• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 119
  • 90
  • 47
  • 18
  • 12
  • 8
  • 8
  • 6
  • 2
  • 2
  • 2
  • 2
  • 1
  • Tagged with
  • 367
  • 85
  • 55
  • 44
  • 43
  • 40
  • 37
  • 33
  • 33
  • 32
  • 32
  • 31
  • 30
  • 29
  • 27
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
151

Molecular Characterization of Experimental Traumatic Brain Injury

Israelsson, Charlotte January 2006 (has links)
Traumatic brain injury (TBI) is the most common cause of mortality and disability in the younger (<50 years) Swedish population with an incidence rate of 20,000 cases per year. This thesis aims to increase the understanding of brain injury mechanisms, especially in a molecular and cellular context. Bone morphogenetic protein (BMP) signalling was examined in three genetically modified mice (two “loss-of-function”, one “gain-of-function”) exposed to TBI (controlled cortical impact, CCI) with CaMKII used as promoter for Cre-driven recombination in postnatal forebrain neurons. The mice survived, developed normally and did not show any obvious phenotypes except for an upregulation in Mtap2 mRNA in mice with impaired BMP signalling. Reactive Gfap and Timp1 mRNA expression measured using quantitative RT-PCR (qRT-PCR) was reduced in the mice overexpressing BMP signals. The BMP signalling pathway was further studied in cultured PC12 cells with BMP4 and NGF added. Egr3 expression was substantially increased by these growth factors. Blocking Egr or Junb functions reduced neurite outgrowth. TBI-induced mRNA expression changes in 100 selected genes in C57BL/6J mouse neocortex and hippocampus were measured using qRT-PCR at different time points post-injury. Several distinct gene clusters with similar expression patterns were identified. GeneChip analysis (Affymetrix) of the injured mouse neocortex at three days revealed 146 transcripts significantly upregulated, confirming and extending the qRT-PCR results. The findings demonstrate marked increases after injury among chemokine transcripts and activation of many genes involved in inflammation. In conclusion, the present study has revealed transcriptional changes in specific signalling pathways after brain injury. The results may help to identify novel targets for neuroprotective interventions after traumatic brain injury.
152

Analyse des effets de l'exercice physique dans un modèle murin de Sclérose Latérale Amyotrophique

Deforges, Séverine 08 April 2011 (has links) (PDF)
La sclérose latérale amyotrophique (SLA) est une maladie neurodégénérative fatale du sujet adulte caractérisée par la perte sélective des motoneurones (MN) rapides, des paralysies musculaires et un hypermétabolisme lipidique. L'exercice physique pourrait être envisagé comme traitement thérapeutique de la SLA. Sur les souris SLA, des protocoles de course différents ont des effets contradictoires en fonction de leur intensité. Dans ce contexte scientifique, nous avons comparé les adaptations neuromusculaires de souris sauvages et SLA SOD1 G93A soumises à un entraînement à la course sur tapis roulant et à la nage en piscine à courant que nous avons développée. Dans nos conditions expérimentales, la nage recrute préférentiellement les unités motrices rapides et active les MN de large surface, contrairement à la course qui recrute préférentiellement les unités motrices lentes (activation des MN de surface réduite et induction de transitions de fibres musculaires rapides vers lentes). Seule la nage retarde l'apparition des symptômes et allonge la durée de vie des souris SLA. Elle limite la mort des MN dans la moelle épinière lombaire et maintient l'intégrité des muscles squelettiques. Ces effets de la nage sont corrélés à l'adaptation du métabolisme énergique menant à une augmentation de l'utilisation du glucose et à la préservation des réserves lipidiques. Nos données mettent en évidence une relation entre l'activation et la protection des unités motrices rapides sensibles à la SLA. La détermination des mécanismes de neuroprotection induits par la nage dans les souris SLA nous permettrait de proposer de nouvelles voies thérapeutiques médicamenteuses aux patients atteints de SLA.
153

K(ATP) Channel blockade instructs microglia to foster brain repair and neurogenesis after stroke

Ortega González, Fco. Javier 13 April 2012 (has links)
Stroke causes CNS injury associated with strong fast microglial activation as part of the inflammatory response. Fast activation of microglia in response to neuronal damage requires the rapid availability of a large amount of energy to trigger diverse cytotoxic or neuroprotective signals. ATP-dependent potassium (K(ATP)) channels play important roles in many cellular functions by coupling cell metabolism to electrical activity. K(ATP) channels were first detected in cardiac myocytes and later found in beta-cells of the pancreas, skeletal muscle, neurons, smooth muscle, heart, pituitary, and tubular cells of the kidney. Our group and others have also demonstrated its expression in reactive microglia after brain injury. In rat models of stroke, blockade of the sulfonylurea receptor (SUR), with glibenclamide (Gbc) reduced cerebral edema and infarct volume. Furthermore, clinical data suggest the effectiveness of Gbc to treat stroke. Gbc close the K(ATP) channel by interaction with two drug-binding sites on SUR subunits, as well as, the astroglial NC(Ca-ATP) channel, which mediates the Gbc-induced prevention of edema after cerebral ischemia. In these studies however, the function of the K(ATP) channel remained unclear. Therefore, as Gbc may bind to constitute functional K(ATP) channels after ischemic stroke, other possible effects of Gbc might explain the effectiveness of this drug in the treatment of stroke. Giving the fact that, SUR1-regulated channels are exquisitely sensitive to changes in the metabolic state of the cell, and that microglia are sensing the environment, the expression of K(ATP) channels in activated microglia, will couple cell energy to membrane potential. We herein postulate, that the effectiveness of Gbc to treat stoke, at least in part, is caused by the KATP channel closure expressed by activated microglia, which may then be critical in determining, their participation in the pathogenic process. Given the analogy with beta-cells, K(ATP) channel blockade in microglia would response faster and more efficiently to the external signals released after brain injury. If true, blockade of microglial K(ATP) channel with low doses of Gbc during the early stages of stroke might foster neuroprotective microglial activity, could enhance ischemia-induced neurogenesis in the SVZ, and consequently will lead to an improved functional outcome. The work presented in this thesis demonstrates that, Gbc improves functional neurological outcome in stroke, accompanied by neuron preservation in the core of the ischemic brain. In this region, reactive microglia from tMCAO rats upregulate the K(ATP) channel, which makes microglia a target to Gbc actions in the early stages of stroke. Furthermore, Gbc also strengthens the neuroprotective role of microglia in the acute phase after focal cerebral ischemia, enhance long-term neurogenesis and brain repair processes. As such, identify microglial K(ATP) channels as a key target for stroke treatment. Overall, these results provide new therapeutic avenues for the treatment of other neurological disorders that involve microglia.
154

Lack of neuroprotective effects by platelet-derived growth factor against beta-amyloid induced toxicity uncovers a novel hypothesis of Alzheimer's disease pathology

Liu, Hui 04 May 2012 (has links)
Aβ oligomer-induced neurotoxicity has become an important area of therapeutic development in treating Alzheimer’s disease. Platelet-derived growth factor (PDGF) has been shown to be able to protect neurons against several neuronal insults such as ischemia and HIV1 toxin induced cytotoxicity. These neuroprotective effects correlate well with our previous results that demonstrate the neuroprotective effects of PDGF-BB, one of the PDGF receptor ligand subtypes, against NR2B containing NMDA receptor induced excitotoxicity, a possible underlying cause of Aβ oligomer induced synaptic dysfunction and neuronal death. This project examines the neuroprotective effect of PDGF-BB against Aβ1-42 oligomer induced cytotoxicity in both SH-SY5Y cells and primary hippocampal neurons. Cell viability was monitored by MTT assay and the affected signaling pathways were examined using pharmacological methods and Western blotting. The results demonstrated that Aβ1-42 oligomer elicited a dose-dependent toxicity with a sign of saturation at higher dosages, PDGF-BB failed to protect neurons against Aβ1-42 oligomer induced cytotoxicity. In contrast, Aβ1-42 oligomers strongly inhibit PDGF-BB induced mitogenesis in both SH-SY5Y cells and primary neurons. Further investigation using Western blotting to measure PDGF receptor expression and phosphorylation in SH-SY5Y cells showed that Aβ1-42 oligomer can inhibit PDGF-BB induced phosphorylation of PDGF β-receptor on Tyr1021, a site that is crucial for PLCγ mediated mitogenesis. These findings not only explained the poor neuroprotective effect elicited by PDGF-BB against Aβ1-42 oligomers, but also led to a novel hypothesis that Aβ1-42 oligomer may interfere with neurotrophic factor induced neuronal survival, either selectively or perhaps globally. Further exploration on this hypothesis will be able to shed light on this potentially novel mechanism of pathogenesis in Alzheimer’s disease.
155

Neuroprotective Role of Ubiquitin Carboxyl-Terminal Hydrolase L1 and Heat Shock Protein 70 at the Rostral Ventrolateral Medulla During Mevinphos Intoxication in the Rat

Chang, Chi 23 May 2005 (has links)
In eukaryotic cells, most proteins in the cytosol and nucleus are degraded via the ubiquitin-proteasome pathway. Ubiquitin is best known for its role in targeting proteins for degradation by the proteasome. Ubiquitin carboxyl-terminal hydrolase L1 (UCH-L1) is found specifically in central and peripheral neurons, and is responsible for the removal of small peptide fragments from the ubiquitin chain and for co-translational processing of ubiquitin gene products to generate free monomeric ubiquitin. In response to extreme conditions, cells exhibit an up-regulation of heat shock protein (HSP) expression, which contributes to repair and protective mechanisms. Within the HSP family, HSP70 is the major inducible member that protects against cell death. Based on the pharmacologic property of organophosphates as an inhibitor of cholinesterase, it is generally contended that manifestations of organophosphate poisoning, including secretion and muscle fasciculation, stupor, cardiopulmonary collapse, respiratory failure, coma or death, result from accumulation of, and over-stimulation by acetylcholine at peripheral of central synapses. One approach in furthering our understanding on organophosphate poisoning is delineation of its potential protective mechanisms. In this regard, the information on the cellular and molecular mechanisms that underlie organophosphate poisoning has received attention. Our laboratory demonstrated previously that a crucial brain site via which mevinphos (Mev), an organophosphate insecticide of the P=O type, acts is the rostral ventrolateral medulla (RVLM), the medullary origin of premotor sympathetic neurons that are responsible for the maintenance of vasomotor tone. The phasic changes in cardiovascular events over the course of acute Mev intoxication also parallel fluctuations of the ¡§life-and-death¡¨ signals that emanate form the RVLM. Based on a rat model of organophosphate poisoning that provides continuous information on cellular and molecular mechanisms in the RVLM, the present study was undertaken to evaluate whether changes in protein level of UCH-L1 or HSP70 are associated with death arising from Mev intoxication. We also evaluated the efficacy of both of them in the neuroprotection against fatality during Mev intoxication. The first part of this study investigated whether UCH-L1 plays a neuroprotective role at the RVLM, where Mev acts to elicit cardiovascular toxicity. In Sprague-Dawley rats maintained under propofol anesthesia, Mev (960 µg/kg, i.v.) induced a parallel and progressive augmentation in UCH-L1 or ubiquitin expression at the ventrolateral medulla during the course of Mev intoxication. The increase in UCH-L1 level was significantly blunted on pretreatment with microinjection bilaterally into the RVLM of a transcription inhibitor, actinomycin D (5 nmol) or a translation inhibitor, cycloheximide (20 nmol). Compared to artificial cerebrospinal fluid (aCSF) or sense uch-L1 oligonucleotide (100 pmol) pretreatment, microinjection of an antisense uch-L1 oligonucleotide (100 pmol) bilaterally into the RVLM significantly increased mortality, reduced the duration of the phase I (¡§pro- life¡¨ phase), blunted the increase in ubiquitin expression in ventrolateral medulla, and augmented the induced hypotension in rats that received Mev. The second part of this study investigated whether HSP70 plays a neuroprotective role at the RVLM. Intravenous administration of Mev (960
156

Dysfunction of Mitochondrial Respiratory Chain in Rostral Ventrolateral Medulla During Experimental Endotoxemia

Chuang, Yao-Chung 08 January 2003 (has links)
Dysfunction of Mitochondrial Respiratory Chain in Rostral Ventrolateral Medulla During Experimental Endotoxemia Sepsis is a complex pathophysiologic state resulting from an exaggerated whole-body inflammatory response to infection or injury. Metabolic disturbances, abnormal regulation of blood flow and diminished utilization of oxygen at the cellular level may account for tissue damage and lead to multiple organ failure and death. As the primary site of cellular energy generation is the mitochondrion, it presents itself as an important target for the septic cascade. In this regard, the notion that bioenergetic failure due to mitochondrial dysfunction contributes to organ failure during sepsis has received attention. We established the low frequency fluctuations in the systemic arterial pressure signals are related to the sympathetic neurogenic vasomotor tone, and reflect the functional integrity of the brain stem. Their origin is subsequently traced to the premotor sympathetic neurons at the rostral ventrolateral medulla (RVLM), whose neuronal activity is intimately related to the ¡§life-and-death¡¨ process. Based on a rat model of experimental endotoxemia that provides continuous information on changes in neuronal activity in the RVLM, the present study was undertaken to evaluate whether changes in mitochondrial respiratory functions are associated with death arising from sepsis. We also evaluated the efficacy of a new water-soluble coenzyme Q10 (CoQ10, ubiquinone) formula in the protection against fatality during endotoxemia by microinjection into bilateral RVLM. Dysfunction of Mitochondrial Respiratory Chain in Rostral Ventrolateral Medulla During Experimental Endotoxemia in the Rat We investigated the functional changes in mitochondrial respiratory chain at the RVLM in an experimental model of endotoxemia that mimics systemic inflammatory response syndrome. Experiments were carried out in adult male Sprague-Dawley rats that were maintained under propofol anesthesia. Intravenous administration of E. coli lipopolysaccharide (LPS; 30 mg/kg) induced progressive hypotension, with death ensued within 4 hours. The sequence of cardiovascular events during this LPS-induced endotoxemia can be divided into a reduction (Phase I), followed by an augmentation (Phase II; ¡§pro-life¡¨ phase) and a secondary decrease (Phase III; ¡§pro-death¡¨ phase) in the power density of the vasomotor components (0-0.8 Hz) of systemic arterial pressure (SAP) signals. Enzyme assay revealed significant decrease of the activity of NADH cytochrome c reductase (Complex I+III) and cytochrome c oxidase (Complex IV) in the RVLM during all 3 phases of endotoxemia. On the other hand, the activity of succinate cytochrome c reductase (Complex II+III) remained unaltered. Neuroprotective Effects of Coenzyme Q10 at Rostral ventrolateral Medulla Against Fatality During Experimental Endotoxemia in the Rat CoQ10 is a highly mobile electron carrier in the mitochondrial respiratory chain that also acts as an antioxidant. We evaluated the neuroprotective efficacy of CoQ10 against fatality in an experimental model of endotoxemia, using a novel water-soluble formulation of this quinone derivative. In Sprague-Dawley rats maintained under propofol anesthesia, intravenous administration of E. coli LPS (30 mg/kg) induced experimental endotoxemia. Pretreatment by microinjection bilaterally of CoQ10 (1 or 2 mg) into RVLM significantly diminished mortality, prolonged survival time, and reduced the slope or magnitude of the LPS-induced hypotension. CoQ10 pretreatment also significantly prolonged the duration of Phase II endotoxemia and augmented the total power density of the vasomotor components of SAP signals in Phase II endotoxemia. The increase in superoxide anion production induced by LPS at the RVLM during Phases II and III endotoxemia was also significantly blunted. Conclusion The present study revealed that selective dysfunction of respiratory enzyme Complexes I and IV in the mitochondrial respiratory chain at the RVLM is closely associated with fatal endotoxemia. CoQ10 provides neuroprotection against fatality during endotoxemia by acting on the RVLM. We further found that a reduction in superoxide anion produced during endotoxemia at the RVLM may be one of the mechanisms that underlie the elicited neuroprotection of CoQ10. These findings therefore open a new direction for future development of therapeutic strategy in this critical, complicated and highly fatal condition known as sepsis.
157

Le système EPOergique cérébral : caractérisation et implication dans la protection neuronale au cours de l'épileptogenèse

Nadam, Jérémie 27 June 2007 (has links) (PDF)
Notre travail de thèse est le premier à mettre en évidence chez le rat ayant subit un état de mal cérébral (EMC), que la production locale et réactionnelle d'érythropoïétine (Epo) par les astrocytes, quand elle est accompagnée de l'administration exogène d'Epo, représente une condition optimale pour le maintien des populations neuronales les plus fragiles. La neuroprotection induite par l'Epo implique un récepteur spécifique qui serait composé par l'association entre le récepteur à l'Epo (Epo-R) et la chaîne bêta commune (βc). L'absence de régulation coordonnée entre l'Epo-R et la βc que nous dénonçons, aussi bien dans des conditions physiologiques (développement et vieillissement du SNC) que physiopathologiques (EMC), est un argument fort permettant de réfuter l'hypothèse selon laquelle la βc constituerait une des sous-unités du récepteur de l'Epo impliqué dans la neuroprotection.
158

Temporal deregulation of genes and microRNAs in neurons during prion-induced neurodegeneration

Majer, Anna 18 June 2010 (has links)
Prion diseases are fatal and incurable neurodegenerative diseases that share many pathological similarities to other neurodegenerative diseases such as Alzheimer’s or Parkinson’s disease. One of the earliest pathological signs commonly detected in all of these diseases is the dysfunction followed by loss of neuronal synapses, spines and eventually dendrites that collectively contribute to disruption of normal brain function. These pathologies tend to progressively accumulate within the brain tissue such that extensive damage typically precedes clinical symptom manifestation and ultimate death of neurons. Clearly, understanding the molecular processes responsible for these pathologies could uncover critical pathway(s) that are responsible for propagating this brain damage and could therefore be exploited for therapy development. However, molecular mechanisms implicated in this early pathology remain unidentified. To address this gap in knowledge, this thesis describes a transcriptional approach coupled with specific isolation of neuronal-enriched tissue which was used to help delineate cellular pathways involved in prion-induced neurodegeneration. Profiling cell bodies of CA1 hippocampal neurons known to be affected during early prion disease revealed temporal alteration in both gene and microRNA (gene regulators) expression throughout disease. On a gene expression level, changes in transcript expression during preclinical disease were reminiscent of an activity-dependent neuroprotective gene signature previously described in the literature. These neuroprotective genes were induced during preclinical disease, diminished as disease progressed and were abolished at clinical disease. In support of this process, upregulation of the phosphorylated form of the neuroprotective transcription factor CREB was detected during preclinical disease in these neurons. Furthermore, several genes known to be induced by CREB were also upregulated at preclinical disease in prion-infected mice. Interestingly, expression of numerous deregulated microRNAs paralleled the neuroprotective gene signature of which several are known to remodel neuronal spines and dendrites. To determine whether other preclinically induced microRNAs were also capable of remodeling neuronal structures, gain-of-function studies were performed in primary mouse hippocampal neurons for the uncharacterized miR-26a-5p. Neurons over-expressing miR-26a-5p had enhanced spine density and dendrite arborization, similar to other preclinically-induced microRNAs. Together, these data suggests that CA1 hippocampal neurons induce a neuroprotective transcriptional signature that is evident early in the course of disease within CA1 hippocampal neurons and is abolished by clinical disease. Reestablishment of key molecules that can induce this neuroprotective signature at a time when these genes begin to dissipate could prolong prion disease onset and delay clinical symptom manifestation. / October 2015
159

Μηχανισμοί νευροεκφύλισης και νευροπροστασίας μετά από τη χορήγηση νευροστεροειδών σε παρκινσονικά μοντέλα

Μούρτζη, Θεοδώρα 29 April 2014 (has links)
Η Νόσος του Πάρκινσον αποτελεί τη δεύτερη συχνότερη νευροεκφυλιστική ασθένεια μετά τη νόσο του Αλτσχάιμερ, η οποία εμφανίζεται στο 2% των ανθρώπων άνω των 65 ετών. Η μέχρι στιγμής αντιμετώπισή της περιορίζεται σχεδόν αποκλειστικά στη χορήγηση του προδρόμου μορίου της ντοπαμίνης L-DOPA, με σκοπό την αντιμετώπιση των κινητικών προβλημάτων της ασθένειας, η οποία επιφέρει όμως ισχυρές παρενέργειες. Για το λόγο αυτό κρίνεται σκόπιμη η εύρεση νευροπροστατευτικών ουσιών οι οποίες θα καθυστερούν ή θα αναστέλλουν την εξέλιξη της νόσου, με τις ελάχιστες δυνατές παρενέργειες. Ο μυς weaver, φέρει μία αυτοσωμική υπολειπόμενη μετάλλαξη στο γονίδιο Girk2 και αποτελεί το μοναδικό γενετικό μοντέλο της νόσου του Πάρκινσον, το οποίο εμφανίζει προοδευτική απώλεια των ντοπαμινεργικών νευρώνων της μέλαινας ουσίας, η οποία συμβαίνει ενδογενώς. Για το λόγο αυτό θεωρείται ιδανικό για μελέτες νευροπροστασίας. Τόσο in vitro, όσο και in vivo μελέτες στο μοντέλο weaver αλλά και σε άλλα παρκινσονικά μοντέλα (συμπεριλαμβανομένων προηγούμενων αποτελεσμάτων της ομάδας μας, (Διδακτορική Διατριβή Κωνσταντίνου Μποτσάκη, Σεπτέμβριος 2013) αναδεικνύουν ότι το ενδογενές νευροστεροειδές δεϋδροεπιανδροστερόνη (DHEA), καθώς και ο θειικός της εστέρας (DHEA-S) ασκούν ισχυρότατη νευροπροστατευτική δράση στους ντοπαμινεργικούς νευρώνες της μέλαινας ουσίας των παρκινσονικών αυτών ζώων. Επιπλέον, το συνθετικό ανάλογο του DHEA 17β-spiro[5-androstene-17,20-oxiran]-3β-ol (ΒΝΝ-50), το οποίο δεν μεταβολίζεται ενδογενώς σε οιστρογόνα (και θα μπορούσε κατά συνέπεια να είναι κατάλληλο για κλινική χρήση), φαίνεται να ασκεί την ίδια νευροπροστατευτική επίδραση στους ντοπαμινεργικούς νευρώνες της μέλαινας ουσίας των ομοζυγωτικών μυών weaver (Δ.Δ Κωνσταντίνου Μποτσάκη, Σεπτέμβριος 2013), αλλά και σε κυτταροκαλλιέργειες PC12 κυττάρων. Σκοπός της παρούσας εργασίας ήταν η διερεύνηση του μηχανισμού δράσης των νευροπροστατευτικών αυτών ουσιών. Για το λόγο αυτό διερευνήθηκε η πιθανή αντιαποπτωτική δράση των DHEA-S και ΒΝΝ-50, μέσω προσδιορισμού του λόγου των επιπέδων της αντιαποπτωτικής πρωτεΐνης Bcl-2 προς τα επίπεδα της προαποπτωτικής πρωτεΐνης Bax. Ο προσδιορισμός αυτός πραγματοποιήθηκε στο μεσεγκέφαλο φυσιολογικών μυών και μυών weaver ηλικίας Ρ21 ημερών, μετά από χρόνια χορήγηση των νευροστεροειδών (από την Ρ1 έως την Ρ21). Επιπλέον, διερευνήθηκε η πιθανή αντιοξειδωτική δράση του νευροστεροειδούς ΒΝΝ-50, μέσω του προσδιορισμού της ελεύθερης (frMDA) και προσδεδεμένης σε πρωτεΐνες (prMDA) μαλονικής διαλδεΰδης, στο μεσεγκέφαλο φυσιολογικών μυών και μυών weaver ηλικίας Ρ21 ημερών, μετά από χρόνια χορήγηση του αναλόγου (από την Ρ1 έως την Ρ21). Τα αποτελέσματα ανέδειξαν ότι τόσο το DHEA-S, όσο και το BNN-50 ασκούν ισχυρή αντιαποπτωτική δράση στο μεσεγκέφαλο των μυών weaver, αυξάνοντας τον λόγο Bcl-2/Bax (o οποίος εμφανίζεται μειωμένος στo μεσεγκέφαλο των wv/wv ζώων, σε σχέση με τα φυσιολογικά ζώα) κατά 74% και 83% αντίστοιχα, σε σχέση με τα weaver μύες που έλαβαν φυσιολογικό ορό. Η δράση αυτή επιτυχάνεται με διαφορετικό τρόπο, ανάμεσα στα δύο νευροστεροειδή. Πιο συγκεκριμένα, το DHEA-S επαναφέρει πλήρως τα μειωμένα επίπεδα της αντιαποπτωτικής πρωτεΐνης Bcl-2 στo μεσεγκέφαλο των wv/wv ζώων χωρίς να επηρεάζει τα επίπεδα της προαποπτωτικής πρωτεΐνης Bax, ενώ το ΒΝΝ-50 επαναφέρει πλήρως τα αυξημένα επίπεδα της προαποπτωτικής πρωτεΐνης Βax στo μεσεγκέφαλο των wv/wv ζώων, χωρίς να επηρεάζει τα επίπεδα της αντιαποπτωτικής πρωτεΐνης Bcl-2. Επιπλέον, κατέστει σαφές ότι το στεροειδές ΒΝΝ-50 κατέχει και αντιοξειδωτική δράση, αφού επάγει τη δραματική μείωση των επιπέδων της ολικής μαλονικής διαλδεΰδης, τελικού προϊόντος της υπεροξείδωσης των λιπιδίων που εμφανίζεται αυξημένη στo μεσεγκέφαλο των wv/wv ζώων, φέρνοντάς τα κοντά στα φυσιολογικά επίπεδα. Η μείωση αυτή οφείλεται αποκλειστικά στη μείωση των επιπέδων της προσδεδεμένης σε πρωτεΐνες και όχι της ελεύθερης ΜDA, πιθανότατα λόγω της πολύ μεγαλύτερης αύξησης της πρώτης σε σχέση με τη δεύτερη, στο μεσεγκέφαλο των ομοζυγωτικών weaver μυών. Τα παραπάνω αποτελέσματα οδηγούν στο συμπέρασμα ότι τα νευροστεροειδή DHEA-S και ΒΝΝ-50 ασκούν τη νευροπροστατευτική τους επίδραση δρώντας πλειοτροπικά, τουλάχιστον μέσω δύο γραμμών δράσης, της αντιαποπτωτικής και αντιοξειδωτικής. Το γεγονός αυτό είναι ιδιαίτερα σημαντικό καθώς για πρώτη φορά, γίνεται μελέτη του μηχανισμού της δράσης του στεροειδούς ΒΝΝ-50 in vivo. Καθώς το BNN-50 δεν μεταβολίζεται ενδογενώς σε φυλετικές ορμόνες, και θα μπορούσε συνεπώς να είναι κατάλληλο για κλινική χρήση, κρίνεται απαραίτητη η περαιτέρω διερεύνηση του μηχανισμού δράσης του, ώστε να διευκρινιστεί η πιθανή ικανότητα χρήσης του για αντιμετώπιση της σοβαρής αυτής νόσου, μελλοντικά. / Parkinson’s Disease is the second most common neurodegenerative disease, affecting about 2% of the population aged over 65 years old. The most common treatment of the disease until now, is the administration of L-DOPA, a dopamine precursor, in order to reduce the locomotive defects caused by the disease, a drug that causes severe side effects. Hence, the discovery of neuroprotective compounds that can prohibit or at least prolong the progression of the disease is highly required. The weaver mouse carries an autosomic recessive mutation at the Girk-2 gene and consists the only non-invasive genetic model of Parkinson’s Disease that exhibits progressive neurodegeneration of the nigrostriatal dopaminergic neurons. Thus, it is ideal for neuroprotection studies. The endogenous neurosteroif dehydroepiandrosterone (DHEA) as well as its sulphated ester (DHEA-S) exhibit a significant neuroprotective effect on the dopaminergic neurons of the weaver mouse (K. Botsakis phD thesis, 2013), as well as in other in vitro and in vivo studies. Additionally, the synthetic analogue of DHEA 17β-spiro[5-androstene-17,20-oxiran]-3β-ol (BNN-50), that is not metabolized to estrogens in vivo, exhibits the same neuroprotective effect on the dopaminergic neurons of the weaver mouse as DHEA-S (K. Botsakis phD thesis, 2013) as well as in PC12 cell cultures. The aim of this study was the investigation of the mechanism of action of DHEA-S and BNN-50 in vivo. To that extend, we investigated the possible antiapoptotic action of DHEA-S and BNN -50 by determining the ratio of the levels of the antiapoptotic protein Bcl-2 to the levels of pro-apoptotic protein Bax. The assay was performed in the midbrain of control mice and omozygous weaver mice at P21, after chronic administration of neurosteroids ( from P1 to P21 ) . Furthermore , we investigated the potential antioxidant properties of the neurosteroid BNN -50 , through the determination of free (frMDA) and protein-bound (prMDA) malonic dialdeyde in the midbrain of control mice and omozygous weaver mice at P21 after chronic administration of analogue ( from P1 to P21 ) . The results revealed that DHEA-S as well as the BNN-50 exert a very important antiapoptotic action in the weaver mouse midbrain, increasing the ratio of Bcl-2/Bax (that is reduced in the midbrain of the wv / wv animals, compared to control) by 74 % and 83 % respectively, compared with the weaver mice that received saline. This antiapoptotic action is achieved in different ways for the two neurosteroids . More specifically, DHEA-S fully restore the reduced levels of the antiapoptotic protein Bcl-2 in the wv/wv midbrain, without affecting the levels of the proapoptotic protein Bax, while the BNN -50 fully restored the elevated levels of proapoptotic protein Bax in the wv/wv midbrain, without affecting the levels of the antiapoptotic protein Bcl-2. Moreover , it became clear that the steroid BNN -50 possesses significant antioxidant activity, inducing a dramatic reduction in the levels of total MDA , the end product of lipid peroxidation that is displayed increased in the wv/wv midbrain, bringing the MDA level almost to control. The decrease is due to the reduction of the levels of protein-bound and not free MDA , probably due to the much higher increase of the first than the second , in the midbrain of homozygous weaver mice . The aforementioned results suggest that thee neurosteroids DHEA-S and BNN -50 exert their neuroprotective effects by acting pleiotropically , at least through two lines of action, one antiapoptotic and one antioxidant . This is particularly important because for the first time , there is a study of the mechanism of action of steroid BNN -50 in vivo. As BNN-50 is not metabolized endogenously in sex hormones , it could therefore be suitable for clinical use. Hence, it is necessary to further investigate the mechanism of action of the steroid, in order to clarify its possible usability for treating this serious disease in the future .
160

The modulating effect of sildenafil on cell viability and on the function of selected pharmacological receptors in cell cultures / B.E. Eagar

Eager, Blenerhassit Edward January 2004 (has links)
Since sildenafil's (Viagra®), a phospodiesterase type 5 (PDE5) inhibitor, approval for the treatment of male erectile dysfunction (MED) in the United States early 1998, 274 adverse event reports were filed by the Food and Drug Administration (FDA) between 4 Jan. 1998 and 21 Feb. 2001 with sildenafil as the primary suspect of various neurological disturbances, including amnesia and aggressive behaviour (Milman and Arnold, 2002). These and other research findings have prompted investigations into the possible central effects of sildenafil. The G protein-coupled muscarinic adetylcholine receptors (mAChRs) and serotonergic receptors (5HT-Rs), have been linked to antidepressant action (Brink et al. 2004). GPCRs signal through the phosphatidylinositol signal transduction pathway known to activate protein kinases (PKs). Since the nitric oxide (NO)-guanylyl cyclase signal transduction pathway is also known to involve the activation of PKs (via cyclic guanosine monophosphate (cGMP)), the scope is opened for sildenafil to possibly modulate the action of antidepressants by elevating cGMP levels. It is generally assumed that excitotoxic delayed cell death is pathologically linked to an increase in the release of excitatory neurotransmitters e.g. glutamate. Glutamate antagonists, especially those that block the define NMDA-receptors, are neuroprotective, showing the importance of the NMDA-NO-cGMP pathway in neuroprotection (Brandt et al., 2003). Sildenafil may play a role in neuroprotection by elevating cGMP levels. Aims: The aims of the study were to investigate any neuroprotective properties of sildenafil, as well as modulating effects of sildenafil pre-treatment on mAChR function. Methods: Human neuroblastoma SH-SY5Y or human epithelial HeLa cells were seeded in 24-well plates and pre-treated for 24 hours in serum-free medium with no drug (control), PDE5 inhibitors sildenafil (100nM and 450 nM), dipiridamole (20 µM) or zaprinast (20 µM), non-selective PDE inhibitor 3-isobutyl-I-methylxanthine (IBMX - ImM), cGMP analogue N2,2'-0-dibutyrylguanosine 3'5'-cyclic monophosphate sodium salt (500 µM), guanylcyclase inhibitor 1H-[1 ,2,4]oxadiazolo[4,3-a]quinoxalin-I-one (ODQ - 3 µM) or sildenafil + ODQ (450 nM and 3 µM respectively). Thereafter cells were used to determine mAChR function by constructing dose-response curves of methacholine or to determine cell viability utilising the Trypan blue, propidium iodide and MTT tests for cell viability. Results: Sildenafil pre-treatments induced a 2.5-fold increase in ,the Emax value of methacholine in neuronal cells but did not show a significant increase in epithelial cells The Trypan blue test suggests that neither the PDE5 inhibitors nor a cGMP analogue show any neuroprotection. Rather, sildenafil 450 nM, dipiridamole and IBMX displayed a neurodegenerative effect. The MTT test was not suitable, since pre-treatment with the abovementioned drugs inhibited the formation of forrnazan. The propidium iodide assay could also not be used, due to severe cell loss. Conclusion: Sildenafil upregulates mAChR function in SH-SY5Y cells and displays a neurodegenerative, and not a protective property, in neuronal cells. This is not likely to be associated with its PDE5 inhibitory action, but may possibly be linked to an increase in cGMP levels via the NO-cGMP pathway. / Thesis (M.Sc. (Pharmacology))--North-West University, Potchefstroom Campus, 2005.

Page generated in 0.0731 seconds