Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 5520, 2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35414699

ABSTRACT

Meteorites preserve evidence of processes on their parent bodies, including alteration, metamorphism, and shock events. Here we show that the Kakowa (L6) ordinary chondrite (OC) preserves both shock-melt veins and pockets of detrital grains from a brecciated and altered object, including corundum, albite, silica, fayalite, forsterite, and margarite in a Pb- and Fe-rich matrix. Preservation of the observed mineralogy and texture requires a sequence of at least two impacts: first, a high-velocity collision formed the shock melt veins containing the high-pressure minerals ringwoodite, wadsleyite, majorite, and albitic jadeite; later, a low-velocity impact formed fractures and filled them with the detrital material. Oxygen and Pb isotope ratios suggest an OC origin for these detrital minerals. Although fluid alteration is common in carbonaceous chondrites, the discovery of margarite with an OC oxygen isotopic signature is novel. Kakowa extends both the impact and alteration history of L6 ordinary chondrites in general.

2.
J Therm Spray Technol ; 31(1-2): 206-216, 2022.
Article in English | MEDLINE | ID: mdl-38624750

ABSTRACT

Grinding wheels are usually manufactured by powder metallurgical processes, i.e., by molding and sintering. Since this requires the production of special molds and the sintering is typically carried out in a continuous furnace, this process is time-consuming and cost-intensive. Therefore, it is only worthwhile for medium and large batches. Another influencing factor of the powder metallurgical process route is the high thermal load during the sintering process. Due to their high thermal sensitivity, superabrasives such as diamond or cubic boron nitride are very difficult to process in this way. In this study, a novel and innovative approach is presented, in which superabrasive grinding wheels are manufactured by thermal spraying. For this purpose, flat samples as well as grinding wheel bodies were coated by low-pressure (LP) cold gas spraying with a blend of a commercial Cu-Al2O3 cold gas spraying powder and nickel-coated diamonds. The coatings were examined metallographically in terms of their composition. A well-embedded superabrasive content of 12 % was achieved. After the spraying process, the grinding wheels were conditioned and tested for the grinding application of cemented carbides and the topographies of both the grinding wheel and the cemented carbide were evaluated. Surface qualities of the ground surface that are comparable to those of other finishing processes were reached. This novel process route offers great flexibility in the combination of binder and hard material as well as a cost-effective single-part and small-batch production.

3.
Astrobiology ; 16(12): 949-963, 2016 12.
Article in English | MEDLINE | ID: mdl-27905827

ABSTRACT

Nitrogen is a major nutrient for all life on Earth and could plausibly play a similar role in extraterrestrial biospheres. The major reservoir of nitrogen at Earth's surface is atmospheric N2, but recent studies have proposed that the size of this reservoir may have fluctuated significantly over the course of Earth's history with particularly low levels in the Neoarchean-presumably as a result of biological activity. We used a biogeochemical box model to test which conditions are necessary to cause large swings in atmospheric N2 pressure. Parameters for our model are constrained by observations of modern Earth and reconstructions of biomass burial and oxidative weathering in deep time. A 1-D climate model was used to model potential effects on atmospheric climate. In a second set of tests, we perturbed our box model to investigate which parameters have the greatest impact on the evolution of atmospheric pN2 and consider possible implications for nitrogen cycling on other planets. Our results suggest that (a) a high rate of biomass burial would have been needed in the Archean to draw down atmospheric pN2 to less than half modern levels, (b) the resulting effect on temperature could probably have been compensated by increasing solar luminosity and a mild increase in pCO2, and (c) atmospheric oxygenation could have initiated a stepwise pN2 rebound through oxidative weathering. In general, life appears to be necessary for significant atmospheric pN2 swings on Earth-like planets. Our results further support the idea that an exoplanetary atmosphere rich in both N2 and O2 is a signature of an oxygen-producing biosphere. Key Words: Biosignatures-Early Earth-Planetary atmospheres. Astrobiology 16, 949-963.


Subject(s)
Atmosphere , Climate , Exobiology , Geology , Models, Theoretical , Nitrogen/analysis , Aerobiosis , Anaerobiosis , Carbon/analysis , Earth, Planet , Time Factors
4.
Glia ; 64(12): 2219-2230, 2016 12.
Article in English | MEDLINE | ID: mdl-27641725

ABSTRACT

Oxidative stress critically contributes to the pathogenesis of a variety of neurodegenerative diseases such as multiple sclerosis. Astrocytes are the main regulators of oxidative homeostasis in the brain and dysregulation of these cells likely contributes to the accumulation of oxidative damage. The nuclear factor erythroid 2-related factor 2 (Nrf2) is the main transcriptional regulator of the anti-oxidant stress defense. In this study, we elucidate the effects of astrocytic Nrf2-activation on brain-intrinsic inflammation and lesion development. Cells deficient for the Nrf2 repressor kelch-like ECH-associated protein 1 (Keap1) are characterized by hyperactivation of Nrf2-signaling. Therefore, wild type mice and mice with a GFAP-specific Keap1-deletion were fed with 0.25% cuprizone for 1 or 3 weeks. Cuprizone intoxication induced pronounced oligodendrocyte loss, demyelination and reactive gliosis in wild type animals. In contrast, astrocyte-specific Nrf2-activation was sufficient to prevent oligodendrocyte loss and demyelination, to ameliorate brain intrinsic inflammation and to counteract axonal damage. Our results highlight the potential of the Nrf2/ARE system for the treatment of neuroinflammation in general and of multiple sclerosis in particular. © GLIA 2016;64:2219-2230.


Subject(s)
Astrocytes/metabolism , Demyelinating Diseases/etiology , Gene Expression Regulation/physiology , Multiple Sclerosis/complications , Multiple Sclerosis/pathology , NF-E2-Related Factor 2/metabolism , Animals , Astrocytes/drug effects , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Cuprizone/toxicity , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monoamine Oxidase Inhibitors/toxicity , Multiple Sclerosis/chemically induced , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/metabolism
5.
Br J Pharmacol ; 172(1): 80-92, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25220526

ABSTRACT

BACKGROUND AND PURPOSE: Modulation of the sphingosine 1-phosphate receptor is an approved treatment for relapsing multiple sclerosis because of its anti-inflammatory effect of retaining lymphocytes within the lymph nodes. Here, we evaluated the potential of an agonist at this receptor, FTY720 (fingolimod), to activate the promyelinating pathways within the brain to encourage remyelination and neuroprotection. EXPERIMENTAL APPROACH: In this study, we used the cuprizone model in male C57BL/6 mice and tested the promyelinating and neuroprotective effects of FTY720 after acute and chronic toxin-induced experimental demyelination. We used histological, immunohistochemical and gene expression methods. KEY RESULTS: The midline of the corpus callosum was severely demyelinated after acute and chronic cuprizone-induced demyelination. Robust endogenous remyelination was evident after acute, but impaired after chronic, demyelination. FTY720 treatment modestly accelerated myelin recovery after acute but not chronic cuprizone exposure. Markers of gliosis (astrocyte and microglia activation) were not affected by FTY720 treatment. Remarkably, the accumulation of amyloid precursor protein-positive spheroids in axons was less distinct in FTY720-treated animals, indicating that this compound alleviated ongoing axonal damage. CONCLUSIONS AND IMPLICATIONS: We show that even during endogenous remyelination, axonal degeneration continued at a low level, accumulating over time. This continuous neurodegenerative process was ameliorated by FTY720 treatment. FTY720 preserved CNS integrity by direct interaction with brain resident cells, the actions of which are still to be defined.


Subject(s)
Demyelinating Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Propylene Glycols/therapeutic use , Receptors, Lysosphingolipid/agonists , Sphingosine/analogs & derivatives , Amyloid beta-Protein Precursor/metabolism , Animals , Corpus Callosum/drug effects , Corpus Callosum/pathology , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Fingolimod Hydrochloride , Gene Expression Regulation/drug effects , Liver/drug effects , Liver/metabolism , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Phosphatidate Phosphatase/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Propylene Glycols/pharmacology , Sphingosine/pharmacology , Sphingosine/therapeutic use
6.
Exp Brain Res ; 224(2): 155-64, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23064810

ABSTRACT

Middle cerebral artery occlusion (MCAO) models have become well established as the most suitable way to simulate stroke in experimental studies. The high variability in the size of the resulting infarct due to filament composition, rodent strain and vessel anatomy makes the setup of such models very complex. Beside controllable variables of homeostasis, the choice of anesthetics and the grade of ischemia and reperfusion played a major role for extent of neurological injury. Transient MCAO was induced during either isoflurane or ketamine/xylazine (ket/xyl) anesthesia with simultaneously measurement of cerebral blood flow (CBF) in 60 male Wistar rats (380-420 g). Neurological injury was quantified after 24 h. Isoflurane compared with ket/xyl improved mortality 24 h after MCAO (10 vs. 50 %, p = 0.037) and predominantly led to striatal infarcts (78 vs. 18 %, p = 0.009) without involvement of the neocortex and medial caudoputamen. Independent of anesthesia type, cortical infarcts could be predicted with a sensitivity of 67 % and a specificity of 100 % if CBF did not exceed 35 % of the baseline value during ischemia. In all other cases, cortical infarcts developed if the reperfusion values remained below 50 %. Hyperemia during reperfusion significantly increased infarct and edema volumes. The cause of frequent striatal infarcts after isoflurane anesthesia might be attributed to an improved CBF during ischemia (46 ± 15 % vs. 35 ± 19 %, p = 0.04). S-100ß release, edema volume and upregulation of IL-6 and IL-1ß expression were impeded by isoflurane. Thus, anesthetic management as well as the grade of ischemia and reperfusion after transient MCAO demonstrated important effects on neurological injury.


Subject(s)
Anesthesia/methods , Anesthetics/therapeutic use , Brain Injuries/drug therapy , Brain Injuries/etiology , Cerebrovascular Circulation/drug effects , Infarction, Middle Cerebral Artery/complications , Animals , Blood Pressure/drug effects , Brain Edema/etiology , Brain Injuries/mortality , Cerebral Infarction/drug therapy , Cerebral Infarction/etiology , Cytokines/metabolism , Disease Models, Animal , Encephalitis/drug therapy , Encephalitis/etiology , Heart Rate/drug effects , Isoflurane/therapeutic use , Ketamine/therapeutic use , Laser-Doppler Flowmetry , Male , Neurologic Examination , Rats , Rats, Wistar , Reperfusion
7.
J Mol Neurosci ; 49(1): 80-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23054589

ABSTRACT

The cuprizone model is a suitable animal model of de- and remyelination secondary to toxin-induced oligodendrogliopathy. From a pharmaceutical point of view, the cuprizone model is a valuable tool to study the potency of compounds which interfere with toxin-induced oligodendrocyte cell death or boost/inhibit remyelinating pathways and processes. The aim of this study was to analyze the vulnerability of neighboring white mater tracts (i.e., the fornix and cingulum) next to the midline of the corpus callosum which is the region of interest of most studies using this model. Male mice were fed cuprizone for various time periods. Different white matter areas were analyzed for myelin (anti-PLP), microglia (anti-IBA1), and astrocyte (anti-GFAP) responses by means of immunohistochemistry. Furthermore, Luxol fast blue-periodic acid Schiff stains were performed to validate loss of myelin-reactive fibers in the different regions. Cuprizone induced profound demyelination of the midline of the corpus callosum and medial parts of the cingulum that was paralleled by a significant astrocyte and microglia response. In contrast, lateral parts of the corpus callosum and the cingulum, as well as the fornix region which is just beneath the midline of the corpus callosum appeared to be resistant to cuprizone exposure. Furthermore, resistant areas displayed reduced astrogliosis and microgliosis. This study clearly demonstrates that neighboring white matter tracts display distinct vulnerability to toxin-induced demyelination. This important finding has direct relevance for evaluation strategies in this frequently used animal model for multiple sclerosis.


Subject(s)
Chelating Agents/toxicity , Corpus Callosum/pathology , Cuprizone/toxicity , Nerve Fibers, Myelinated/pathology , Animals , Astrocytes/drug effects , Astrocytes/pathology , Fornix, Brain/chemistry , Fornix, Brain/pathology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Myelin Proteins/analysis , Myelin Proteins/drug effects , Nerve Fibers, Myelinated/drug effects
8.
J Mol Neurosci ; 48(1): 66-76, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22528463

ABSTRACT

Brain inflammation plays a central role in multiple sclerosis (MS). Besides lymphocytes, the astroglia and microglia mainly contribute to the cellular composition of the inflammatory infiltrate in MS lesions. Several studies were able to demonstrate that cortical lesions are characterized by lower levels of inflammatory cells among activated microglia/macrophages. The underlying mechanisms for this difference, however, remain to be clarified. In the current study, we compared the kinetics and extent of microglia and astrocyte activation during early and late cuprizone-induced demyelination in the white matter tract corpus callosum and the telencephalic gray matter. Cellular parameters were related to the expression profiles of the chemokines Ccl2 and Ccl3. We are clearly able to demonstrate that both regions are characterized by early oligodendrocyte stress/apoptosis with concomitant microglia activation and delayed astrocytosis. The extent of microgliosis/astrocytosis appeared to be greater in the subcortical white matter tract corpus callosum compared to the gray matter cortex region. The same holds true for the expression of the key chemokines Ccl2 and Ccl3. The current study defines a model to study early microglia activation and to investigate differences in the neuroinflammatory response of white vs. gray matter.


Subject(s)
Cerebral Cortex/immunology , Chemokine CCL2/genetics , Chemokine CCL3/genetics , Corpus Callosum/immunology , Cuprizone/toxicity , Demyelinating Diseases/immunology , Encephalitis/immunology , Animals , Cerebral Cortex/pathology , Chemokine CCL2/immunology , Chemokine CCL3/immunology , Corpus Callosum/pathology , Demyelinating Diseases/chemically induced , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/genetics , Gene Expression Regulation/immunology , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Monoamine Oxidase Inhibitors/toxicity , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Oligodendroglia/immunology , Oligodendroglia/pathology
9.
Mult Scler ; 18(3): 258-63, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22383435

ABSTRACT

FTY720 (fingolimod; Gilenya®), a sphingosine 1-phosphate (S1P) receptor modulator, is the first oral disease-modifying therapy to be approved for the treatment of relapsing-remitting multiple sclerosis. FTY720 is rapidly converted in vivo to the active S-fingolimod-phosphate, which binds to S1P receptors. This action inhibits egress of lymphocytes from the lymph nodes, preventing entry into the blood and thus infiltration into the central nervous system. More recent studies, however, convincingly show that FTY720 crosses the blood-brain barrier, where it is thought to act on S1P receptors on cells within the central nervous system, such as astrocytes, oligodendrocytes or microglia. Here we discuss the evidence showing that FTY720 also plays a role in remyelination and repair within the brain. While the mechanisms of action still require firm elucidation, it is clear that FTY720 could also be reparative, extending its therapeutic potential for multiple sclerosis.


Subject(s)
Multiple Sclerosis/drug therapy , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Animals , Astrocytes/metabolism , Axons/metabolism , Central Nervous System/metabolism , Disease Models, Animal , Fingolimod Hydrochloride , Humans , Lysophospholipids/metabolism , Mice , Microglia/metabolism , Myelin Sheath/drug effects , Myelin Sheath/physiology , Neurons/metabolism , Oligodendroglia/metabolism , Propylene Glycols/pharmacology , Rats , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/physiology , Sphingosine/metabolism , Sphingosine/pharmacology , Sphingosine/therapeutic use
10.
J Neuroendocrinol ; 24(1): 62-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21592237

ABSTRACT

Sex steroids have been demonstrated as powerful compounds to protect neurones and neural tissue from neurotoxic challenges and during neurodegeneration. A multitude of cellular actions have been attributed to female gonadal steroid hormones, including the regulation of pro-survival and anti-apoptotic factors, bioenergetic demands and radical elimination, growth factor allocation and counteracting against excitotoxicity. In recent years, immune-modulatory and anti-inflammatory characteristics of oestrogen and progesterone have also come under scrutiny. To date, each of these physiological responses has been considered to be partially and selectively integrated in the mediation of steroid-mediated cell protection and tested in suitable animal models and in vitro systems. To what extent these individual effects contribute to the overall neural protection remains sketchy. One idea is that a battery of cellular mechanisms operates at the same time. On the other hand, interactions and the control of the brain-intrinsic and peripheral immune system may play an additional and perhaps pioneering function in this scenario, notwithstanding the importance of secondary adjuvant mechanisms. In the present review, we highlight neuroprotective effects of oestrogen and progesterone in two different disease models of the brain, namely acute ischaemic and demyelination damage, which represent the most common acute and degenerative neurological disorders in humans. Besides other inflammatory parameters, we discuss the idea that chemokine expression and signalling appear to be early hallmarks in both diseases and are positively affected by sex steroids. In addition, the complex interplay with local brain-resident immune-competent cells appears to be controlled by the steroid environment.


Subject(s)
Brain Ischemia/metabolism , Demyelinating Diseases/metabolism , Encephalitis/metabolism , Gonadal Steroid Hormones/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Animals , Chemokines/metabolism
11.
Physiol Res ; 60(Suppl 1): S49-60, 2011.
Article in English | MEDLINE | ID: mdl-21777034

ABSTRACT

Over a century ago, hyperplasia and hypertrophy of astrocytes was noted as a histopathological hallmark of multiple sclerosis and was hypothesized to play an important role in the development and course of this disease. However until today, the factual contribution of astrocytes to multiple sclerosis is elusive. Astrocytes may play an active role during degeneration and demyelination by controlling local inflammation in the CNS, provoking damage of oligodendrocytes and axons, and glial scarring but might also be beneficial by creating a permissive environment for remyelination and oligodendrocyte precursor migration, proliferation, and differentiation. Recent findings from our lab suggest that brain lipid binding protein (FABP7) is implicated in the course of multiple sclerosis and the regulation of astrocyte function. The relevance of our findings and data from other groups are highlighted and discussed in this paper in the context of myelin repair.


Subject(s)
Astrocytes/metabolism , Carrier Proteins/metabolism , Demyelinating Diseases/metabolism , Fatty Acid-Binding Proteins/metabolism , Multiple Sclerosis/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Astrocytes/pathology , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Fatty Acid-Binding Protein 7 , Humans , Multiple Sclerosis/pathology , Myelin Sheath/metabolism
12.
J Neuroendocrinol ; 23(9): 839-48, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21790808

ABSTRACT

Motoneurones located in the ventral horn of the spinal cord conciliate cholinergic innervation of skeletal muscles. These neurones appear to be exceedingly affected in neurodegenerative diseases such as amyotrophic lateral sclerosis. The dysfunction of motoneurones is typically accompanied by alterations of cholinergic metabolism and signalling, as demonstrated by a decrease in choline acetyltransferase (ChAT) expression. 17 ß-Oestradiol (E(2)) is generally accepted as neuroprotective factor in the brain under acute toxic and neurodegenerative conditions and also appears to exert a protective role for motoneurones. In the present study, we attempted to analyse the role of E(2) signalling on ChAT expression in the motoneurone-like cell line NSC-34 and in vivo. In a first step, we demonstrated the presence of oestrogen receptor α and ß in NSC-34 cells, as well as in the cervical and lumbar parts, of the male mouse spinal cord. Subsequently, we investigated the effect of E(2) treatment on ChAT expression. The application of E(2) significantly increased the transcription of ChAT in NSC-34 cells and in the cervical but not lumbar part of the spinal cord. Our results indicate that E(2) can influence the cholinergic system by increasing ChAT expression in the mouse spinal cord. This mechanism might support motoneurones, in addition to survival-promoting mechanisms, in the temporal balance toxic or neurodegenerative challenges.


Subject(s)
Choline O-Acetyltransferase/metabolism , Estradiol/pharmacology , Motor Neurons/drug effects , Motor Neurons/enzymology , Spinal Cord/drug effects , Spinal Cord/enzymology , Animals , Cell Differentiation , Cell Line , Choline O-Acetyltransferase/genetics , Estradiol/physiology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Enzymologic , Humans , Male , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Nuclear Receptor Coactivators/genetics , Nuclear Receptor Coactivators/metabolism , Signal Transduction , Spinal Cord/cytology
13.
Brain Behav Immun ; 25(8): 1554-68, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21620951

ABSTRACT

Several lines of evidence indicate that remyelination represents one of the most effective mechanisms to achieve axonal protection. For reasons that are not yet understood, this process is often incomplete or fails in multiple sclerosis (MS). Activated astrocytes appear to be able to boost or inhibit endogenous repair processes. A better understanding of remyelination in MS and possible reasons for its failure is needed. Using the well-established toxic demyelination cuprizone model, we created lesions with either robust or impaired endogenous remyelination capacity. Lesions were analyzed for mRNA expression levels by Affymetrix GeneChip® arrays. One finding was the predominance of immune and stress response factors in the group of genes which were classified as remyelination-supporting factors. We further demonstrate that lesions with impaired remyelination capacity show weak expression of the radial-glia cell marker brain lipid binding protein (BLBP, also called B-FABP or FABP7). The expression of BLBP in activated astrocytes correlates with the presence of oligodendrocyte progenitor cells. BLBP-expressing astrocytes are also detected in experimental autoimmune encephalomyelitis during the remission phase. Furthermore, highest numbers of BLBP-expressing astrocytes were evident in lesions of early MS, whereas significantly less are present at the rim of (chronic)-active lesions from patients with long disease duration. Transfection experiments show that BLBP regulates growth factor expression in U87 astrocytoma cells. In conclusion, we provide evidence that expression of BLBP in activated astrocytes negatively correlates with disease duration and in parallel with remyelination failure.


Subject(s)
Astrocytes/metabolism , Carrier Proteins/biosynthesis , Demyelinating Diseases/metabolism , Fatty Acid-Binding Proteins/biosynthesis , Multiple Sclerosis/metabolism , Nerve Tissue Proteins/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Adult , Aged , Animals , Blotting, Western , Cell Count , Cell Line, Tumor , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fatty Acid-Binding Protein 7 , Female , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Fluorescent Antibody Technique, Indirect , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis/pathology , Oligonucleotide Array Sequence Analysis , Osteopontin/biosynthesis , Platelet-Derived Growth Factor/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Transfection
14.
Inflamm Res ; 60(8): 723-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21516513

ABSTRACT

OBJECTIVE AND DESIGN: It has been demonstrated that changes in the normal-appearing white matter (NAWM) in multiple sclerosis precede the appearance of classical lesions. The understanding of NAWM biology in an established disease model might help to clarify why some of them progress to active demyelinating lesions. MATERIAL OR SUBJECTS: C57BL6 male mice (19-21 g) were used in this study. TREATMENT: Demyelination was induced by feeding mice a diet containing 0.2% cuprizone for up to 5 weeks. METHODS: Routine stainings (luxol fast blue, and hematoxylin and eosin) and immunohistochemistry were performed to assess myelin status and the inflammatory infiltrate. RESULTS: We demonstrated that, in the toxic demyelination cuprizone model, the corpus callosum is severely demyelinated after a 5-week cuprizone challenge (acute demyelination) whereas the fimbria of the hippocampus appear normal in routine myelin stainings. Microgliosis but not astrogliosis is evident after acute demyelination in the fimbria. Interestingly, both regions, the fimbria and the corpus callosum, demonstrated early oligodendrocyte apoptosis as well as intense microglia accumulation and activation. However, only the corpus callosum progresses to actively demyelination lesions whereas the fimbria does not. CONCLUSIONS: The applied model appears suitable for elucidating pathways which promote progression of affected tissue to an active lesion.


Subject(s)
Cuprizone/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Multiple Sclerosis/chemically induced , Multiple Sclerosis/pathology , Neuroprotective Agents/pharmacology , Animals , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Corpus Callosum/cytology , Corpus Callosum/metabolism , Corpus Callosum/pathology , Cuprizone/therapeutic use , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Hippocampus/anatomy & histology , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Nerve Fibers, Myelinated/pathology , Neuroprotective Agents/therapeutic use , Young Adult
15.
J Neuroendocrinol ; 23(7): 601-11, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21507085

ABSTRACT

Corticosteroids (CS) are effective in the treatment of many brain disorders, such as multiple sclerosis (MS) or traumatic brain injury. This has been scrutinised in different experimental animal models. However, neither the mechanisms, nor the site of CS action are fully understood. Short-term high-dose CS treatment improves MS symptoms and severity of clinical disability during an acute inflammatory exacerbation of disease. In the present study, we analysed the influence of CS on the expression of cellular and molecular markers of spontaneous endogenous remyelination in the toxic non-immune cuprizone animal model at early (9 days) and intermediate (21 days) remyelination, as well as steroidal effects in primary astrocytes and oligodendrocyte progenitor cultures. Dexamethasone (Dex) and methylprednisolone (MP) induced a higher expression of the differentiation markers myelin basic protein and proteolipid protein (PLP) in cultured oligodendrocyte progenitor cells (OPC). CS exposure of primary cultured astrocytes resulted in a greater expression of those genes involved in OPC proliferation [fibroblast growth factor 2 (FGF2) and platelet-derived growth factor (PDGF)-αα] and a reduced expression of the pro-maturation factor insulin-like growth factor 1. Pro-maturating effects of CS were completely blocked by FGF2 and PDGF-αα co-application in OPC cultures. MP treatment in vivo resulted in a reduced recovery of PLP-staining intensity, whereas the re-population of the demyelinated corpus callosum with adenomatous polyposis coli-expressing oligodendrocytes was not affected. The numbers of brain intrinsic inflammatory cells, microglia and astrocytes during remyelination were similar in placebo and MP-treated animals. Our findings suggest that treatment with CS might have, in addition to the well-known benefical effects on inflammatory processes, a negative influence on remyelination.


Subject(s)
Adrenal Cortex Hormones/adverse effects , Corpus Callosum/drug effects , Cuprizone , Demyelinating Diseases/chemically induced , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Adrenal Cortex Hormones/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Chelating Agents , Corpus Callosum/metabolism , Demyelinating Diseases/pathology , Dexamethasone/adverse effects , Dexamethasone/pharmacology , Drug Evaluation, Preclinical , Male , Methylprednisolone/adverse effects , Methylprednisolone/pharmacology , Mice , Mice, Inbred C57BL , Nerve Regeneration/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Rats
16.
J Neuroendocrinol ; 22(8): 926-35, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20403091

ABSTRACT

The regulation of mitochondrial energy metabolism is not only important for normal functioning of neurones, but also appears to be essential during acute damage and neurodegeneration in the central nervous system. This makes mitochondria an interesting regulatory target for therapeutic approaches. Oestrogen is well-recognised as a protective hormone in the central nervous system under pathological threats. In the present study, we analysed the influence of oestrogen on the expression of mitochondria-encoded genes and mitochondrial activity in spinal cord cells both in vitro and vivo. Hormone application increased the transcription of mitochondrial respiratory chain enzymes (MRCE). This effect was observed in cultured spinal cord neurones, where it was inhibited by a nuclear oestrogen receptor (ER) antagonist and mainly mediated by the activation of ERbeta. No effect of oestrogen was observed in cultured spinal cord astroglia. In addition, the mitochondrial transcription factor A and nuclear respiratory factor 1 were up-regulated by oestrogen in a similar way as MRCE in vitro, and ATP levels were elevated after the application of the specific ERbeta agonist 2,3-bis(4-hydroxyphenyl)-propionitrile in cultured spinal cord nerve cells. The exposure of young male mice to oestrogen yielded increased levels of MRCE transcripts in the spinal cord. These data clearly show that systemic application of oestrogen stimulates MRCE expression in the spinal cord and predominantly in neurones. Further studies are required to demonstrate the potency of oestrogen to counteract pathological damage by stabilising mitochondrial performance.


Subject(s)
Electron Transport/physiology , Estrogens/pharmacology , Mitochondria/enzymology , Spinal Cord , Transcription, Genetic/drug effects , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/enzymology , Astrocytes/physiology , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Estrogen Antagonists/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Neurons/physiology , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/enzymology , Transcription Factors/genetics , Transcription Factors/metabolism
17.
J Neuroendocrinol ; 21(10): 841-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19686448

ABSTRACT

Oestrogens are powerful endogenous and exogenous neuroprotective hormones in animal models of brain injury, including focal cerebral ischaemia. This protective effect has been demonstrated under a variety of different treatments and injury paradigms, such as in vivo and in vitro stroke conditions. Neuroprotection in the central nervous system by progesterone is less defined. In the present study, cultured cortical and midbrain mouse neurones and human neuroblastoma cells (SH-SY5Y) were exposed to combined glucose-serum deprivation (CGSD), which is regarded as a reliable model mimicking the effects of ischaemia in vitro. Cell viability was assayed using lactate dehydrogenase release and metabolic activity. Conditions for CGSD treatment were chosen to yield half-maximal cell death rates. The validity of CGSD in vitro was compared with permanent middle cerebral artery occlusion (MCAO) in vivo. CGSD for 4 h induced half-maximal neuronal cell death. MCAO in vivo for the same period resulted in significant neuronal loss, also suggesting the validity of CGSD as a suitable stroke-like in vitro model. Combined steroid treatment (17beta-oestradiol and progesterone) but not the application of single steroids abolished CGSD-induced cell death of cortical neurones in vitro. By contrast, no cell protection was found in midbrain neurones or neuroblastoma cells. The co-application of oestrogen (ICI 182,780) or progesterone (RU-486) receptor antagonists did not obviously counteract the protective steroid effects. This suggests the operation of nonclassical steroid mechanisms and their implication in mediation of hormonal effects. The surplus of combined protective hormonal effects might be a result of the observed influence of progesterone application on neuronal oestradiol synthesis. The data obtained in the present study clearly highlight the potential of a combined steroid treatment under toxic degenerative brain pathologies.


Subject(s)
Cerebral Cortex/drug effects , Estradiol/pharmacology , Mesencephalon/drug effects , Neuroblastoma/drug therapy , Neuroprotective Agents/pharmacology , Progesterone/pharmacology , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Drug Therapy, Combination , Estradiol/administration & dosage , Glucose/deficiency , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Mesencephalon/pathology , Mesencephalon/physiology , Mice , Mice, Inbred BALB C , Neuroblastoma/physiopathology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Neuroprotective Agents/administration & dosage , Progesterone/administration & dosage , Rats , Rats, Wistar
18.
J Neuroendocrinol ; 19(10): 819-22, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17850464

ABSTRACT

Besides microglia, astrocytes exert an important regulatory function in the initiation and control of neuro-inflammatory processes in the central nervous system. Clinical and experimental data suggest that sex steroids are neuroprotective and that neurological/neurodegenerative disorders display sex-specific characteristics. Astroglia is known to respond to toxic stimuli by secretion of distinct pro-inflammatory/apoptotic cytokines. In the present study, we investigated the influence of oestrogen and progesterone on the expression of the cytokines tumour necrosis factor (TNF)-alpha and interleukin (IL)-18 in primary astrocytes obtained from neonatal mouse midbrain and cerebral cortex after the stimulation with lipopolysaccharides (LPS). LPS strongly induced the expression of TNF-alpha in astrocytes from both brain regions and IL-18 in those from midbrain. Oestrogen significantly attenuated LPS-induced TNF-alpha expression in the midbrain glia but not in the cortex glia. Combined treatment with oestrogen and progesterone together diminished LPS-induced IL-18 expression in the midbrain completely. Both steroid effects could be specifically antagonised by the steroid hormone receptor antagonists ICI 182 780 and mifepristone. We conclude that neuroprotective oestrogen and progesterone effects in the midbrain might be in part the consequence of a reduced pro-inflammatory response of astroglia.


Subject(s)
Astrocytes/metabolism , Estrogens/physiology , Interleukin-18/metabolism , Progesterone/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/immunology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Gene Expression Regulation/physiology , Interleukin-18/genetics , Lipopolysaccharides/immunology , Mesencephalon/cytology , Mesencephalon/immunology , Mice , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/genetics
19.
J Neuroendocrinol ; 19(9): 682-90, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17680883

ABSTRACT

Dopamine is actively and specifically eliminated from the extracellular space by astrocytes and neurones through dopamine transporters (DAT) and, afterwards, either recycled into vesicles or metabolised. The availability of dopamine reflects a critical point in the regulation of dopamine activity within the nigrostriatal circuit under normal and pathological conditions. From previous studies, we know that oestrogen regulates the efficacy of dopaminergic neurones at the synaptic level and improves dopamine function during Parkinson's disease. Accordingly, we investigated the contribution of local astroglial for extracellular dopamine elimination and the impact of oestrogen on DAT expression and activity. Using neonatal striatal and midbrain astrocyte cultures, we could demonstrate that astrocytes possess a specific dopamine uptake machinery and express DAT at considerable levels. The application of 17beta-oestradiol decreased the expression of DAT by 80% and 60% in midbrain and striatal astroglia cultures, respectively. The unspecific dopamine transporters (OCT3, VMAT2) were not detected in astroglia. Functionally, oestrogen exposure inhibited the clearance of dopamine from the extracellular space by 45% and 35% compared to controls in midbrain and striatal astroglia, respectively. The effect on DAT expression and activity was completely antagonised by the oestrogen receptor antagonist ICI 182 780. In conclusion, our data suggest that the positive reinforcement of dopamine transmission under physiological conditions and the alleviative impact of oestrogen under pathological conditions may be the result of a decline in DAT expression and therefore delayed dopamine uptake by astroglia.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/metabolism , Estrogens/metabolism , Substantia Nigra/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Cells, Cultured , Corpus Striatum/cytology , Dopamine Plasma Membrane Transport Proteins/genetics , Gene Expression Regulation , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , Substantia Nigra/cytology
20.
Mol Cell Biol ; 20(20): 7480-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003645

ABSTRACT

SARs (scaffold attachment regions) are candidate DNA elements for partitioning eukaryotic genomes into independent chromatin loops by attaching DNA to proteins of a nuclear scaffold or matrix. The interaction of SARs with the nuclear scaffold is evolutionarily conserved and appears to be due to specific DNA binding proteins that recognize SARs by a mechanism not yet understood. We describe a novel, evolutionarily conserved protein domain that specifically binds to SARs but is not related to SAR binding motifs of other proteins. This domain was first identified in human scaffold attachment factor A (SAF-A) and was thus designated SAF-Box. The SAF-Box is present in many different proteins ranging from yeast to human in origin and appears to be structurally related to a homeodomain. We show here that SAF-Boxes from four different origins, as well as a synthetic SAF-Box peptide, bind to natural and artificial SARs with high specificity. Specific SAR binding of the novel domain is achieved by an unusual mass binding mode, is sensitive to distamycin but not to chromomycin, and displays a clear preference for long DNA fragments. This is the first characterization of a specific SAR binding domain that is conserved throughout evolution and has DNA binding properties that closely resemble that of the unfractionated nuclear scaffold.


Subject(s)
Chromatin/metabolism , Conserved Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Chromatin/genetics , Chromomycins/pharmacology , Cloning, Molecular , DNA/genetics , DNA-Binding Proteins/genetics , Distamycins/pharmacology , Humans , Molecular Sequence Data , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins , Sequence Homology, Amino Acid , Substrate Specificity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...