LPS induced a strong inflammatory response in the cortex as well in the substantia nigra. Similar results could be obtained after NM injection, while the injection of PBS or gold suspension showed only moderate or no glial activation. However, the inflammatory response declined during the time course. In the SN group, there was, apart from strong microglia activation, a significant dopaminergic cell loss after 1 week of survival time. Our findings clearly indicate that extracellular NM could be one of the key molecules leading to microglial activation and neuronal cell death in the substantia nigra. This may be highly relevant to the elucidation of therapeutic strategies in PD.


Subject(s)
Inflammation/immunology , Melanins/immunology , Microglia/immunology , Nerve Degeneration/pathology , Parkinsonian Disorders/pathology , Substantia Nigra/pathology , Animals , Cell Death , Humans , Immunohistochemistry , Inflammation/etiology , Inflammation/pathology , Male , Nerve Degeneration/immunology , Neurons/pathology , Parkinsonian Disorders/immunology , Rats , Rats, Wistar , Substantia Nigra/immunology , Tyrosine 3-Monooxygenase/metabolism
5.
J Neurochem ; 101(3): 718-28, 2007 May.
Article in English | MEDLINE | ID: mdl-17448144

ABSTRACT

Recent studies suggest that the formyl-peptide-receptor-like-1 (FPRL1) plays an essential role in inflammatory responses in the host defence mechanisms and neurodegenerative disorders. Furthermore, it may be involved in proinflammatory processes of prion diseases. However, little is known about the induction and regulation of PrP106-126-induced receptor endocytosis. We have thus analysed whether PrP106-126 increases the activity of phospholipase D (PLD) via FPRL1, an enzyme involved in the regulation of the secretion, endocytosis and receptor signalling, in glial cells. PLD activity was determined using a transphosphatidylation assay and the internalization of PrP106-126, and FPRL1 was assessed by fluorescence microscopy and quantified by ELISA. We could show that PLD is activated by PrP106-126 both in astrocytes and microglia, and moreover that PrP106-126 is rapidly internalized via FPRL1 in astrocytes and microglia cells. The determination of receptor activity by extracellular signal-regulated kinases 1/2 phosphorylation and cAMP level measurement verified the PrP106-126-induced activation of FPRL1. FPRL1-mediated PrP106-126 uptake was blocked by the receptor antagonist chenodeoxycholic acid. These studies indicate the involvement of FPRL1-mediated cellular signalling in PrP106-126-endocytosis and may allow the development of therapeutic agents interfering with prion uptake and/or PLD function, using either PLD or the FPRL1 as a possible pharmaceutical target.


Subject(s)
Astrocytes/drug effects , Peptide Fragments/metabolism , Prions/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Animals , Animals, Newborn , Brain/cytology , Cells, Cultured , Clathrin/metabolism , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Humans , Peptide Fragments/pharmacology , Phospholipase D/metabolism , Prions/pharmacology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors
6.
Brain Res ; 1129(1): 43-52, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17169340

ABSTRACT

In the diseased central nervous system, astrogliosis is accompanied by microglial activation. Depending on the context of their activation, reactive astrocytes are involved in neuronal survival and regeneration in an either protective or impedimental way. Major reactive changes of astrocytes in vivo are the upregulation of the intermediate filaments GFAP (glial fibrillary acidic protein) and vimentin with accompanying cellular hypertrophy and/or hyperplasia. To examine the involvement of activated microglia in the onset and maintenance of astrogliosis, we used an in vitro model of purified cultures of astrocytes and assessed as parameters for astrogliosis GFAP, vimentin, astroglial hypertrophy and cell growth after treatment with medium conditioned by LPS (lipopolysaccarides)-stimulated microglia. Furthermore, IL-6 as a typically upregulated cytokine in proinflammatory processes in the brain was determined in treated astrocytes. GFAP, the classical marker for astrogliosis, was downregulated on its protein and in parallel with vimentin on its mRNA level. The expression of actin, another cytoskeleton protein used as control, remained unchanged. Ultrastructural studies of astroglial intermediate filaments supported these findings. No hypertrophy was found. Nevertheless, LPS-activated microglia stimulated astrocytes as demonstrated by an increased cell number and an enhanced mRNA expression of IL-6. Resting microglia did not change any of the determined parameters. Our results suggest that the role of activated microglia in astrogliotic processes following injury of the brain has to be reevaluated, as microglia in their activated state might support the onset of astrogliosis on the one hand, but might delay or reduce subsequent glial scar formation on the other hand.


Subject(s)
Astrocytes/metabolism , Brain Diseases/metabolism , Gliosis/metabolism , Microglia/metabolism , Animals , Animals, Newborn , Astrocytes/ultrastructure , Biomarkers/metabolism , Brain Diseases/physiopathology , Cell Communication/physiology , Cell Enlargement , Cells, Cultured , Culture Media, Conditioned/pharmacology , Down-Regulation/physiology , Encephalitis/chemically induced , Encephalitis/metabolism , Encephalitis/physiopathology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Gliosis/physiopathology , Interleukin-6/genetics , Interleukin-6/metabolism , Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Lipopolysaccharides/pharmacology , Microglia/ultrastructure , Microscopy, Electron, Transmission , RNA, Messenger/metabolism , Rats , Rats, Wistar , Vimentin/genetics , Vimentin/metabolism
7.
Toxicol Appl Pharmacol ; 204(1): 36-45, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15781292

ABSTRACT

Microglia participates in most acute and chronic neuropathologies and its activation appears to involve interactions with neurons and other glial cells. Trimethyltin (TMT)-induced brain damage is a well-characterized model of neurodegeneration, in which microglial activation occurs before neuronal degeneration. The aim of this in vitro study was to investigate the role of astroglia in TMT-induced microgliosis by using nitric oxide (NO), inducible NO synthase (iNOS), and morphological changes as parameters for microglial activation. Our investigation discusses (a) whether microglial cells can be activated directly by TMT; (b) if astroglial cells are capable of triggering or modulating microglial activation; (c) how the morphology and survival of microglia and astrocytes are affected by TMT treatment; and (d) whether microglial-astroglial interactions depend on direct cell contact or on soluble factors. Our results show that microglia are more vulnerable to TMT than astrocytes are and cannot be activated directly by TMT with regard to the examined parameters. In bilayer coculture with viable astroglial cells, microglia produce NO in significant amounts at subcytotoxic concentrations of TMT (20 micromol/l). At these TMT concentrations, microglial cells in coculture convert into small round cells without cell processes, whereas flat, fibroblast-like astrocytes convert into thin process bearing stellate cells with a dense and compact cell body. We conclude that astrocytes trigger microglial activation after treatment with TMT, although the mechanisms of this interaction remain unknown.


Subject(s)
Astrocytes/drug effects , Cell Communication/drug effects , Microglia/drug effects , Trimethyltin Compounds/toxicity , Animals , Astrocytes/pathology , Astrocytes/physiology , Cells, Cultured , Coculture Techniques , Hydrogen Peroxide , Lipopolysaccharides , Microglia/metabolism , Microglia/pathology , Microscopy, Phase-Contrast , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Proteins/analysis , Rats , Rats, Wistar , Tetrazolium Salts , Thiazoles
8.
Brain Res Dev Brain Res ; 144(2): 191-9, 2003 Sep 10.
Article in English | MEDLINE | ID: mdl-12935916

ABSTRACT

Astrogliosis is a common phenomenon seen in most neuropathological changes of the central nervous system. Several in vitro models have been used to study the mechanisms and conditions for the induction of astrogliosis, however many do not take into account that the metabolic and structural characteristics of astrocytes change with time in culture. Thus, it appears difficult to attribute changes of, e.g., GFAP to the normal change in vitro as opposed to additional changes due to an astrogliotic reaction. The present study was therefore undertaken to characterize these developmental changes in purified astroglial secondary cultures during cultivation to provide a basis for further investigations of astrogliosis in vitro. During 6 weeks of cultivation (3-43 days) GFAP (ELISA) increased much more (22-fold) than the cell number (2.5-fold) and the total protein (3.5-fold). The GFAP/protein ratio increased during the first 4 weeks of cultivation and reached a plateau thereafter, which was accompanied by a significant increase of GFAP mRNA (Northern blot). At the ultrastructural level (transmission electron microscopy) gliofilaments in the perinuclear region as well as in the cell processes of 4-day-old astrocytes showed a dispersed pattern, whereas an accumulation of gliofilaments was found in 39-day-old cells, which formed large aggregated bundles localized mostly in the cell processes. Our results show that in vitro astrocytes undergo developmental changes in their accumulation of GFAP and intermediate filaments which reach a stable steady state after 4 weeks in culture. These 'normal' developmental changes will have to be taken into account, when experiments with variations of the level of GFAP are performed. Stable culture conditions for experimentation appear to be present after 4 weeks in culture.


Subject(s)
Astrocytes/physiology , Cerebellum/metabolism , Glial Fibrillary Acidic Protein/metabolism , Gliosis , Animals , Animals, Newborn , Astrocytes/ultrastructure , Blotting, Northern , Cell Count , Cell Division , Cells, Cultured , Cerebellum/growth & development , Cerebellum/ultrastructure , Colorimetry , Enzyme-Linked Immunosorbent Assay , Glial Fibrillary Acidic Protein/genetics , Gliosis/genetics , Gliosis/metabolism , Gliosis/pathology , Gliosis/physiopathology , Microscopy, Electron , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
9.
J Neurotrauma ; 20(3): 297-307, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12820684

ABSTRACT

Immunophilins belong to the large family of peptidyl-prolyl-cis-trans-isomerases known to be involved in many cellular processes (e.g., protein trafficking and transcriptional regulation). Beside the widespread therapeutic use of ligands of immunophilins as immunosuppressants, it has been shown that some of these compounds such as FK506 and V-10,367 may mediate neuroprotection and improve axonal regeneration following damage to peripheral nerve fibers. Here, we have analyzed the effects of these two compounds on neurite outgrowth of retinal explants in vitro and on axonal regeneration of retinal ganglion cells, a population of central intrinsic neurons, ten days following optic nerve crush in vivo. FK506 enhanced neurite outgrowth/regrowth in vitro in a dose dependent manner up to 135% (control = 100%), while V-10,367 was more effective (up to 168%). In vivo, intravitreal V-10,367 and FK506 significantly reduced the number of dying retinal ganglion cells as demonstrated by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling. Local application of FK506 into the vitreous body, but not V-10,367, immediately provided after the optic nerve crush induced the elongation of regenerating fibers across the lesion site for around 1.2 mm. Our data provide evidence that the ligands of the FK506-binding proteins FK506 and V-10,367 protect (otherwise dying) retinal ganglion cells from optic nerve crush-induced cell death, promote neurite outgrowth in vitro and that locally applied FK506 enhances the sprouting of axotomized central intrinsic neurons such as retinal ganglion cells in vivo after optic nerve crush.


Subject(s)
Neurites/drug effects , Neuroprotective Agents/pharmacology , Pyridines/pharmacology , Retinal Ganglion Cells/drug effects , Tacrolimus/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Cyclohexanols/pharmacology , Dose-Response Relationship, Drug , GAP-43 Protein/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Nerve Crush , Nerve Regeneration/drug effects , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , Pyrans/pharmacology , Rats , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Tacrolimus/administration & dosage
10.
FASEB J ; 17(3): 500-2, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12631585

ABSTRACT

It has been suggested that microglial inflammation augments the progression of Parkinson's disease (PD). However, endogenous factors initiating microglial activation are largely unknown. We therefore investigated the effects of human neuromelanin (NM) on the release of neurotoxic mediators and the underlying signaling pathways from rat microglia in vitro. The addition of NM to microglial cultures induced positive chemotactic effects, activated the proinflammatory transcription factor nuclear factor kappaB (NF-kappaB) via phosphorylation and degradation of the inhibitor protein kappaB (IkappaB), and led to an up-regulation of tumor necrosis factor alpha, interleukin-6, and nitric oxide. The impairment of NF-kappaB function by the IkappaB kinase inhibitor sulfasalazine was paralleled by a decline in neurotoxic mediators. NM also activated p38 mitogen-activated protein kinase (MAPK), the inhibition of this pathway by SB203580 diminished phosphorylation of the transactivation domain of the p65 subunit of NF-kappaB. These findings demonstrate a crucial role of NM in the pathogenesis of PD by augmentation of microglial activation, leading to a vicious cycle of neuronal death, exposure of additional neuromelanin, and chronification of inflammation. The antagonization of microglial activation by a pharmacological intervention targeting microglial NF-kappaB or p38 MAPK could point to additional venues in the treatment of PD.


Subject(s)
MAP Kinase Signaling System , Melanins/pharmacology , Microglia/physiology , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Animals , Cells, Cultured , Chemotaxis , Humans , I-kappa B Proteins/metabolism , Inflammation/etiology , Inflammation Mediators/metabolism , Microglia/cytology , Microglia/drug effects , Microglia/enzymology , NF-KappaB Inhibitor alpha , Parkinson Disease/immunology , Phosphorylation , Rats , p38 Mitogen-Activated Protein Kinases
11.
Cell Tissue Res ; 308(2): 167-75, 2002 May.
Article in English | MEDLINE | ID: mdl-12037575

ABSTRACT

(-)-Deprenyl ( L-deprenyl, selegiline hydrochloride), a selective monoamine oxidase B (MAO-B) inhibitor employed in the pharmacological therapy of Parkinson's disease, increases neuronal survival in both animal models of neurodegenerative disorders and acute CNS lesions. Despite intensive investigations, the mechanisms of (-)-deprenyl-mediated neuroprotection remain poorly understood. To test the hypothesis that (-)-deprenyl might have a beneficial effect not only on neuronal survival, but also on axonal regeneration, we describe here experiments performed in vitro and in vivo which clearly demonstrate that (-)-deprenyl fails to promote axonal regeneration of severed rat retinal ganglion cells (RGCs). Furthermore, (-)-deprenyl was not able to overcome free-radical-induced RGC axon degeneration. These results challenge the notion that (-)-deprenyl might be useful as a monotherapy for acute CNS lesions and give rise to a more critical viewpoint of the trophic-like function of this widely used therapeutic agent.


Subject(s)
Axons/drug effects , Molsidomine/analogs & derivatives , Monoamine Oxidase Inhibitors/pharmacology , Nerve Regeneration/drug effects , Neuroprotective Agents/pharmacology , Retinal Ganglion Cells/drug effects , Selegiline/pharmacology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Survival/drug effects , Free Radicals/toxicity , GAP-43 Protein/biosynthesis , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/growth & development , Gene Expression Regulation/drug effects , Genes, bcl-2/genetics , Immunohistochemistry , In Vitro Techniques , Male , Molsidomine/pharmacology , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Neurites/drug effects , Optic Nerve/cytology , Optic Nerve/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar , Receptor, trkA/biosynthesis , Receptor, trkB/biosynthesis
12.
Invest Ophthalmol Vis Sci ; 43(6): 2021-6, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12037014

ABSTRACT

PURPOSE: Recent studies have reported neuroprotective effects of erythropoietin (EPO) and vascular endothelial growth factor (VEGF). The purpose of the present study was to clarify their influence on neurite outgrowth and regeneration of rat retinal ganglion cells (RGCs) in vitro and to elucidate the expression of corresponding receptors in the rat retina in vivo. METHODS: Retinal explants from postnatal rats were stimulated with VEGF alone; VEGF in combination with anti-VEGF-receptor (VEGF-R)-2 antibody or T-type Ca2+ channel blocker ethosuximide (ESX); EPO alone; or EPO in combination with anti-EPO-receptor antibody or ESX. The presence of the corresponding receptors in the rat retina was assessed by reverse transcription-polymerase chain reaction (RT-PCR) and by immunohistochemistry. RESULTS: EPO induced a stable improvement of neurite outgrowth of RGCs in a dose-dependent manner (5 x 10(-15) M to 5 x 10(-13) M) up to 169% (P < 0.05). Treatment of the explants with anti-EPO-R antibody (1:80 dilution) and with ESX (5 microM) totally inhibited EPO-mediated effects on RGCs. In comparison, VEGF (50 ng/mL), induced neurite outgrowth of retina explants up to 167% (P < 0.05), which again was inhibited in the presence of anti-VEGF-R2 antibody or ESX. Transcripts of EPO-R, VEGF-R1, and VEGF-R2 were detected by RT-PCR. Intense immunoreactivity for VEGF-R1, VEGF-R2, and EPO-R were found in the RGC layer of the retina. CONCLUSIONS: The data demonstrate for the first time that EPO and VEGF have a significant and specific biological effect on neurite regrowth of axotomized RGCs. Therefore, these results imply that EPO and VEGF have not only a neuroprotective but also a neuroregenerative role in ischemic retinal conditions.


Subject(s)
Endothelial Growth Factors/pharmacology , Erythropoietin/pharmacology , Lymphokines/pharmacology , Neurites/physiology , Retinal Ganglion Cells/drug effects , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Drug Combinations , Ethosuximide/pharmacology , Female , Immunohistochemistry , Male , Nerve Regeneration/drug effects , Proto-Oncogene Proteins/metabolism , Rats , Rats, Inbred F344 , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Erythropoietin/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Retinal Ganglion Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
14.
CNS Drugs ; 16(3): 145-53, 2002.
Article in English | MEDLINE | ID: mdl-11888335

ABSTRACT

In addition to the systemic renin-angiotensin system (RAS), a local RAS has been identified. Recent research has focused on this latter system and has investigated the effects of locally generated angiotensin II, especially in the kidney, heart and CNS. In the mammalian brain, all components of the RAS are present including angiotensin AT(1) and AT(2) receptor subtypes. While the AT(1) receptor is responsible for the classical effects of angiotensin II, it has been found that the AT(2) receptor displays totally different signalling mechanisms and this has revealed hitherto unknown functions of angiotensin II. AT(2) receptors are expressed at low density in many healthy adult tissues, but are up-regulated in pathological circumstances, e.g. stroke or nerve lesion. Evidence has now emerged that the actions of angiotensin II that are exerted via the AT(2) receptor are directly opposed to those mediated by the AT(1 )receptor. For example, the AT(2) receptor has antiproliferative properties and therefore opposes the growth-promoting effect linked to AT(1) receptor stimulation. It has been reported that the AT(2) receptor regulates several functions of nerve cells, e.g. ionic fluxes, cell differentiation and axonal regeneration, but also modulates programmed cell death. It is possible that a more extensive knowledge of the AT(2) receptor could contribute to the understanding of the clinically beneficial effects of AT(1) receptor antagonists, as this treatment may unmask AT(2) receptor activity. This review presents selected aspects of advances in AT(2) receptor pharmacology, molecular biology and signal transduction with particular reference to possible novel therapeutic options for CNS diseases.


Subject(s)
Angiotensin II/metabolism , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Receptors, Angiotensin/physiology , Angiotensin II/pharmacology , Angiotensin II/therapeutic use , Angiotensin Receptor Antagonists , Animals , Humans , Ligands , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...