• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 5
  • Tagged with
  • 5
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 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.
1

Mechanisms Underlying a Unique Form of Neuroendocrine Adaptation in Osmosensitive Supraoptic Neurons

2014 August 1900 (has links)
The neurohormonal mechanisms underlying the regulation of extracellular osmolality are of critical physiological importance. These mechanisms act to maintain the osmolality of human plasma close to a “set-point” of about 290 milliosmoles per litre. The magnocellular neurosecretory cells (MNCs) of the supraoptic nucleus (SON), synthesize and secrete the neurohypophysial hormones vasopressin (VP) and oxytocin (OT). The primary hormonal regulator of osmolality is VP, which is released by the MNCs as a function of plasma osmolality and acts by controlling water reabsorption at the kidneys. MNCs decrease their volume and thus plasma membrane tension in response to acute increases in external osmolality and lack the compensatory mechanisms that limit volume changes in most cell types. This enables them to transduce changes in osmolality into changes in excitability via a mechanosensitive cation channel. It has been shown in vivo that sustained increases in plasma osmolality, however, cause marked hypertrophy of the MNCs that is part of a structural and functional adaptation that is thought to enable the MNCs to secrete large quantities of VP for prolonged periods. The mechanism of this important structural and functional adaptation of MNCs is difficult to address in an in vivo preparation and so an in vitro model of acutely isolated MNCs was used to pharmacologically assess the hypertrophy. It was observed that MNCs exposed to sustained hypertonic solutions, underwent an immediate shrinkage followed by a hypertrophy over 90 minutes and quickly recovered when reintroduced to isotonic conditions. This effect was found to depend on the size of the increase in osmolality, as smaller increases in osmolality resulted in smaller shrinkage and hypertrophy of the MNCs. Hypertrophy was shown to be independent of cell volume regulatory processes as inhibitors of the Na+-K+-Cl- cotransporter did not affect hypertrophy. Hypertrophy was also shown to be dependent on activation of phospholipase C (PLC) and protein kinase C (PKC), as adding inhibitors of these enzymes to the hypertonic solution prevented hypertrophy. Hypertrophy could occur in isotonic conditions by inducing cell depolarization, increasing intracellular calcium ([Ca2+]i) and by activating PKC, thus showing each of these processes are involved in hypertrophy. Recovery from hypertrophy depends upon dynamin-mediated endocytosis as blocking dynamin function prevented recovery. In addition, exposing the MNCs to hypotonic solution resulted in an immediate enlargement followed by a sustained decrease in cell size. Finally, exposing acutely isolated MNCs to hypertonic solution for two hours resulted in a 37% increase in the immunolabeling of the L-type Ca2+ channel CaV1.2 subunit. This increase in CaV1.2 immunolabeling does not depend on action potential firing as adding tetrodotoxin (TTX) to the hypertonic solution failed to prevent the increase. This project will help to elucidate the mechanisms underlying this interesting example of neuroendocrine adaptation and will help us to understand the regulation of body fluid balance during chronic challenges as seen in the elderly and chronically ill.
2

Ion currents regulated by acute and chronic osmotic stimuli in rat supraoptic nucleus neurons

Zhang, Wenbo 25 February 2009
The magnocellular neurosecretory cells (MNCs) of the hypothalamus are able to change their firing rate and pattern in response to small changes in external osmolality due to the involvement of osmosensitive ion channels. The firing rate and pattern determine the release of vasopressin (VP), a primary hormone regulating osmolality by controlling water excretion from the kidney. Both VP- and oxytocin (OT)-MNCs display irregular and infrequent fire when plasma osmolality is near normal, and they progressively increase the frequency of firing to fast continuous firing with increases in osmolality. VP-MNCs also respond to osmotic stimulation by adopting a phasic pattern of firing, which maximizes neuropeptide secretion. Sustained dehydration also causes structural and functional adaptations in MNCs.<p> Voltage-dependent Ca2+ channels play many important roles not only in the regulation of cell excitability but also in intracellular signal transduction, and L-type Ca2+ channel-mediated Ca2+ signals initiate intracellular signal transduction events that activate long-lasting changes in brain function and behavior. Our electrophysiological and immunocytochemical studies demonstrate that 16-24 h of water deprivation causes a significant increase in the amplitude of L-type Ca2+ current (from 55.5 ± 6.2 to 99.1 ± 10.0 pA) but not in other types of Ca2+ current. This increase occurred in both VP- and OT-MNCs. Such an increase in L-type Ca2+ current may contribute to modulation of firing rate and pattern, regulation of vasopressin release, structural adaptation in MNCs during sustained dehydration.<p> The mechanisms underlying the transition of the electrical behaviour are not completely understood. Ion channels, especially osmosensitive ion channels, play key roles in the modulation of MNC firing. A voltage-gated, 4-AP- and TEA-insensitive slowly activating outward current displayed a significant increase in about 66% of MNCs when the osmolality of the external solution was acutely increased from 295 to 325 mosmol kg-1. The responding cells showed an increase in net outward current from 12.3 ± 1.3 pA/pF to 21.4 ± 1.8 pA/pF. The reversal potential of this current was near the equilibrium for K+ and shifted with changes of K+ concentrations in external solution, suggesting that this current is a K+-selective current. The KCNQ/M current selective blockers linopirdine (150 µM) and XE991 (5 µM) suppressed this current. The IC50 of XE991 blockade was 3.9 ìM. The KCNQ/M channel openers retigabine (10 µM) and flupirtine (10 µM) significantly increased the current and shifted its activation curve toward more negative potentials. E4031, a specific blocker of ERG K+ channels, did not significantly block this current. The results from immunocytochemistry suggest that MNCs express KCNQ2, KCNQ3, KCNQ4, and KCNQ5, but not KCNQ1. These data suggest that this osmosensitive current could be a KCNQ/M current. Studies using single unit extracellular recording in hypothalamic explants showed that 10 µM XE991 increased MNC firing rate and that 20 µM retigabine decreased firing rate or caused a cessation of firing. These data suggest that a KCNQ/M current contributes to the regulation of MNC firing. KCNQ/M channels play key roles in regulating neuronal excitability in many types of central neurons. Slow activation of this current during firing might suppress activity by hyperpolarizing the cells and thus contribute to a transition between fast continuous and burst firing.<p> Our studies will be beneficial to understand the mechanisms that control VP and OT in response to acute changes in osmolality and also the mechanisms underlying MNC adaptation during sustained dehydration.
3

Ion currents regulated by acute and chronic osmotic stimuli in rat supraoptic nucleus neurons

Zhang, Wenbo 25 February 2009 (has links)
The magnocellular neurosecretory cells (MNCs) of the hypothalamus are able to change their firing rate and pattern in response to small changes in external osmolality due to the involvement of osmosensitive ion channels. The firing rate and pattern determine the release of vasopressin (VP), a primary hormone regulating osmolality by controlling water excretion from the kidney. Both VP- and oxytocin (OT)-MNCs display irregular and infrequent fire when plasma osmolality is near normal, and they progressively increase the frequency of firing to fast continuous firing with increases in osmolality. VP-MNCs also respond to osmotic stimulation by adopting a phasic pattern of firing, which maximizes neuropeptide secretion. Sustained dehydration also causes structural and functional adaptations in MNCs.<p> Voltage-dependent Ca2+ channels play many important roles not only in the regulation of cell excitability but also in intracellular signal transduction, and L-type Ca2+ channel-mediated Ca2+ signals initiate intracellular signal transduction events that activate long-lasting changes in brain function and behavior. Our electrophysiological and immunocytochemical studies demonstrate that 16-24 h of water deprivation causes a significant increase in the amplitude of L-type Ca2+ current (from 55.5 ± 6.2 to 99.1 ± 10.0 pA) but not in other types of Ca2+ current. This increase occurred in both VP- and OT-MNCs. Such an increase in L-type Ca2+ current may contribute to modulation of firing rate and pattern, regulation of vasopressin release, structural adaptation in MNCs during sustained dehydration.<p> The mechanisms underlying the transition of the electrical behaviour are not completely understood. Ion channels, especially osmosensitive ion channels, play key roles in the modulation of MNC firing. A voltage-gated, 4-AP- and TEA-insensitive slowly activating outward current displayed a significant increase in about 66% of MNCs when the osmolality of the external solution was acutely increased from 295 to 325 mosmol kg-1. The responding cells showed an increase in net outward current from 12.3 ± 1.3 pA/pF to 21.4 ± 1.8 pA/pF. The reversal potential of this current was near the equilibrium for K+ and shifted with changes of K+ concentrations in external solution, suggesting that this current is a K+-selective current. The KCNQ/M current selective blockers linopirdine (150 µM) and XE991 (5 µM) suppressed this current. The IC50 of XE991 blockade was 3.9 ìM. The KCNQ/M channel openers retigabine (10 µM) and flupirtine (10 µM) significantly increased the current and shifted its activation curve toward more negative potentials. E4031, a specific blocker of ERG K+ channels, did not significantly block this current. The results from immunocytochemistry suggest that MNCs express KCNQ2, KCNQ3, KCNQ4, and KCNQ5, but not KCNQ1. These data suggest that this osmosensitive current could be a KCNQ/M current. Studies using single unit extracellular recording in hypothalamic explants showed that 10 µM XE991 increased MNC firing rate and that 20 µM retigabine decreased firing rate or caused a cessation of firing. These data suggest that a KCNQ/M current contributes to the regulation of MNC firing. KCNQ/M channels play key roles in regulating neuronal excitability in many types of central neurons. Slow activation of this current during firing might suppress activity by hyperpolarizing the cells and thus contribute to a transition between fast continuous and burst firing.<p> Our studies will be beneficial to understand the mechanisms that control VP and OT in response to acute changes in osmolality and also the mechanisms underlying MNC adaptation during sustained dehydration.
4

Yeast Chorismate Mutase: Molecular Evolution of an Allosteric Enzyme / Die Chorismatemutase der Bäckerhefe: Molekulare Evolution eines Allosterischen Enzyms

Helmstaedt, Kerstin 31 October 2002 (has links)
Die Chorismatmutase (CM, EC 5.4.99.5), kodiert durch ARO7, katalysiert die Claisen-Umlagerung von Chorismat zu Prephenat in der Biosynthese von Tyrosin und Phenylalanin. Das relativ kleine, dimere Enzym der Hefe Saccharomyces cerevisiae wird allosterisch durch Tryptophan aktiviert und allosterisch durch Tyrosin inhibiert. In der vorliegenden Arbeit wurde die Theorie widerlegt, dass die Chorismatemutase an der Osmoregulation und Vakuolenentstehung beteiligt ist. Die Analyse einiger Stämme mit punktmutiertem oder deletiertem ARO7-Gen zeigte ausschließlich eine Funktion in der Aminosäure-Biosynthese. Die Fusion an das grün-fluoreszierende Protein ermöglichte die Lokalisierung der CM in Cytoplasma und Kern der Hefezelle.Auf Proteinebene wurde der intramolekulare Signalübertragungsweg von den allosterischen zu den aktiven Zentren näher untersucht. Es wurden Chimären-Enzyme hergestellt, in denen das molekulare Scharnier L220s zwischen der katalytischen und allosterischen Domäne ausgetauscht wurde gegen den entsprechenden Bestandteil homologer Pilzenzyme. Die kinetische Analyse zeigte, dass dieser Proteinteil essentiell ist für die Unterscheidung zwischen dem Signal Aktivierung bzw. Inhibierung. Diese Region ist auch für die Dimerisierung der CM von Bedeutung. Durch Austausch hydrophober Aminosäuren gegen geladene Reste in und in der Nähe dieses Scharniers wurde eine stabile, monomere Enzymvariante hergestellt. Diese CM zeigte reduzierte Aktivität und keine Regulation, aber das kodierende Gen komplementierte die Tyrosin- und Phenylalanin-Auxotrophie der Zellen. Diese Ergebnisse unterstützen die Theorie, dass das Hefeenzym durch gleichzeitige Evolution von Regulations- und Stabilisierungsmechanismen aus einem monomeren, unregulierten Vorläuferprotein entstanden ist, welches dem der Escherichia coli CM ähnlich war. Um weitere Erkenntnisse über die Prinzipien der Proteinstabilisierung zu erhalten, wurde auch die Chorismatmutase on Thermus thermophilus charakterisiert, nachdem das kodierende Gen kloniert war. Dieses Enzym ist ähnlich zu der strukturell einzigartigen Chorismatmutae aus Bacillus subtilis, wird aber, im Gegensatz zu letzterem durch Tyrosin in seiner Aktivität gehemmt. Modellierungsstudien zeigten, dass wie auch bei anderen Proteinen verstärkte Hydrophilität von Oberflächen, erhöhte Hydrophobizität innerhalb der Struktur wie auch die Versteifung von Loops in der Nähe des aktiven Zentrums zur Stabilisierung dieser Proteinfaltung beitragen.
5

Control of transcription initiation by the stress activated hog1 kinase

Zapater Enrique, Meritxell 01 December 2006 (has links)
En el llevat Saccharomyces cerevisiae els canvis en les condicions osmòtiques del medi extracel.lular són sensades per la MAP cinasa Hog1, la qual permet dur a terme l'adaptació cel.lular mitjançant la modulació de l'expressió gènica, de la traducció i de la progressió del cicle cel.lular. A l'inici d'aquest projecte de tesi, els mecanismes pels quals Hog1 controla l'expressió gènica no eren del tot coneguts. El nostre objectiu va ser caracteritzar el mecanisme molecular pel qual Hog1 modula la transcripció en resposta a estrès osmòtic. Hem aconseguit demostrar que el reclutament de Hog1 als promotors sensibles a estrès osmòtic per part del factor de transcripció és essencial per al reclutament i activació de la RNA polimerasa II, mecanisme que podria estar conservat en les cèl.lules eucariotes. També hem identificat noves activitats remodeladores de cromatina implicades en la resposta gènica a osmoestrès mediada per Hog1. Vàrem realitzar un cribatge genètic per identificar mutacions que provoquessin osmosensibilitat i una reducció en l'expressió de gens de resposta a estrès osmòtic. Aquest cribatge ens va permetre identificar nous reguladors de la transcripció mediada per osmoestrès: la histona deacetilasa Rpd3 i els complexes SAGA i mediador. Els nostres resultats permeten, doncs, definir un important paper per a Rpd3, SAGA i mediador en la inducció gènica mediada per Hog1, i han estat importants per assolir una millor visió de com les cinases activades per estrès regulen la iniciació de la transcripció. / In Saccharomyces cerevisiae, changes in the extracellular osmotic conditions are sensed by the HOG MAPK pathway, which elicits the program for cell adaptation, including modulation of gene expression, translation and cell-cycle progression. At the beginning of this PhD Project, the mechanisms by which Hog1 was controlling gene transcription were not completely understood. Our main objective was to characterize the molecular mechanisms by which the Hog1 MAPK modulates transcription upon osmostress. We have shown that anchoring of Hog1 to osmoresponsive promoters by the transcription factor is essential for recruitment and activation of RNA polymerase II, a mechanism that might be conserved among eukaryotic cells. In addition, we identified novel chromatin modifying and remodelling activities involved in the Hog1-mediated osmostress gene expression. We performed a genome-wide genetic screening searching for mutations that render cells osmosensitive and displayed reduced expression of osmoresponsive genes. Rpd3 histone deacetylase, SAGA and Mediator complexes were identified as novel regulators of osmostress-mediated transcription. Thus, our results define a major role for Rpd3, SAGA and Mediator in the Hog1-mediated osmostress gene induction, and have been important to achieve a better view of how a SAPK regulates transcription initiation.

Page generated in 0.0806 seconds