Дисертації з теми "Receptor guanylyl cyclase C"

Щоб переглянути інші типи публікацій з цієї теми, перейдіть за посиланням: Receptor guanylyl cyclase C.

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся з топ-39 дисертацій для дослідження на тему "Receptor guanylyl cyclase C".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Переглядайте дисертації для різних дисциплін та оформлюйте правильно вашу бібліографію.

1

Nedvetsky, Pavel I. "Regulation of the nitric oxide receptor, soluble guanylyl cyclase." Doctoral thesis, [S.l. : s.n.], 2003. http://deposit.ddb.de/cgi-bin/dokserv?idn=969682026.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Ogawa, Yoshihisa. "Natriuretic peptide receptor guanylyl cyclase-A protects podocytes from aldosterone-induced glomerular injury." Kyoto University, 2012. http://hdl.handle.net/2433/160971.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Birnby, Deborah Ann. "Analysis of daf-11, a transmembrane guanylyl cyclase that mediates chemosensory transduction in C. elegans /." Thesis, Connect to this title online; UW restricted, 1998. http://hdl.handle.net/1773/10300.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Ter-Avetisyan, Gohar [Verfasser]. "The receptor guanylyl cyclase Npr2 regulates the bifurcation of cranial sensory axons / Gohar Ter-Avetisyan." Berlin : Freie Universität Berlin, 2013. http://d-nb.info/104244143X/34.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Nakanishi, Michio. "Role of natriuretic peptide receptor guanylyl cyclase-A in myocardial infarction evaluated using genetically engineered mice." Kyoto University, 2006. http://hdl.handle.net/2433/143854.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Morita, Rinpei. "Atrial natriuretic peptide polarizes human dendritic cells toward a Th2-promoting phenotype through its receptor guanylyl cyclase-coupled receptor-A." Kyoto University, 2003. http://hdl.handle.net/2433/148481.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Liu, Yong [Verfasser]. "Targeted guanylyl cyclase C for optimization of circulating colorectal cancer cells enrichment and isolation / Yong Liu." Berlin : Medizinische Fakultät Charité - Universitätsmedizin Berlin, 2017. http://d-nb.info/1148425500/34.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Yamashita, Yui. "Brain-specific natriuretic peptide receptor-B deletion attenuates high-fat diet-induced visceral and hepatic lipid deposition in mice." Kyoto University, 2016. http://hdl.handle.net/2433/217139.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
9

Kato, Yukiko. "Natriuretic peptide receptor guanylyl cyclase-A pathway counteracts glomerular injury evoked by aldosterone through p38 mitogen-activated protein kinase inhibition." Kyoto University, 2018. http://hdl.handle.net/2433/235033.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
10

Yasoda, Akihiro. "Natriuretic peptide regulation of endochondral ossification-Evidence for possible roles of the C-type natriuretic peptide/guanylyl cyclase-B pathway." Kyoto University, 1999. http://hdl.handle.net/2433/156998.

Повний текст джерела
Анотація:
本文データは平成22年度国立国会図書館の学位論文(博士)のデジタル化実施により作成された画像ファイルを基にpdf変換したものである
Kyoto University (京都大学)
0048
新制・課程博士
博士(医学)
甲第7759号
医博第2112号
新制||医||713(附属図書館)
UT51-99-G353
京都大学大学院医学研究科内科系専攻
(主査)教授 中西 重忠, 教授 中村 孝志, 教授 中尾 一和
学位規則第4条第1項該当
Стилі APA, Harvard, Vancouver, ISO та ін.
11

Nakao, Kazuhiro. "ENDOTHELIUM-DERIVED C-TYPE NATRIURETIC PEPTIDE CONTRIBUTES TO BLOOD PRESSURE REGULATION BY MAINTAINING ENDOTHELIAL INTEGRITY." 京都大学 (Kyoto University), 2017. http://hdl.handle.net/2433/225500.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
12

Shen, Jiaxiao. "Regulation of adenylyl cyclase 2 by protein kinase C and Gq-coupled muscarinic receptor." Thesis, University of Cambridge, 2013. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.648373.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
13

Taye, Ashraf Mohamed Abouelwafa. "Divergent effects of angiotensin II receptor types 1A and 2 on vascular functions involve NADPH oxidase-dependent oxidative stress and no-dependent guanylyl cyclase." [S.l.] : [s.n.], 2005. http://deposit.ddb.de/cgi-bin/dokserv?idn=974104493.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
14

Wolter, Steffen [Verfasser], and Jan [Akademischer Betreuer] Benda. "Loss of function mutation of the receptor guanylyl cyclase B (GC-B) leads to changes in features of auditory processing / Steffen Wolter ; Betreuer: Jan Benda." Tübingen : Universitätsbibliothek Tübingen, 2019. http://d-nb.info/1197611037/34.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
15

Guo, Beichu. "Interaction of PKCbeta with CARMA1 mediates B cell receptor-induced NF-kappaB activation /." Thesis, Connect to this title online; UW restricted, 2003. http://hdl.handle.net/1773/8348.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
16

Welshhans, Kristy. "Neuronal growth cone dynamics are regulated by a nitric oxide-initiated second messenger pathway." unrestricted, 2007. http://etd.gsu.edu/theses/available/etd-09282007-114034/.

Повний текст джерела
Анотація:
Thesis (Ph. D.)--Georgia State University, 2007.
Vincent Rehder, committee chair; Sarah Pallas, Walter William Walthall, committee members. Electronic text (248 p. : ill. (some col.)) : digital, PDF file. Description based on contents viewed Jan. 28, 2008; title from file title page. Includes bibliographical references (p. 218-248).
Стилі APA, Harvard, Vancouver, ISO та ін.
17

Dankworth, Beatrice [Verfasser], та Michaela [Akademischer Betreuer] Kuhn. "Charakterisierung der dynamischen Interaktion des Guanylyl Cyclase-A (GC-A)-Rezeptors mit den Transient Receptor Potential Canonical Type 3 und Type 6 (TRPC3/C6)-Kanälen und Generierung von β-Zell-spezifischen GC-A-knock-out-Mäusen sowie die Analyse der Bedeutung von ANP für die Insulin-Homöostase unter pathophysiologischen Bedingungen / Beatrice Dankworth. Betreuer: Michaela Kuhn". Würzburg : Universitätsbibliothek der Universität Würzburg, 2013. http://d-nb.info/1036836495/34.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
18

Bengtsson, Magnus Wilhelm. "Effects of Orexins, Guanylins and Feeding on Duodenal Bicarbonate Secretion and Enterocyte Intracellular Signaling." Doctoral thesis, Uppsala University, Department of Neuroscience, 2008. http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-8664.

Повний текст джерела
Анотація:

The duodenal epithelium secretes bicarbonate ions and this is regarded as the primary defence mechanism against the acid discharged from the stomach. For an efficient protection, the duodenum must also function as a sensory organ identifying luminal factors. Enteroendocrine cells are well-established intestinal “taste” cells that express signaling peptides such as orexins and guanylins. Luminal factors affect the release of these peptides, which may modulate the activity of nearby epithelial and neural cells.

The present thesis considers the effects of orexins and guanylins on duodenal bicarbonate secretion. The duodenal secretory response to the peptides was examined in anaesthetised rats in situ and the effects of orexin-A on intracellular calcium signaling by human as well as rat duodenal enterocytes were studied in vitro.

Orexin-A, guanylin and uroguanylin were all stimulants of bicarbonate secretion. The stimulatory effect of orexin-A was inhibited by the OX1-receptor selective antagonist SB-334867. The muscarinic antagonist atropine on the other hand, did not affect the orexin-A-induced secretion, excluding involvement of muscarinic receptors. Orexin-A induced calcium signaling in isolated duodenocytes suggesting a direct effect at these cells. Interestingly, orexin-induced secretion and calcium signaling as well as mucosal orexin-receptor mRNA and OX1-receptor protein levels were all substantially downregulated in overnight fasted rats compared with animals with continuous access to food. Further, secretion induced by Orexin-A was shown to be dependent on an extended period of glucose priming.

The uroguanylin-induced bicarbonate secretion was reduced by atropine suggesting involvement of muscarinic receptors. The melatonin receptor antagonist luzindole attenuated the secretory response to intra-arterially administered guanylins but had no effect on secretion when the guanylins were given luminally.

In conclusion, the results suggest that orexin-A as well as guanylins may participate in the regulation of duodenal bicarbonate secretion. Further, the duodenal orexin system is dependent on the feeding status of the animals.

Стилі APA, Harvard, Vancouver, ISO та ін.
19

Arshad, Najla. "Guanylyl Cyclase C Regulation And Pathophysiology." Thesis, 2011. https://etd.iisc.ac.in/handle/2005/2439.

Повний текст джерела
Анотація:
The survival of the any living organism depends on its availability to communicate, and a breakdown of cellular signaling can have dire consequences such as uncontrolled cellular proliferations or even cell death. Environmental cues or ligands are perceived by cognate receptors, expressed primarily on the cell surface, and transmitted to the interior of the cell to elicit a response. This universal phenomenon is termed as signal transduction. During this process, second messengers such as cyclic nucleotides, cAMP and cGMP, are produced which serve to amplify the signal. Cyclic GMP is emerging as a universal second messenger, and is found in both prokaryotes and eukaryotes. It is synthesized from GTP by the action of guanylyl cyclases. Vertebrate guanylyl cyclases are of two forms, soluble and membrane-associated. Soluble guanylyl cyclases are heterodimeric enzymes which are activated by nitic oxide. Membrane-associated guanylyl cyclases on the other hand are homodimeric enzymes that act as receptors for divers polypeptide ligands. In mammals, there are seven isoforms of receptors guanylyl cyclase, GC-A through GC-G. These recptors have a highly conseved modular domin organization with an N-terminal extracellular domain, a single transmembrane domain and a C- terminal intra cellular regions. The intracellular region contains a juxtamembrane domain followed by a protein-kinase domain, a linker region and a catalytic guanylyl cyclase domain. The coordinated actions of these domains ensure fine tuned-regulations of the catalytic domain. Guanylyl cyclase-C (GC-C) is a member of the membrane-bound guanylyl cyclases. GC-C is predominantely present in the intestine, on the apical surface of epithelial cells, but has also been detected in the rat epididymis. In the intestine it serves as the guanylin, uroguanylin and lymphoguanylin which are the endogenous peptide ligands, while heat- stable entrotoxins (ST) peptides secreted by enterotoxigenic E.coil, are exogenic ligands. Activation of GC-C by these ligands results in an increase in intracellular cGMP levels, which then activates cGMP-dependent protein kinase and cross-activates protein kinase A. In turn, these activated kinases phosphorylate and active the cystic fibrosis transmembrane conductance regulator (CFTR), resulting in chloride and water secretion into the intestine lumen, thus regulating salt and water homeostasis in the intestine. ST peptide has a greater affinity for GC-C than the endogenous ligands and activation of GC-C by ST results in masiive fluid and ion efflux from the intestine cells from which manifests as Travelers’ Diarrhea. The GC-C mediated cGMP signal transduction pathway also maintains intestinal crypt-villus axis homeostatis by exerting a cytostatic effect on the epithelial cells, there by regulating their turn over. Over the years multiple mechanisms of regulation of GC-C activity has been identified including allosteric controlled by various domains in the receptor and phosphorylation-mediated regulation of guanylyl cyclase activity. The current study describes aspects of the regulation of GC-C by gycosylation, and also reports the molecular phenotypes of a naturally occurring mutation in GC-C causes sever diarrhea in affected individuals. GC-C is expressed as a differentially glycosylated protein (130k Da and 145kDa). While both forms bind with equal affinity, only one the 145 kDa form is activated by its ligands. It is this higher glycosylated form which is selectively downregulated in the process of decensitization of GC-C in colomn carcinoma cells (Caco2). Give the critical role gycosylation plays in protein folding, trafficking, receptor activity and mediating protein inter actions, its influence on GC-C was analysed.
Стилі APA, Harvard, Vancouver, ISO та ін.
20

Arshad, Najla. "Guanylyl Cyclase C Regulation And Pathophysiology." Thesis, 2011. http://etd.iisc.ernet.in/handle/2005/2439.

Повний текст джерела
Анотація:
The survival of the any living organism depends on its availability to communicate, and a breakdown of cellular signaling can have dire consequences such as uncontrolled cellular proliferations or even cell death. Environmental cues or ligands are perceived by cognate receptors, expressed primarily on the cell surface, and transmitted to the interior of the cell to elicit a response. This universal phenomenon is termed as signal transduction. During this process, second messengers such as cyclic nucleotides, cAMP and cGMP, are produced which serve to amplify the signal. Cyclic GMP is emerging as a universal second messenger, and is found in both prokaryotes and eukaryotes. It is synthesized from GTP by the action of guanylyl cyclases. Vertebrate guanylyl cyclases are of two forms, soluble and membrane-associated. Soluble guanylyl cyclases are heterodimeric enzymes which are activated by nitic oxide. Membrane-associated guanylyl cyclases on the other hand are homodimeric enzymes that act as receptors for divers polypeptide ligands. In mammals, there are seven isoforms of receptors guanylyl cyclase, GC-A through GC-G. These recptors have a highly conseved modular domin organization with an N-terminal extracellular domain, a single transmembrane domain and a C- terminal intra cellular regions. The intracellular region contains a juxtamembrane domain followed by a protein-kinase domain, a linker region and a catalytic guanylyl cyclase domain. The coordinated actions of these domains ensure fine tuned-regulations of the catalytic domain. Guanylyl cyclase-C (GC-C) is a member of the membrane-bound guanylyl cyclases. GC-C is predominantely present in the intestine, on the apical surface of epithelial cells, but has also been detected in the rat epididymis. In the intestine it serves as the guanylin, uroguanylin and lymphoguanylin which are the endogenous peptide ligands, while heat- stable entrotoxins (ST) peptides secreted by enterotoxigenic E.coil, are exogenic ligands. Activation of GC-C by these ligands results in an increase in intracellular cGMP levels, which then activates cGMP-dependent protein kinase and cross-activates protein kinase A. In turn, these activated kinases phosphorylate and active the cystic fibrosis transmembrane conductance regulator (CFTR), resulting in chloride and water secretion into the intestine lumen, thus regulating salt and water homeostasis in the intestine. ST peptide has a greater affinity for GC-C than the endogenous ligands and activation of GC-C by ST results in masiive fluid and ion efflux from the intestine cells from which manifests as Travelers’ Diarrhea. The GC-C mediated cGMP signal transduction pathway also maintains intestinal crypt-villus axis homeostatis by exerting a cytostatic effect on the epithelial cells, there by regulating their turn over. Over the years multiple mechanisms of regulation of GC-C activity has been identified including allosteric controlled by various domains in the receptor and phosphorylation-mediated regulation of guanylyl cyclase activity. The current study describes aspects of the regulation of GC-C by gycosylation, and also reports the molecular phenotypes of a naturally occurring mutation in GC-C causes sever diarrhea in affected individuals. GC-C is expressed as a differentially glycosylated protein (130k Da and 145kDa). While both forms bind with equal affinity, only one the 145 kDa form is activated by its ligands. It is this higher glycosylated form which is selectively downregulated in the process of decensitization of GC-C in colomn carcinoma cells (Caco2). Give the critical role gycosylation plays in protein folding, trafficking, receptor activity and mediating protein inter actions, its influence on GC-C was analysed.
Стилі APA, Harvard, Vancouver, ISO та ін.
21

Mahaboobi, *. "Receptor Guanylyl Cyclase C : Insights Into Expression And Regulation." Thesis, 2004. https://etd.iisc.ac.in/handle/2005/1287.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
22

Mahaboobi, *. "Receptor Guanylyl Cyclase C : Insights Into Expression And Regulation." Thesis, 2004. http://etd.iisc.ernet.in/handle/2005/1287.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
23

Basu, Nirmalya. "Insights Into Cytostatic Mechanisms Regulated By Receptor Guanylyl Cyclase C." Thesis, 2012. https://etd.iisc.ac.in/handle/2005/2590.

Повний текст джерела
Анотація:
All cells are equipped to sense changes in their environment and make adaptive responses according to the stimuli. Signal recognition usually occurs at the cell membrane (with the exception of steroid signalling) where the ligand, which can be a small molecule, a peptide or a protein, binds its cognate receptor. This results in a change in the conformation of the receptor which in turn can regulate the production of second messengers. Second messengers can now modulate specific pathways which control gene expression and modify various aspects of cell behaviour. The signalling cascade is terminated by the removal of second messenger and/or by desensitisation of the receptor to the extracellular signal. Cyclic guanosine monophosphate (cGMP) was first identified in the rat urine and since then has emerged as an important second messenger regulating diverse cell processes. Subsequent to its discovery in mammalian cells, enzymes responsible for its synthesis (guanylyl cyclases), hydrolysis (phosphodiesterases) and its most common effectors (cGMP-dependent protein kinases) were identified. Guanylyl cyclases exist in two forms, cytosolic and membrane bound. Both have a conserved guanylyl cyclase domain, but differ in their choice of ligands, overall structure and tissue localization. It is now known that cytosolic and the membrane-bound forms are involved in eliciting distinct cellular responses. Receptor guanylyl cyclase C (GC-C) was identified as the target for a family of heat-stable enterotoxin toxins (ST) produced by enterotoxigenic E.coli. Stable toxin-mediated diarrhoeas are observed frequently in infants and contribute significantly to the incidence of Travellers’ Diarrhea. Early studies demonstrated that the effects of ST were mediated by an increase in intracellular cGMP levels in intestinal cells, and the receptor for ST was almost exclusively expressed in the apical microvilli of the intestinal brush-border epithelia. Effectors of cGMP in intestinal cells include protein kinase G (PKG), cyclic nucleotide gated ion channel 3 (CNG), and the cystic fibrosis transmembrane conductance regulator (CFTR). ST is an exogenous ligand which serves as a hyperagonist for GC-C, in comparison with the endogenous ligands guanylin and uroguanylin, which maintain fluid-ion homeostasis in the intestinal epithelia. The GC-C/cGMP signal transduction pathway also modulates intestinal cell proliferation along the crypt-villus axis by exerting a cytostatic effect on the epithelial cells, thereby regulating their turnover and neoplastic transformation. The current study describes in molecular detail two signalling pathways, one impinging on and one emerging from GC-C, which regulate colonic cell proliferation. The first part identifies the cross-talk and cross-regulation of GC-C and c-src. The second part delves into the molecular basis of GC-C/cGMP-mediated cytostasis and its effect on colonic tumorigenesis. Cross-talk between signalling pathways is believed to play a key role in regulating cell physiology. Phosphorylation of signalling molecules by protein kinases is frequently used as a means of achieving this cross-regulation. Aberrant hyperactivation of the c-src tyrosine kinase is an early event in the progression of colorectal cancer, and activated c-src specifically phosphorylates a number of proteins in the cell. It was found that c-src can phosphorylate GC-C in T84 colorectal carcinoma cells, as well as in the rat intestinal epithelia. Tyrosine phosphorylation of GC-C resulted in attenuation of ligand-mediated cGMP production; an effect which was reversed by chemical or transcriptional knockdown of c-src. These effects were found to be cell line-independent and relied only on the extent of c-src expression and activation in the cell. Mutational analysis revealed GC-C to be phosphorylated on a conserved tyrosine residue (Y820) in the guanylyl cyclase domain. The sequence of GC-C around Y820 allowed for efficient phosphorylation by c-src, and indeed, kinase assays indicated that the affinity of c-src for the GC-C Y820 peptide was one of the highest reported till date. A phospho-mimetic mutation at this site, which mimics a constitutively phosphorylated receptor, resulted in a sharp reduction of guanylyl cyclase activity of the receptor, reiterating the inhibitory role of Y820 phosphorylation on GC-C activity. Phosphorylation of GC-C at Y820 generated a docking site for the SH2 domain of c-src which could interact and thereby co-localize with GC-C on the cell membrane. Intriguingly, this interaction resulted in activation of c-src, setting-up a feed-forward loop of inhibitory GC-C phosphorylation and c-src activation. Treatment of colorectal carcinoma cells with ligands for GC-C reduces cell proliferation and inhibits tumorigenesis. It was observed that this cytostatic effect can be modulated by the status of c-src activation, and consequently, the fraction of tyrosine phosphorylated GC-C in these cells. Since activation of c-src is a frequent event in intestinal neoplasia, phosphorylation of GC-C by active c-src may be one of the means by which the cytostatic effects of GC-C agonists (guanylin and uroguanylin) in the intestine are bypassed, thereby leading to cancer progression. Colonisation of the gut with enteropathogenic microorganisms induces secretion of IFNγ from the host mucosal immune system, which subsequently activates c-src in intestinal epithelial cells. Ligand-stimulated activity of GC-C was found to be reduced in IFNγ treated cells. This could be one of the host defence mechanisms initiated in response to enterotoxigenic E. coli infection. These results provide the first evidence of cross-talk between a receptor guanylyl cyclase and a tyrosine kinase that results in heterologous desensitisation of the receptor. Populations with a higher incidence of enterotoxigenic E.coli infections appear to be protected from intestinal neoplasia. It was found that mice lacking GC-C, and therefore unable to respond to ST, displayed an increased cell proliferation in colonic crypts and enhanced carcinogen-induced aberrant crypt foci formation, which is a surrogate marker for colorectal carcinogenesis. However, pharmacological elevation of cGMP was able to efficiently induce cytostasis even in GC-C knockout mice, indicating a key role for cGMP in regulating colonic cell proliferation. Through microarray analyses, genes regulated by ST-induced GC-C activation in T84 colorectal carcinoma cells were identified. Genes involved in a number of cellular pathways were differentially expressed, including those involved in signal transduction, protein and solute secretion, transcriptional regulation and extracellular matrix formation. One of the genes found to be significantly up-regulated was the cell-cycle inhibitor, p21. The increase in p21 expression was validated at both the transcript and protein level. This p53-independent up-regulation of p21 was coupled to the activation of the cGMP-responsive kinase, PKGII, since knockdown of PKGII using specific siRNAs abolished ST-induced p21 induction. Activation of PKGII led to phosphorylation and activation of the stress responsive p38 MAPK. Similar to what was seen following knockdown of PKGII, inhibition of p38 MAPK activity attenuated the up-regulation of p21 in response to cGMP, indicating that PKGII and p38 MAPK could be a part of a pathway regulating p21 expression. It was found that active p38 MAPK phosphorylated the ubiquitous transcription factor SP1, enhancing its occupancy at the proximal p21 promoter. Therefore, SP1 could be one of the factors linking cGMP to transcription of the p21 mRNA. Chronic activation of GC-C led to nuclear accumulation of p21 in colonic cells, which entered a quiescent state. These cells arrested in the G1 phase of the cell cycle, consequent to p21-dependent inhibition of the G1 cyclin-CDK complexes. A fraction of these quiescent cells stochastically initiated a cGMP-dependent senescence programme and displayed all the hallmarks of senescent cells, including flattened cell morphology, expression of SA- galactosidase and formation of senescence-associated heterochromatic foci. Activation of senescence and loss of tumorigenicity in these cells was crucially dependent on the up-regulation of p21. This irreversible exit from the cell cycle due to cGMP-mediated activation of the PKGII/p38/p21 axis was well correlated with reduced colonic polyp formation in mice exposed to ST. In summary, these observations may provide a possible explanation for the low incidence of colorectal carcinoma seen in countries with a high incidence of ST-mediated diarrhoea. Interestingly, c-src mediated tyrosine phosphorylation of GC-C prevented p21 accumulation following ligand application. The findings described in this thesis may have important implications in understanding the molecular mechanisms involved in the progression and treatment of colorectal cancer.
Стилі APA, Harvard, Vancouver, ISO та ін.
24

Basu, Nirmalya. "Insights Into Cytostatic Mechanisms Regulated By Receptor Guanylyl Cyclase C." Thesis, 2012. http://etd.iisc.ernet.in/handle/2005/2590.

Повний текст джерела
Анотація:
All cells are equipped to sense changes in their environment and make adaptive responses according to the stimuli. Signal recognition usually occurs at the cell membrane (with the exception of steroid signalling) where the ligand, which can be a small molecule, a peptide or a protein, binds its cognate receptor. This results in a change in the conformation of the receptor which in turn can regulate the production of second messengers. Second messengers can now modulate specific pathways which control gene expression and modify various aspects of cell behaviour. The signalling cascade is terminated by the removal of second messenger and/or by desensitisation of the receptor to the extracellular signal. Cyclic guanosine monophosphate (cGMP) was first identified in the rat urine and since then has emerged as an important second messenger regulating diverse cell processes. Subsequent to its discovery in mammalian cells, enzymes responsible for its synthesis (guanylyl cyclases), hydrolysis (phosphodiesterases) and its most common effectors (cGMP-dependent protein kinases) were identified. Guanylyl cyclases exist in two forms, cytosolic and membrane bound. Both have a conserved guanylyl cyclase domain, but differ in their choice of ligands, overall structure and tissue localization. It is now known that cytosolic and the membrane-bound forms are involved in eliciting distinct cellular responses. Receptor guanylyl cyclase C (GC-C) was identified as the target for a family of heat-stable enterotoxin toxins (ST) produced by enterotoxigenic E.coli. Stable toxin-mediated diarrhoeas are observed frequently in infants and contribute significantly to the incidence of Travellers’ Diarrhea. Early studies demonstrated that the effects of ST were mediated by an increase in intracellular cGMP levels in intestinal cells, and the receptor for ST was almost exclusively expressed in the apical microvilli of the intestinal brush-border epithelia. Effectors of cGMP in intestinal cells include protein kinase G (PKG), cyclic nucleotide gated ion channel 3 (CNG), and the cystic fibrosis transmembrane conductance regulator (CFTR). ST is an exogenous ligand which serves as a hyperagonist for GC-C, in comparison with the endogenous ligands guanylin and uroguanylin, which maintain fluid-ion homeostasis in the intestinal epithelia. The GC-C/cGMP signal transduction pathway also modulates intestinal cell proliferation along the crypt-villus axis by exerting a cytostatic effect on the epithelial cells, thereby regulating their turnover and neoplastic transformation. The current study describes in molecular detail two signalling pathways, one impinging on and one emerging from GC-C, which regulate colonic cell proliferation. The first part identifies the cross-talk and cross-regulation of GC-C and c-src. The second part delves into the molecular basis of GC-C/cGMP-mediated cytostasis and its effect on colonic tumorigenesis. Cross-talk between signalling pathways is believed to play a key role in regulating cell physiology. Phosphorylation of signalling molecules by protein kinases is frequently used as a means of achieving this cross-regulation. Aberrant hyperactivation of the c-src tyrosine kinase is an early event in the progression of colorectal cancer, and activated c-src specifically phosphorylates a number of proteins in the cell. It was found that c-src can phosphorylate GC-C in T84 colorectal carcinoma cells, as well as in the rat intestinal epithelia. Tyrosine phosphorylation of GC-C resulted in attenuation of ligand-mediated cGMP production; an effect which was reversed by chemical or transcriptional knockdown of c-src. These effects were found to be cell line-independent and relied only on the extent of c-src expression and activation in the cell. Mutational analysis revealed GC-C to be phosphorylated on a conserved tyrosine residue (Y820) in the guanylyl cyclase domain. The sequence of GC-C around Y820 allowed for efficient phosphorylation by c-src, and indeed, kinase assays indicated that the affinity of c-src for the GC-C Y820 peptide was one of the highest reported till date. A phospho-mimetic mutation at this site, which mimics a constitutively phosphorylated receptor, resulted in a sharp reduction of guanylyl cyclase activity of the receptor, reiterating the inhibitory role of Y820 phosphorylation on GC-C activity. Phosphorylation of GC-C at Y820 generated a docking site for the SH2 domain of c-src which could interact and thereby co-localize with GC-C on the cell membrane. Intriguingly, this interaction resulted in activation of c-src, setting-up a feed-forward loop of inhibitory GC-C phosphorylation and c-src activation. Treatment of colorectal carcinoma cells with ligands for GC-C reduces cell proliferation and inhibits tumorigenesis. It was observed that this cytostatic effect can be modulated by the status of c-src activation, and consequently, the fraction of tyrosine phosphorylated GC-C in these cells. Since activation of c-src is a frequent event in intestinal neoplasia, phosphorylation of GC-C by active c-src may be one of the means by which the cytostatic effects of GC-C agonists (guanylin and uroguanylin) in the intestine are bypassed, thereby leading to cancer progression. Colonisation of the gut with enteropathogenic microorganisms induces secretion of IFNγ from the host mucosal immune system, which subsequently activates c-src in intestinal epithelial cells. Ligand-stimulated activity of GC-C was found to be reduced in IFNγ treated cells. This could be one of the host defence mechanisms initiated in response to enterotoxigenic E. coli infection. These results provide the first evidence of cross-talk between a receptor guanylyl cyclase and a tyrosine kinase that results in heterologous desensitisation of the receptor. Populations with a higher incidence of enterotoxigenic E.coli infections appear to be protected from intestinal neoplasia. It was found that mice lacking GC-C, and therefore unable to respond to ST, displayed an increased cell proliferation in colonic crypts and enhanced carcinogen-induced aberrant crypt foci formation, which is a surrogate marker for colorectal carcinogenesis. However, pharmacological elevation of cGMP was able to efficiently induce cytostasis even in GC-C knockout mice, indicating a key role for cGMP in regulating colonic cell proliferation. Through microarray analyses, genes regulated by ST-induced GC-C activation in T84 colorectal carcinoma cells were identified. Genes involved in a number of cellular pathways were differentially expressed, including those involved in signal transduction, protein and solute secretion, transcriptional regulation and extracellular matrix formation. One of the genes found to be significantly up-regulated was the cell-cycle inhibitor, p21. The increase in p21 expression was validated at both the transcript and protein level. This p53-independent up-regulation of p21 was coupled to the activation of the cGMP-responsive kinase, PKGII, since knockdown of PKGII using specific siRNAs abolished ST-induced p21 induction. Activation of PKGII led to phosphorylation and activation of the stress responsive p38 MAPK. Similar to what was seen following knockdown of PKGII, inhibition of p38 MAPK activity attenuated the up-regulation of p21 in response to cGMP, indicating that PKGII and p38 MAPK could be a part of a pathway regulating p21 expression. It was found that active p38 MAPK phosphorylated the ubiquitous transcription factor SP1, enhancing its occupancy at the proximal p21 promoter. Therefore, SP1 could be one of the factors linking cGMP to transcription of the p21 mRNA. Chronic activation of GC-C led to nuclear accumulation of p21 in colonic cells, which entered a quiescent state. These cells arrested in the G1 phase of the cell cycle, consequent to p21-dependent inhibition of the G1 cyclin-CDK complexes. A fraction of these quiescent cells stochastically initiated a cGMP-dependent senescence programme and displayed all the hallmarks of senescent cells, including flattened cell morphology, expression of SA- galactosidase and formation of senescence-associated heterochromatic foci. Activation of senescence and loss of tumorigenicity in these cells was crucially dependent on the up-regulation of p21. This irreversible exit from the cell cycle due to cGMP-mediated activation of the PKGII/p38/p21 axis was well correlated with reduced colonic polyp formation in mice exposed to ST. In summary, these observations may provide a possible explanation for the low incidence of colorectal carcinoma seen in countries with a high incidence of ST-mediated diarrhoea. Interestingly, c-src mediated tyrosine phosphorylation of GC-C prevented p21 accumulation following ligand application. The findings described in this thesis may have important implications in understanding the molecular mechanisms involved in the progression and treatment of colorectal cancer.
Стилі APA, Harvard, Vancouver, ISO та ін.
25

Rasool, Insha. "Molecular Phenotyping of Mutations in Guanylyi Cyclase C Associated with Congenital Diarrhea." Thesis, 2014. http://etd.iisc.ac.in/handle/2005/3192.

Повний текст джерела
Анотація:
Guanylyl cyclase C (GC-C) is a member of particulate guanylyl cyclases, discovered primarily as the target of a family of heat stable enterotoxins (ST), produced by enterotoxigenic Escherichia coli (ETEC). ST is acknowledged as a prime cause of traveller’s diarrhea and the leading cause of child mortality under the age of 5 years in developing nations. The bacterial expression of ST peptides represents molecular mimicry where the pathogen has exploited a gastrointestinal tract-signaling pathway to disperse and propagate. GC-C is primarily expressed on the apical or the brush border membranes of intestinal epithelial cells. GC-C agonists elaborated in the gastrointestinal tract are a family of guanylin peptides, which are responsible for maintaining fluid-ion homeostasis, essential for normal gut physiology. The signal of liigand binding to the extracellular domain of GC-C is transduced to the catalytic guanylyl cyclase domain, which results in production of intracellular cGMP. The elevated levels of cGMP influence multiple downstream targets, which finally regulate ion-flux through the transporters present on the membrane of an enterocyte. The ST peptide, a GC-C superagonist, produces physiologically abnormal levels of cGMP that manifest as secretory diarrhea. The purview of GC-C misregulation was confined to the notion of its hyperactivation caused by ETEC infection and the ensuing diarrhea. Recently, two seminal studies widened the scope of pathologies associated with GC-C. Studies described point mutations in GUCY2C, which were associated with human disease. One study identified a Norwegian family whose members demonstrated a dominantly inherited syndrome of frequent diarrhea associated with hyperactive GC-C. Following this study, inactivating mutations in GC-C in a small Bedouin population was reported. The current study reports the molecular phenotypes associated with the first germ line mutations in GC-C that result in a severe form of congenital sodium diarrhea. Our collaborators from Austria (Thomas Muller & Andreas Janecke, Department of Pediatrics Innsbruck Medical University) communicated to us their study of patients who had clinical diagnosis of congenital sodium diarrhea, with proportionally high fecal sodium loss, metabolic acidosis and dehydration. Exome sequencing in a cohort of 6 unrelated patients revealed four heterozygous missense mutations in GC-C (R792S, L775P, K507E, N850D). Novel GC-C mutations were de novo spontaneous mutations with the carrier being the only affected family member in contrast to the previous two reports with familial history. Biochemical characterization revealed that the mutants (GC-CR792S, GC-CL775P) were constitutively active with GC-CR792S, GC-CK507E, and GC-CN850D showing further stimulation upon treatment with ST and guanylin family of peptides. Interestingly, there was no change in the binding affinities of the ligands for the mutant receptors compared to wild type. However, a significant decrease (ranging from 10-100 fold) in ligand EC50 for the mutant GC-C receptors was prominent. The in vitro assays suggested that the mutations occupying different domains of GC-C might have resulted in distinct structural consequences reflected in the repertoire of phenotypes that were observed. The results presented in this thesis illustrate the molecular basis of the severe form of congenital diarrhea associated with the GC-C gain-of-function mutations. This study has also elaborated our understanding of the regulation of GC-C activity by its various domains.
Стилі APA, Harvard, Vancouver, ISO та ін.
26

Rasool, Insha. "Molecular Phenotyping of Mutations in Guanylyi Cyclase C Associated with Congenital Diarrhea." Thesis, 2014. http://hdl.handle.net/2005/3192.

Повний текст джерела
Анотація:
Guanylyl cyclase C (GC-C) is a member of particulate guanylyl cyclases, discovered primarily as the target of a family of heat stable enterotoxins (ST), produced by enterotoxigenic Escherichia coli (ETEC). ST is acknowledged as a prime cause of traveller’s diarrhea and the leading cause of child mortality under the age of 5 years in developing nations. The bacterial expression of ST peptides represents molecular mimicry where the pathogen has exploited a gastrointestinal tract-signaling pathway to disperse and propagate. GC-C is primarily expressed on the apical or the brush border membranes of intestinal epithelial cells. GC-C agonists elaborated in the gastrointestinal tract are a family of guanylin peptides, which are responsible for maintaining fluid-ion homeostasis, essential for normal gut physiology. The signal of liigand binding to the extracellular domain of GC-C is transduced to the catalytic guanylyl cyclase domain, which results in production of intracellular cGMP. The elevated levels of cGMP influence multiple downstream targets, which finally regulate ion-flux through the transporters present on the membrane of an enterocyte. The ST peptide, a GC-C superagonist, produces physiologically abnormal levels of cGMP that manifest as secretory diarrhea. The purview of GC-C misregulation was confined to the notion of its hyperactivation caused by ETEC infection and the ensuing diarrhea. Recently, two seminal studies widened the scope of pathologies associated with GC-C. Studies described point mutations in GUCY2C, which were associated with human disease. One study identified a Norwegian family whose members demonstrated a dominantly inherited syndrome of frequent diarrhea associated with hyperactive GC-C. Following this study, inactivating mutations in GC-C in a small Bedouin population was reported. The current study reports the molecular phenotypes associated with the first germ line mutations in GC-C that result in a severe form of congenital sodium diarrhea. Our collaborators from Austria (Thomas Muller & Andreas Janecke, Department of Pediatrics Innsbruck Medical University) communicated to us their study of patients who had clinical diagnosis of congenital sodium diarrhea, with proportionally high fecal sodium loss, metabolic acidosis and dehydration. Exome sequencing in a cohort of 6 unrelated patients revealed four heterozygous missense mutations in GC-C (R792S, L775P, K507E, N850D). Novel GC-C mutations were de novo spontaneous mutations with the carrier being the only affected family member in contrast to the previous two reports with familial history. Biochemical characterization revealed that the mutants (GC-CR792S, GC-CL775P) were constitutively active with GC-CR792S, GC-CK507E, and GC-CN850D showing further stimulation upon treatment with ST and guanylin family of peptides. Interestingly, there was no change in the binding affinities of the ligands for the mutant receptors compared to wild type. However, a significant decrease (ranging from 10-100 fold) in ligand EC50 for the mutant GC-C receptors was prominent. The in vitro assays suggested that the mutations occupying different domains of GC-C might have resulted in distinct structural consequences reflected in the repertoire of phenotypes that were observed. The results presented in this thesis illustrate the molecular basis of the severe form of congenital diarrhea associated with the GC-C gain-of-function mutations. This study has also elaborated our understanding of the regulation of GC-C activity by its various domains.
Стилі APA, Harvard, Vancouver, ISO та ін.
27

Vivek, T. N. "Receptor Guanylyl Cyclase C Cross-talk With Tyrosine Kinases And The Adaptor Protein, Crk." Thesis, 2006. https://etd.iisc.ac.in/handle/2005/412.

Повний текст джерела
Анотація:
Signal transduction is a crucial event that enables cells to sense and respond to cues from their immediate environment. Guanylyl cyclase C (GC-C) is a member of the family of receptor guanylyl cyclases. GC-C is a single transmembrane protein that responds to its ligands by the production of the second messenger cGMP. The guanylin family of peptides, (including the bacterially produced heat-stable enterotoxin ST) is the ligand for GC-C, elevates intracellular cGMP levels and activates downstream pathways. GC-C regulates the cystic fibrosis transmembrane conductance regulator (CFTR) by inducing phosphorylation by protein kinase G, resulting in chloride ion and fluid efflux. GC-C also regulates cell cycle progression through cGMP-gated Ca2+ channels. These functions are seen in the intestinal epithelium, the primary site for GC-C expression. GC-C as a molecule has been studied in detail, but its functioning in the context of other signaling pathways remains unknown. The aim of the present investigation was to understand the regulation of signal transduction by GC-C and its cross-talk with other signaling pathways operating in the cell. Molecular events that commonly connect components in a signaling pathway are protein phosphorylation and protein-protein interaction. These two aspects are explored in this thesis. The possibility of tyrosine phosphorylation of GC-C has been explored earlier in our laboratory. In vitro studies indicated that the residue Tyr820 was a site for phosphorylation by the Src family of non-receptor tyrosine kinases and those studies also suggested that phosphorylated Tyr820 could bind to the SH2 domain of Src. We generated a nonphosphorylatable mutant of GC-C, GC-CY820F, and a phosphomimetic mutant GC-CY820E to study the effect of phosphorylation of Tyr820, on the functioning of GC-C. A stable cell line of HEK293:GC-CY820F cells was generated and compared with HEK293:GC-CWT. Dose response to ST in the two cell lines showed that cGMP accumulation by GC-CY820F was greater than that of GC-CWT, although the EC50 remained unchanged. The phosphomimetic GC-CY820E mutant receptor was non-responsive to ST. Further in HEK293 cells, phosphorylation of GC-CWT by constitutively active v-Src resulted in decreased ST stimulation and this effect of v-Src was reduced with GC-CY820F. Inhibition of ST stimulation brought about by v-Src required catalytically active Src, as the kinase inactive v-SrcK295R did not inhibit ST stimulation. These results were corroborated by in vitro studies by using the recombinant catalytic domain of GC-C expressed in insect cells and by phosphorylation using a purified kinase, Hck. Observations suggested that phosphorylation of Tyr820 in the catalytic domain of GC-C compromises the guanylyl cyclase activity of GC-C. T84 and Caco-2 colon carcinoma cells endogenously express GC-C. The effect of tyrosine phosphorylation of GC-C was studied by using HgCl2, a known activator of Src kinases, and by the inhibition of protein tyrosine phosphatases using pervanadate, an irreversible inhibitor. Both these ways of achieving increased tyrosine phosphorylation resulted in decreased ST-stimulated cGMP production by GC-C, as suggested from v-Src transfection studies. This decrease was reversed by using a Src kinase specific inhibitor PP2, confirming the role of Src kinases in the inhibition of GC-C activity. Interestingly, in Caco-2 cells that differentiate in culture, the effect of pervanadate on the inhibition of ST-stimulated GC-C activation was dependent on the differentiation stage. Crypt-like cells showed higher inhibition with pervanadate. As they matured into villus-like cells, the effect of pervanadate on GC-C activation was gradually lost. This effect also correlated with a decrease in the expression of Lck, suggesting that in the context of the intestine there could be differential regulation of tyrosine phosphorylation of GC-C along the crypt-villus axis. Intestinal ligated loop assays in rats demonstrated that ST-induced fluid accumulation in the intestine was abrogated on pervanadate treatment. Reduction in this fluid accumulation by pervanadate was not observed with 8-Br-cGMP, a cell permeable analogue of cGMP. This indicated that tyrosine phosphorylation of proteins is important for ST-induced fluid accumulation, and perhaps pervanadate modulates this by phosphorylation of GC-C, thereby causing a reduction in fluid accumulation. Earlier in vitro studies on Src-SH2 binding from the laboratory had suggested the possibility of activation of Src family kinases by GC-C. The activation status of Src kinases was monitored by using phosphorylation-state specific antibody, pSFK416. ST stimulation in T84 cells increased Tyr416 phosphorylation of Src kinases in a time dependent manner, indicating that Src kinases are activated downstream of GC-C. This activation of Src kinases was also seen with the endogenous ligand of GC-C, uroguanylin. Interestingly, 8-Br-cGMP a cell permeable analogue of cGMP that is known to mimic other cellular effects of GC-C, namely Cl-secretion and cell cycle progression, did not activate Src kinases, suggesting that the mechanism of Src kinase activation by GC-C could be independent of cGMP. Binding affinities of Src, Lck, Fyn and Yes SH2 domains to Tyr820 phosphorylated GCC peptide were in the nM range, indicating a high affinity of interaction. In vitro GST-SH2 pull down experiments suggested that phosphorylation of Tyr820 in full length GC-C allows interaction of GC-C to the SH2 domain of Src. These studies suggest a dual cross-talk between Src kinases and GC-C; Src phosphorylation inhibits GC-C signaling and stimulation of GC-C by its ligands activates Src kinases. Interaction of proteins containing SH2 and SH3 domains are commonly found in signaling molecules. In accordance with the observation that there are three PXXP motifs in GCC, many SH3 domains could interact with GC-C. GC-C appears to show a preference to bind the SH3 domains of Fyn, Hck, Abl tyrosine kinases, Grb2 and Crk adaptor proteins, the α-subunit of P85 PI3 kinase, PLC-γ and cortactin to various extents. The SH3 domains of spectrin and Nck did not show any detectable interaction with GC-C. In SH3 pull-down assays, the N-terminal SH3 domain of Crk, CrkSH3 (N), bound GC-C maximally, suggesting that Crk is a good candidate for interaction with GC-C. By overlay analysis, the region of GC-C that binds CrkSH3 (N) was narrowed down to the catalytic domain of GC-C containing a ‘PGLP’ motif. Mutations were generated in GC-C at this site to generate GC-CP916Q and GC-CW918R. These mutations compromised the binding of full length receptor to CrkSH3 (N). In cells, CrkII and GC-C co-transfection inhibited the ST stimulation of GC-C. A CrkII mutant, that has compromised binding through its SH3 domain, did not inhibit the activity of GC-C. CrkII from T84 cells co-immunoprecipitated with GC-C and interestingly, the phosphorylated form of CrkII did not, indicating that GC-C - Crk interaction could be regulated by the phosphorylation of Crk. In summary, this study places GC-C, in the context of tyrosine kinase signaling pathway and interaction with the adaptor protein Crk. These studies suggest that GC-C signal transduction can be altered by cross-talk with other signaling events in the cell. Reversible phosphorylation of tyrosine residues inhibits the activity of GC-C, and this is mediated by Src family kinases. Src kinases themselves are activated on stimulation of GC-C by its ligands, possibly because of SH2 domain interaction with GC-C. Association of Crk by its SH3 domain regulates GC-C functioning primarily by inhibiting ST-stimulated cGMP production. This opens up the possibility of GC-C signaling through a multimeric complex involving other binding partners of Crk, and these cross-talks involving GC-C with the two proto-oncogenes, Src and Crk, might have far reaching consequences in the regulation of cellular functions.
Стилі APA, Harvard, Vancouver, ISO та ін.
28

Vivek, T. N. "Receptor Guanylyl Cyclase C Cross-talk With Tyrosine Kinases And The Adaptor Protein, Crk." Thesis, 2006. http://hdl.handle.net/2005/412.

Повний текст джерела
Анотація:
Signal transduction is a crucial event that enables cells to sense and respond to cues from their immediate environment. Guanylyl cyclase C (GC-C) is a member of the family of receptor guanylyl cyclases. GC-C is a single transmembrane protein that responds to its ligands by the production of the second messenger cGMP. The guanylin family of peptides, (including the bacterially produced heat-stable enterotoxin ST) is the ligand for GC-C, elevates intracellular cGMP levels and activates downstream pathways. GC-C regulates the cystic fibrosis transmembrane conductance regulator (CFTR) by inducing phosphorylation by protein kinase G, resulting in chloride ion and fluid efflux. GC-C also regulates cell cycle progression through cGMP-gated Ca2+ channels. These functions are seen in the intestinal epithelium, the primary site for GC-C expression. GC-C as a molecule has been studied in detail, but its functioning in the context of other signaling pathways remains unknown. The aim of the present investigation was to understand the regulation of signal transduction by GC-C and its cross-talk with other signaling pathways operating in the cell. Molecular events that commonly connect components in a signaling pathway are protein phosphorylation and protein-protein interaction. These two aspects are explored in this thesis. The possibility of tyrosine phosphorylation of GC-C has been explored earlier in our laboratory. In vitro studies indicated that the residue Tyr820 was a site for phosphorylation by the Src family of non-receptor tyrosine kinases and those studies also suggested that phosphorylated Tyr820 could bind to the SH2 domain of Src. We generated a nonphosphorylatable mutant of GC-C, GC-CY820F, and a phosphomimetic mutant GC-CY820E to study the effect of phosphorylation of Tyr820, on the functioning of GC-C. A stable cell line of HEK293:GC-CY820F cells was generated and compared with HEK293:GC-CWT. Dose response to ST in the two cell lines showed that cGMP accumulation by GC-CY820F was greater than that of GC-CWT, although the EC50 remained unchanged. The phosphomimetic GC-CY820E mutant receptor was non-responsive to ST. Further in HEK293 cells, phosphorylation of GC-CWT by constitutively active v-Src resulted in decreased ST stimulation and this effect of v-Src was reduced with GC-CY820F. Inhibition of ST stimulation brought about by v-Src required catalytically active Src, as the kinase inactive v-SrcK295R did not inhibit ST stimulation. These results were corroborated by in vitro studies by using the recombinant catalytic domain of GC-C expressed in insect cells and by phosphorylation using a purified kinase, Hck. Observations suggested that phosphorylation of Tyr820 in the catalytic domain of GC-C compromises the guanylyl cyclase activity of GC-C. T84 and Caco-2 colon carcinoma cells endogenously express GC-C. The effect of tyrosine phosphorylation of GC-C was studied by using HgCl2, a known activator of Src kinases, and by the inhibition of protein tyrosine phosphatases using pervanadate, an irreversible inhibitor. Both these ways of achieving increased tyrosine phosphorylation resulted in decreased ST-stimulated cGMP production by GC-C, as suggested from v-Src transfection studies. This decrease was reversed by using a Src kinase specific inhibitor PP2, confirming the role of Src kinases in the inhibition of GC-C activity. Interestingly, in Caco-2 cells that differentiate in culture, the effect of pervanadate on the inhibition of ST-stimulated GC-C activation was dependent on the differentiation stage. Crypt-like cells showed higher inhibition with pervanadate. As they matured into villus-like cells, the effect of pervanadate on GC-C activation was gradually lost. This effect also correlated with a decrease in the expression of Lck, suggesting that in the context of the intestine there could be differential regulation of tyrosine phosphorylation of GC-C along the crypt-villus axis. Intestinal ligated loop assays in rats demonstrated that ST-induced fluid accumulation in the intestine was abrogated on pervanadate treatment. Reduction in this fluid accumulation by pervanadate was not observed with 8-Br-cGMP, a cell permeable analogue of cGMP. This indicated that tyrosine phosphorylation of proteins is important for ST-induced fluid accumulation, and perhaps pervanadate modulates this by phosphorylation of GC-C, thereby causing a reduction in fluid accumulation. Earlier in vitro studies on Src-SH2 binding from the laboratory had suggested the possibility of activation of Src family kinases by GC-C. The activation status of Src kinases was monitored by using phosphorylation-state specific antibody, pSFK416. ST stimulation in T84 cells increased Tyr416 phosphorylation of Src kinases in a time dependent manner, indicating that Src kinases are activated downstream of GC-C. This activation of Src kinases was also seen with the endogenous ligand of GC-C, uroguanylin. Interestingly, 8-Br-cGMP a cell permeable analogue of cGMP that is known to mimic other cellular effects of GC-C, namely Cl-secretion and cell cycle progression, did not activate Src kinases, suggesting that the mechanism of Src kinase activation by GC-C could be independent of cGMP. Binding affinities of Src, Lck, Fyn and Yes SH2 domains to Tyr820 phosphorylated GCC peptide were in the nM range, indicating a high affinity of interaction. In vitro GST-SH2 pull down experiments suggested that phosphorylation of Tyr820 in full length GC-C allows interaction of GC-C to the SH2 domain of Src. These studies suggest a dual cross-talk between Src kinases and GC-C; Src phosphorylation inhibits GC-C signaling and stimulation of GC-C by its ligands activates Src kinases. Interaction of proteins containing SH2 and SH3 domains are commonly found in signaling molecules. In accordance with the observation that there are three PXXP motifs in GCC, many SH3 domains could interact with GC-C. GC-C appears to show a preference to bind the SH3 domains of Fyn, Hck, Abl tyrosine kinases, Grb2 and Crk adaptor proteins, the α-subunit of P85 PI3 kinase, PLC-γ and cortactin to various extents. The SH3 domains of spectrin and Nck did not show any detectable interaction with GC-C. In SH3 pull-down assays, the N-terminal SH3 domain of Crk, CrkSH3 (N), bound GC-C maximally, suggesting that Crk is a good candidate for interaction with GC-C. By overlay analysis, the region of GC-C that binds CrkSH3 (N) was narrowed down to the catalytic domain of GC-C containing a ‘PGLP’ motif. Mutations were generated in GC-C at this site to generate GC-CP916Q and GC-CW918R. These mutations compromised the binding of full length receptor to CrkSH3 (N). In cells, CrkII and GC-C co-transfection inhibited the ST stimulation of GC-C. A CrkII mutant, that has compromised binding through its SH3 domain, did not inhibit the activity of GC-C. CrkII from T84 cells co-immunoprecipitated with GC-C and interestingly, the phosphorylated form of CrkII did not, indicating that GC-C - Crk interaction could be regulated by the phosphorylation of Crk. In summary, this study places GC-C, in the context of tyrosine kinase signaling pathway and interaction with the adaptor protein Crk. These studies suggest that GC-C signal transduction can be altered by cross-talk with other signaling events in the cell. Reversible phosphorylation of tyrosine residues inhibits the activity of GC-C, and this is mediated by Src family kinases. Src kinases themselves are activated on stimulation of GC-C by its ligands, possibly because of SH2 domain interaction with GC-C. Association of Crk by its SH3 domain regulates GC-C functioning primarily by inhibiting ST-stimulated cGMP production. This opens up the possibility of GC-C signaling through a multimeric complex involving other binding partners of Crk, and these cross-talks involving GC-C with the two proto-oncogenes, Src and Crk, might have far reaching consequences in the regulation of cellular functions.
Стилі APA, Harvard, Vancouver, ISO та ін.
29

Bose, Avipsa. "Elucidating the Role of Cyclic GMP in Diarrhoea and Intestinal Inflammation." Thesis, 2022. https://etd.iisc.ac.in/handle/2005/5939.

Повний текст джерела
Анотація:
Cyclic guanosine 3’,5’-monophosphate (cGMP) performs a wide range of functions in various cell types and tissues. The cellular levels of cGMP are maintained by the enzymatic conversion of guanosine 5’-triphosphate (GTP) to cGMP by guanylyl cyclases, which can also be membrane-associated receptors. Receptor guanylyl cyclase C (GC-C; gene GUCY2C) is predominantly expressed on the apical surface of the intestinal epithelial cells. GC-C is activated by peptide hormones guanylin and uroguanylin and the heat-stable enterotoxin (ST) produced by enterotoxigenic E.coli that causes traveller’s diarrhoea. Several disease-causing mutations in GUCY2C have been reported. Patients with gain-of-function mutations show diarrhoea and inflammatory bowel disease (IBD). Several cases of paediatric IBD have also been associated with mutations in GUCY2C. This study addresses the physiological implications of increased cGMP using a transgenic mouse model harbouring the first-identified hyperactive mutation in human patients leading to familial GUCY2C diarrhoea syndrome (FGDS). Mice with hyperactive GC-C showed increased levels of cGMP in intestinal epithelial cells, which led to activation of Cftr and inhibition of Nhe3, resulting in diarrhoea-like symptoms and increased luminal pH and faecal sodium levels. Global transcriptome analysis of the distal colon revealed activation of the interferon signalling pathway, and transgenic mice showed greater susceptibility to DSS-induced colitis. Histological analysis of the terminal ileum revealed a reduction in functional Paneth cells, goblet cells, and mucus barrier. The barrier integrity of the small intestine was compromised in these mice. Global transcriptome analysis of the terminal ileum revealed a Th1-type gene signature. Immune cell profiling across the gut-associated lymphoid tissue (GALT) showed reduced regulatory dendritic cells and an increased abundance of CD4+ Th cells. Increased levels of Stat1 were observed in the ileal epithelial cells of these mice, along with elevated expression of interferon-stimulated genes. Small intestinal organoids were prepared from wild type and transgenic mice. The organoids from transgenic mice showed greater swelling in the presence of ST and uroguanylin due to increased fluid secretion into the lumen. Administration of cGMP increased Stat1 phosphorylation in the intestinal organoids. Our observations suggest that high cGMP levels have epithelial cell-extrinsic and cell-intrinsic roles in inducing intestinal inflammation. Furthermore, the administration of zinc inhibited the activity of GC-C and reduced diarrhoea and intestinal inflammation in the transgenic mice. Thus, the similarities observed in these transgenic mice with that of chronic diarrhoea and IBD patients indicate that they can be used as a pre-clinical model to understand the effects of chronically elevated cGMP on intestinal pathophysiology and for identifying novel therapeutic strategies for patients with hyperactivating mutations in GUCY2C.
Стилі APA, Harvard, Vancouver, ISO та ін.
30

Nedvetsky, Pavel I. [Verfasser]. "Regulation of the nitric oxide receptor, soluble guanylyl cyclase / vorgelegt von Pavel I. Nedvetsky." 2003. http://d-nb.info/969682026/34.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
31

Srivastava, Rashi [Verfasser]. "Molecular mechanisms affecting expression of the NO receptor soluble guanylyl cyclase (sGC) / von Rashi Srivastava." 2006. http://d-nb.info/982425651/34.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
32

Ciou, Yi-Yun, and 邱逸雲. "Functional Characterization of a Novel Receptor Mouse Guanylyl Cyclase G (mGC-G) in Reproduction System." Thesis, 2006. http://ndltd.ncl.edu.tw/handle/00720078368604347713.

Повний текст джерела
Анотація:
碩士
臺北醫學大學
醫學研究所
94
We recently identified a novel testis-enriched receptor guanylyl cyclase (GC) in the mouse, designated mGC-G. To further investigate its protein expression and function, we generated a neutralizing antibody specifically against the extracellular domain of this receptor. Reverse transcriptase (RT)-PCR and immunohistochemical analyses show that mGC-G is predominantly expressed from round spermatids to spermatozoa in mouse testis at both mRNA and protein levels. In female genital tract, mGC-G also has a significant expression in granulosa cell and zona pellucida in the ovary by immunohistochemistry. Flow cytometry and confocal immunofluorescence reveal that mGC-G is a cell-surface protein restricted to the plasma membrane overlying the acrosome and the midpiece of the flagellum in mature sperm. Interestingly, Western-blot analysis demonstrates that testicular mGC-G harbors an apparent molecular mass of approximately 180 kDa, but is subject to a limited proteolysis during epididymal sperm transport, resulting in a smaller fragment tethered on mature sperm surface. By utilizing Fluo-3 cytometrical analysis and computer-assisted sperm assay, we found that albumin-induced elevation of sperm [Ca2+]i level, protein tyrosine phosphorylation associated with capacitation and progressive motility are markedly reduced by pre-incubation of the anti-mGC-G neutralizing antibody. To further characterize mGC-G fuction in vivo, we generate mGC-G knock-out mice. Preliminary results show that the fertility of mGC-G null mice is reduced both in vivo and in vitro. Together, this study provides evidence that mGC-G is proteolytically modified in mature sperm membrane and suggests that mGC-G-mediated signaling may play a critical role in gamete/reproductive biology.
Стилі APA, Harvard, Vancouver, ISO та ін.
33

Michel, Jennifer Melanie Lisa. "Gene expression profiling in hearts of transgenic mice overexpressing guanylyl cyclase domain of the GC-A receptor." Thèse, 2004. http://hdl.handle.net/1866/15538.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
34

"Guanylyl cyclase-A/natriuretic peptide receptor-A-mediated regulation and inhibition of mitogen-activated protein kinases: Identification of signaling targets." Tulane University, 2010.

Знайти повний текст джерела
Анотація:
Atrial natriuretic peptide (ANP) is a vasoactive peptide hormone generated in the atrium of the heart and released in the circulation. ANP exhibits important functions in blood pressure and volume homeostasis, cardiac hypertrophy, and inhibition of cell proliferation. The binding of ANP to the cell surface guanylyl cyclase-A/natriuretic peptide receptor-A (GC-A/NPRA) activates its intrinsic guanylyl cyclase domain, causing production of the intracellular second messenger cGMP, and subsequently activating cGMP-dependent protein kinase (PKG). Although ANP is known to inhibit cell proliferation and promote apoptosis in numerous cell types, the signaling pathways responsible for the antimitogenic actions of ANP are not well understood. The objective of this study was to elucidate the role of the ANP-NPRA-PKG system in the inhibition of mitogen-activated protein kinases (MAPKs) and in cell growth and proliferation by studying effect of ANP on the activation of MAPKs and the downstream transcription factors cAMP response element binding protein (CREB) and activating protein-1 (AP-1) regulated by MAPKs. These studies have shown the role of the ANP-NPRA signaling system in regulating and inhibiting the proliferation of two types of cells: mouse mesangial cells (MMCs) and Leydig tumor cells (LEDs). We have delineated a pathway for this signaling system, demonstrating that ANP inhibits the activation of MAPKs and two specific proliferation-stimulating transcription factors downstream, CREB and AP-1, effects that were seen to be dependent on PKG and mediated through NPRA rather than through NPRC. These findings help to advance our understanding of the biochemical pathways through which ANP inhibits cell proliferation
acase@tulane.edu
Стилі APA, Harvard, Vancouver, ISO та ін.
35

Börner, Juliane [Verfasser]. "Charakterisierung der Phosphorylierungsstellen der Guanylyl-Cyklase A, dem Rezeptor für das atriale natriuretische Peptid, mittels Massenspektrometrie = Characterization of the phosphorylation sites of Guanylyl cyclase A, the receptor for atrial natriuretic peptide, by mass spectrometry application / vorgelegt von Juliane Börner." 2010. http://d-nb.info/1008729213/34.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
36

Pinto, Isa Eunice Costa Figueiredo Paula. "The role of cGMP on adenosine A1 receptor-mediated inhibition of synaptic transmission at the hippocampus." Master's thesis, 2014. http://hdl.handle.net/10400.6/5663.

Повний текст джерела
Анотація:
The adenosine A1 receptor is highly expressed in hippocampus where it inhibits synaptic transmission and has neuroprotective activity. Similar actions are obtained by increasing the concentration of the second messenger cGMP and recently it was found that activation of adenosine A1 receptor increased cGMP levels in the nervous system, but the role of cGMP on adenosine A1 receptor mediated inhibition of synaptic transmission remains to be established. This work addressed if the adenosine A1 receptor inhibitory effect on neurotransmission is dependent on the cGMP pathway. To answer this question investigated in what extent increasing the levels of cGMP (with a phosphodiesterases inhibitor and a cGMP analog), or blocking the cGMP pathway (using nitric oxide synthase, protein kinase G and soluble guanylyl cyclase inhibitors) modify the inhibitory effect of an A1 receptor agonist on synaptic transmission. The hippocampal slice was used as an experimental model and neurotransmission evaluated through extracelular electrophysiological recording technique, specifically by measuring the slope of field excitatory postsynaptic potentials (fEPSPs) evoked by electrical stimulation. N6-cyclopentyladenosine (CPA, 15nM), a selective adenosine A1 receptor agonist, reversibly decreased the fEPSPs by 48% ± 2.1% (n=5; P<0.05). Incubation of the slices with a phosphodiesterase inhibitor Bay 60-7550 (100 nM), in order to prevent cGMP degradation, did not modify the CPA (15 nM) inhibitory effect on fEPSPs (50%±2.8%, n=5; P>0.05 when compared with CPA alone). The presence of a membrane-permeable analog of cGMP, 8-pCPTP-cGMP (10 µM), also did not significantly affect (P>0.05) the CPA (15nM) inhibitory effect on fEPSPs (59.0%±4.5% in the absence and 50%±8.6% in the presence of 8-pCTP-cGMP, n=4). On the other hand, inhibition of nitric oxide synthase (NOS) by L-NAME (200 µM) decreased (P<0.05) the CPA (15nM) inhibitory effect on fEPSPs (54%±5.3% in the absence and 23%±5.7% in the presence of L-NAME, n=5, female rats). In male rats, the presence of a Protein Kinase G (PKG) inhibitor (KT5823, 1nM) decreased the inhibitory effect of CPA (15nM) on fEPSPs by 45.0%±8.9% (n=4, P<0.05 compared with zero); similar results were obtained in females. Finally the effect of an inhibitor of soluble guanylyl cyclase (sGC), ODQ (10 µM), on the CPA (15 nM) inhibitory action on fEPSPs was investigated. In males ODQ decreased (P<0.05) the CPA inhibitory effect (50%±4.3% in the absence and 39%±6.0% in the presence of ODQ, n=4), but only when adenosine deaminase (1 U/ml) was present; similar results were found in females. In conclusion, the results strongly suggest that inhibitory action of adenosine A1 receptors on glutamatergic neurotransmission at the hippocampus is, at least partially, mediated by activation of the NOS/sGC/cGMP/PKG pathway.
A adenosina é libertada para o meio extracelular em situação de actividade neuronal prolongada ou após insulto neurotóxico. A adenosina, via activação do receptor A1, inibe a transmissão sináptica e previne o dano neuronal causado por isquémia, excitotocicidade ou episódios epiléticos. Os receptores A1 diminuem a neurotransmissão quer pré-sinapticamente, por inibibição da libertação de glutamato, quer pós-sinapticamente, por activação dos canais de potássio que provocam hiperpolarização dos neurónios pós-sinápticos. Os receptores A1 produzem este efeito inibitório através da modulação de vários efectores e mensageiros intracelulares, tais como o cAMP, canais de cálcio e potássio e fosfatos de inositol. O cGMP é produzido pelas ciclases do guanililo que são uma família de enzimas que convertem o GTP em cGMP. Existem dois tipos de ciclases do guanililo: a forma solúvel (sGC) activada pelo oxido nítrico e a forma particulada, a qual constitui um receptor membranar para ligandos extracelulares como o péptido natriurético. O cGMP é degradado por acção de fosfodiesterases que catalisam a hidrólise do cGMP e também do cAMP (algumas formas), desempenhando um importante papel na regulação da via dos nucleótidos cíclicos e na comunicação celular. O cGMP através da activação da cinase G de proteínas (PKG) diminui a transmissão sináptica reduzindo a actividade neuronal e exerce uma acção neuroprotectora face a insultos neurotóxicos. Uma via importante que modula os níveis de cGMP no cérebro em resposta à atividade sináptica é a via oxido nítrico/sGC. O oxido nitrico age como mensageiro retrogrado, uma vez que além de activar a ciclase do guanililo solúvel em neurónio pós-sinápticos pode difundir para o meio extracelular e pré-sinapticamente estimular a sGC produzindo cGMP diminuindo assim a libertação de glutamato. Estudos anteriores mostraram que doadores de oxido nítrico inibiram a transmissão sináptica em fatias de hippocampo e essa inibição foi bloqueada por antagonistas do receptor A1 (Boulton et al., 1994; Broome et al., 1994). Em contraste, a inibição da ciclase do guanililo solúvel não afectou o efeito inibitório do doador de oxido nitrico na transmisão sináptica (Arrigoni et al., 2006). Por outro lado, o óxido nítrico mostrou aumentar o efeito inibitório da 2-chloroadenosine (CADO) na transmissão sináptica e esse aumento foi bloqueado por inibidores da sGC (Fragata et al., 2006). No entanto, o papel do cGMP na mediação do efeito inibitório do receptor A1 na transmissão sináptica permanece por esclarecer. Observámos recentemente que a activação do receptor A1 aumenta a formação de cGMP no cérebro. Com o presente projecto pretende-se investigar se o efeito inibitório do receptor A1 na transmissão sináptica glutamatérgica serão mediados, pelo menos em parte, pela via do cGMP. Para responder à questão anterior investigou-se em que medida fármacos que aumentam os níveis de cGMP (e.g. inibidor de fosfodiesterases e análogo do cGMP) ou que bloqueiem a via do cGMP (e.g. inibidores da sintase do óxido nítrico, da PKG e da sGC) modificarão o efeito de um agonista selectivo do receptor A1 na transmissão sináptica. A fatia de hipocampo de rato foi utilizada como modelo experimental. A neurotransmissão foi avaliada por electrofisiologia através do registo e medição do declive dos potenciais pós-sinápticos excitatórios de campo (fEPSPs) evocados por estimulação eléctrica. A N6-ciclopentiladenosina (CPA, 15 nM), um agonista selectivo do receptor A1 de adenosina diminuiu reversivelmente os fEPSPs em 48% ± 2,1% (n = 5, P<0,05). A incubação das fatias com um inibidor de fosfodiesterases, o Bay 60-7550 (100 nM), a fim de evitar a degradação do cGMP, não alterou o efeito inibitório da CPA (15 nM) nos fEPSPs (50% ± 2,8%, n = 5, P>0,05 quando comparado com a CPA sozinha). A presença de um análogo do cGMP permeável à membrana, o 8-pCPTP-cGMP (10 µM), também não afectou significativamente (P>0,05) o efeito inibitório da CPA (15 nM) nos fEPSPs (59% ± 4,5%, na ausência e 50% ± 8,6% na presença de 8-PCTP-cGMP, n = 4). Por outro lado, a inibição da sintase do óxido nítrico (NOS) pelo L-NAME (200 µM) diminuiu (P<0,05) o efeito inibitório da CPA (15 nM) nos fEPSPs (54% ± 5,3 % na ausência e 23% ± 5,7% na presença de L-NAME, n = 5, ratos fêmeas). Em ratos machos, a presença de um inibidor (KT5823, 1 nM) da PKG diminuiu o efeito inibitório do CPA (15 nM) nos fEPSPs em 45.0%±8.9% (n=4, P<0,05 comparado com zero); resultados semelhantes também foram obtidos em fêmeas. Finalmente, o efeito de um inibidor da ciclase do guanililo solúvel, o ODQ (10 µM), na acção inibitória da CPA (15 nM) nos fEPSPs foi investigado. Em machos o ODQ diminuiu (P<0,05) o efeito inibitório da CPA (50% ± 4,3%, na ausência e 39% ± 6,0% na presença de ODQ, n=4), mas apenas quando a desaminase da adenosina (1 U/ml) estava presente; resultados semelhantes foram obtidos em fêmeas. Em conclusão, os resultados sugerem que a acção inibitória dos receptores A1 da adenosina na neurotransmissão glutamatérgica ao nível do hipocampo é, pelo menos em parte, mediada pela activação da via NOS/sGC/cGMP/PKG.
Стилі APA, Harvard, Vancouver, ISO та ін.
37

Taye, Ashraf Mohamed Abouelwafa [Verfasser]. "Divergent effects of angiotensin II receptor types 1A and 2 on vascular functions involve NADPH oxidase-dependent oxidative stress and no-dependent guanylyl cyclase / submitted by Ashraf Mohamed Abouelwafa Taye." 2005. http://d-nb.info/974104493/34.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
38

Fillion-Forté, Valérie. "Le syndrome métabolique chez les congéniques du rat Dahl : influence de la diète et rôle du récepteur de l'ANP." Thèse, 2010. http://hdl.handle.net/1866/4285.

Повний текст джерела
Анотація:
L’hypertension artérielle et l’obésité sont deux composantes conjointement reliées du syndrome métabolique. Les récepteurs de l’ANP (GCA) et de l’oxyde nitrique (GCs) ont des propriétés diurétiques, natriurétiques, vasodilatatrices et sont liés au contrôle de la pression. Des études récentes ont démontré leur implication dans l’obésité. Hypothèse : Une différence génétique au niveau du gène GCA pourrait contribuer à des différences physiologiques. La composante lipidique et/ou sodique de la diète pourrait influencer la fonction rénale, cardiaque et les valeurs anthropométriques différemment chez les souches congéniques. Objectifs : (1) Déterminer l’effet de la composante lipidique et sodique des diètes; (2) Évaluer l’influence de GCA sur la réponse physiologique des souches congéniques; (3) Expliquer les mécanismes physiologiques procurant une réduction de la pression artérielle chez la souche SM9. Méthodologie : Des modèles congéniques du rat Dahl (DSS) hypertendu, nourri avec une diète riche en gras (HF) ou normale (NF), ont été utilisés pour démontrer l’impact d’un segment chromosomique d’origine normotendue. Résultats : La souche SM9 a une prise de poids plus importante que SM12 et DSS sur diète HF malgré un apport alimentaire équivalent. La souche SM9 présente également un ratio masse adipeuse/masse maigre plus élevé que SM12 et DSS. Nous n’avons observé aucune augmentation de la pression artérielle en réponse à la diète HF pour les 3 souches malgré une augmentation du dommage rénal pour les 3 souches. Le dommage rénal est plus important chez DSS que pour les 2 congéniques. La réponse diurétique à l’ANP est plus élevée chez SM9 et est influencée par le contenu en sel dand la diète. La perte glomérulaire plus importante chez le rat DSS semble compensée par une augmentation de la réponse à l’ANP par les glomérules résiduels. Il y a une corrélation entre l’activité de GCA en réponse à l’ANP, les niveaux d’ARNm et le nombre de répétition du dinucléotide TA dans son promoteur. Le rat DSS présente une hypertrophie cardiaque plus importante que les deux souches congénique et ceci n’est pas modifié par la diète HF. Conclusion : Nos études ont permis de mettre en évidence un effet génétique impliquant le segment chromosomique normotendu contenant GCA dans la réponse à une diète HF chez le rat DSS.
Hypertension and obesity are two related components of the metabolic syndrome. The ANP receptor (GCA) and nitric oxide receptor (sGC) have diuretic, natriuretic, vasodilatory properties, and are linked to blood pressure control. Furthermore a recent study has demonstrated the implication of GCA and sGC in the development of obesity. Hypothesis: A genetic difference in GCA gene could contribute to physiological differences. The differencial lipid and/or sodium composition of the diet could influence the renal, cardiac and anthropometric values. Objectives: (1) To determine the effect of fat and sodium on the physiological parameters; (2) To evaluate the influence of GCA on the physiological response of the congenic rat; (3) To explain the mechanisms of the blood pressure reduction in SM9 rats. Methodology: Congenic model of DSS rat, fed with either high fat (HF) or normal (NF) diet, were used to demonstrate the impact of a chromosome segment from normotensive origin on physiological functions. C2SM9 contains GCA and sGC from normotensive origin while C2SM12 harbours only sGC from normotensive origin. Results: HF diet had negative feature on body composition, renal damage, creatinine clearance and inhibited the diuretic/natriuretic effect of ANP. The normotensive segment including GCA and sGC has reduced the blood pressure, improve the renal damage and increased the diuretic/natriuretic capacity of SM9 in response to ANP injection when compared to SM12 and DSS. GCA mRNA and the clearance receptor ratio were reduced in SM9 in the renal cortex and retroperitoneal fat. SM12 and SM9, containing the chromosomal segment that includes sGC, improve their lipid profile compared with DSS. Conclusion: Our results suggested a compensatory increase in the GCA levels for SM12 and DSS that is insufficient to improve their pathophysiologic status as observed in SM9. HF diet increases the metabolic syndrome in those rats.
Стилі APA, Harvard, Vancouver, ISO та ін.
39

Dankworth, Beatrice. "Charakterisierung der dynamischen Interaktion des Guanylyl Cyclase-A (GC-A)-Rezeptors mit den Transient Receptor Potential Canonical Type 3 und Type 6 (TRPC3/C6)-Kanälen und Generierung von β-Zell-spezifischen GC-A-knock-out-Mäusen sowie die Analyse der Bedeutung von ANP für die Insulin-Homöostase unter pathophysiologischen Bedingungen". Doctoral thesis, 2013. https://nbn-resolving.org/urn:nbn:de:bvb:20-opus-78650.

Повний текст джерела
Анотація:
Das atriale natriuretische Peptid (ANP) beeinflusst den arteriellen Blutdruck und das intravasale Volumen durch Stimulation der intrazellulären Produktion von cGMP über den membranständigen Guanylyl Cyclase-A (GC-A)-Rezeptor. ANP stimuliert außerdem die Angiogenese und ist am Wachstum der Kardio-myozyten beteiligt. Im ersten Teil der vorliegenden Arbeit wurde die dynamische Interaktion zwischen den rezeptoraktivierten Kationenkanälen Transient Receptor Potential Canonical Type 3 und Type 6 (TRPC3/C6) und dem GC-A-Rezeptor untersucht. Erst kürzlich konnte gezeigt werden, dass ANP über GC-A den TRPC-vermittelten Ca2+-Einstrom in Kardiomyozyten auf cGMP-unabhängige Weise stimuliert. Um eine mögliche direkte Interaktion von TRPC3/C6 und GC-A zu zeigen, wurde TRPC3 oder C6 mit Flag-GC-A in HEK293-Zellen koexprimiert. Die Membranfraktion der Zellen wurde nach Immunpräzipitation mit einem anti-Flag-Antikörper im Western Blot untersucht. Es konnte gezeigt werden, dass TRPC3/C6 unabhängig von ANP mit GC-A ko-immunpräzipitieren. Die Interaktion erfolgte auch mit einem modifizierten GC-A-Rezeptor, dem die Cyclase-Domäne fehlt. Um die Interaktion in Kardiomyozyten zu untersuchen, wurde ein transgenes Mausmodell mit einer Überproduktion von HA-GC-A in Kardiomyozyten verwendet. Auch bei diesem Modell konnte mittels anti-HA- Antikörper die Koimmunpräzipitation von GC-A und TRPC3/C6 nachgewiesen werden. Schließlich wurden FRET-basierte Untersuchungen durchgeführt, um die lokale Nähe von GC-A und TRPC3 zu beweisen und eine mögliche ANP-induzierte Konformationsänderung zu untersuchen. Die Koexpression von GC-A-CFP und TRPC3-YFP in HEK293 Zellen führte zu einem FRET-Signal, welches durch ANP konzentrationsabhängig (1-100 nM) gesenkt wurde. Die Gabe des membranpermeablen cGMP-Analagons 8-Br-cGMP führte dagegen zu keiner Veränderung des FRET-Signals. Die Ergebnisse bestätigen das Vorhandensein eines stabilen Proteinkomplexes von GC-A und TRPC3, der für den neuen cGMP-unabhängigen Signalweg von GC-A ausschlaggebend ist. Der zweite Teil der vorliegenden Arbeit beschreibt die Rolle von ANP/GC-A für die Insulinausschüttung der pankreatischen β-Zellen. Es ist bereits bekannt, dass GC-A in den β-Zellen exprimiert wird und dass ANP an isolierten Langerhans’schen Inseln das β-Zell-Wachstum und die Insulinsekretion moduliert. Um langfristig die Bedeutung von ANP für die systemische Glukose-Homöostase zu ergründen, wurde ein Mausmodell mit einer β-Zell-spezifischen GC-A-Deletion generiert. Der Nachweis des konditionellen GC-A knock out (KO) erfolgte mittels genomischer PCR und Immunhistochemie. Eine Detektion von GC-A in den Langerhans’schen Inseln auf Proteinebene war leider nicht möglich. Aber es konnte gezeigt werden, dass der β-Zell-spezifische KO zu keiner Expressionsänderung von GC-A in anderen Geweben führte. Auch der Blutdruck und das Herzgewicht der KO Mäuse blieb unauffällig. Zur Untersuchung der Bedeutung von ANP für die Insulinausschüttung unter pathologischen Bedingungen wurden KO- und Kontrolltiere für 12 Wochen einer fettreichen Ernährung (60% Fett) ausgesetzt um einen Prädiabetes auszulösen. Zu verschiedenen Zeitpunkten der Studie wurden orale Glukose-Toleranz-Tests (oGTT), Blutdruckmessungen und Gewichtsbestimmungen durchgeführt. Bereits vor der Studie wurde beobachtet, dass der Nüchternglukosewert in den weiblichen KO-Mäusen leicht erhöht ist. Daher wurden die oGTT‘s in der Studie geschlechtsspezifisch ausgewertet. Am Ende der Studie zeigten alle Mäuse eine vergleichbare insuffiziente Blutzuckerregulierung. Der Blutdruck war sowohl in KO- als auch in Kontrolltieren um ca. 60% erhöht. Einigen Tieren wurde das Pankreas entnommen und für immunhistologische Zwecke präpariert. Die morphometrische Auswertung der Pankreas-Schnitte ergab eine signifikant vergrößerte durchschnittliche Inselfläche und eine erhöhte durchschnittliche β-Zellfläche der KO-Tiere im Vergleich zu den Kontrollen. Die β-Zellen der KO-Tieren waren im Vergleich zu den Kontrollen hypertroph. Die Studie zeigt also, dass die Deletion von GC-A in den β-Zellen unter pathologischen Bedingungen zu einer Hypertrophie der β-Zellen führt und zu einem geringeren Schutz gegen die Ausbildung eines Prädiabetes beiträgt. Eine mögliche verstärkte periphere Insulinresistenz in den KO-Tieren ist auch nicht auszuschließen. Weitere Studien an dem neuen Mausmodell könnten die Bedeutung des ANP/GC-A-Systems für die Insulinausschüttung näher ergründen und dadurch eventuell neue Therapieansätze für Diabetes mellitus Typ 2 bringen
Atrial natriuretic peptide (ANP) modulates blood pressure and volume by its cGMP generating guanylyl cyclase-A (GC-A) receptor. ANP also stimulates cardiomyocyte growth and angiogenesis. This work concentrates on two separate mechanisms where ANP/GC-A system plays an important role. The first part of this work concerns the dynamic interaction of the transient receptor potential canonical type 3 or type 6 (TRPC3/C6) cation channels with GC-A receptor. Recently, it was indicated that ANP via GC-A stimulates the TRPC-mediated Ca2+ influx in cardiomyocytes in a cGMP-independent manner. To analyze the presumed direct interaction between TRPC3/C6 and GC-A the proteins TRPC3 or C6 and Flag-GC-A were coexpressed in HEK293 cells in presence or absence of ANP. After lysis GC-A was precipitated from the membrane fraction of the cells with anti-Flag-antibodies. TRPC3 and TRPC6 were detected in this fraction. The co-immunoprecipitation was also performed with a modified GC-A receptor lacking the cyclase domain as well as with cardiomyocytes from transgenic mice characterized by cardiomyocyte overexpression of HA-GC-A. In all cases TRPC3/C6 co-immunoprecipitated with GC-A. Finally, FRET-based approaches were used to examine the local distance between GC-A and TRPC3. Coexpression of GC-A-CFP and TRPC3-YFP in HEK293 cells led to a FRET signal which was decreased by ANP (1-100 nM) in a concentration dependent manner. Incubation of the membrane permeable cGMP analog 8-Br-cGMP did not alter the FRET signal. All results confirm the presence of a stable protein interaction between GC-A and TRPC3 or TRPC6. In the second part of this work the role of ANP/GC-A system for (patho)physiologic insulin release in pancreatic β-cells was investigated. It was recently shown that GC-A is expressed in β-cells and that ANP modulates β-cell growth and insulin secretion in isolated pancreatic islets. To analyze the relevance of ANP for the systemic glucose homeostasis a new mouse model was generated characterized by a β-cell-specific GC-A deletion. To prove the conditional GC-A knock out (KO) we used genomic PCR and immunohistochemistral approaches. The specific deletion did alter neither the GC-A expression in other tissues nor blood pressure and heart weight in KO mice. Fasting blood glucose levels were slightly elevated in female KO mice. Therefore all subsequent experiments were evaluated gender-related. Male and female controls and KO mice were fed a high fat diet (60 % fat) for 12 weeks to provoke a prediabetic state and insulin resistance. Oral glucose tolerance tests (oGTT), blood pressure measurements and weight analysis were performed to analyze if the KO affects insulin homeostasis under pathophysiological conditions. All mice developed an insufficient blood glucose regulation which was evident using the oGTT. Blood pressure was increased by 60% both in controls and KO mice. For immunohistochemical studies the pankreata from several animals were dissected and fixed in formalin. The cut organ slices were treated with glucagon and insulin antibodies. All islets were documented and analyzed morphometrically. The mean islet area and the mean β-cell area was significantly increased in KO animals. In summary, the β-cell-specific KO of GC-A led to hypertrophic β-cells under pathophysiological conditions. To uncover the entire role of ANP/GC-A system for the insulin release further studies have to be performed. The new mouse model provides the potential to find new therapeutic strategies for the treatment diabetes mellitus type 2
Стилі APA, Harvard, Vancouver, ISO та ін.
Ми пропонуємо знижки на всі преміум-плани для авторів, чиї праці увійшли до тематичних добірок літератури. Зв'яжіться з нами, щоб отримати унікальний промокод!

До бібліографії