• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 5
  • 2
  • 2
  • 2
  • Tagged with
  • 12
  • 3
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 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

Studies on the genetic control of infection and hepatic disease in schistosoma haematobium and schistosoma japonicum infections in human / Etudes du contrôle génétique des niveaux d'infection et des atteintes hépatiques dans les infections par Schistosoma haematobium et Schistosoma japonicum

He, Hongbin 21 December 2010 (has links)
La bilharziose reste un problème de santé majeur. L'équipe du Pr Dessein a montré que les infections élevées étaient déterminées par un locus majeur en 5q31 et que des polymorphismes dans un gène à ce locus,IL13, aggravent l'infection. Notre premier objectif était d'évaluer si des variants d'autres gènes de la voie de l'IL13 intervenaient dans le contrôle de l'infection. Nous avons observé une association entre le SNP rs324013, dans le promoteur de STAT6,et les niveaux d'infection à S. haematobium. Ce polymorphisme a un effet additif avec le polymorphisme IL13rs1800925. Ce SN modifie la fixation de facteurs nucléaires au niveau du promoteur de STAT6. L'équipe du Pr Dessein avait également montré que les fibres hépatiques avancées et sévères étaient déterminées par un autre locus majeur localisé en 6q23. Notre deuxième objectif fut d'évaluer dans le laboratoire du Pr Dessein et en étroite collaboration avec le laboratoire du Pr Li(Yueyang Institute of Parasitic disease)deux gènes candidats(IFNGR1 et CTGF) situés dans cette région chromosomique. Nous avons observé une association entre les deux polyporphismes(rs17066192 er rs673156)localisés dans le promoteur du gène. Nous avons observé une association entre les deux polymorphismes(rs17066192 et rs673156)localisés dans le promoteur du gène IFNGR1 et la fibrose hépatique: le génotype rs673156A/A et rs17066192C/C sont associés à un risque 7.3 fois et 1.5 fois plus élevé, respectivement, de fibrose avancée. Nous avons également montré que les variants rs9402373 et rs12526196 du gène CTGF sont indépendamment associés à la fibrose chez les fermiers et pêcheurs chinois infectés par S.japonicum. Sur la population chinoise d'étude, les risques relatifs associés aux polymorphismes rs9402373 et rs12526196 sont de 2.8 et 3 / Schistosomiasis remains one of the world’s most prevalent diseases. It comprises a group of chronic diseases caused by helminths of the Schistosoma genus. Schistosoma haematobium causes obstructive nephropathy that can be aggravated by urinary bacterial infections. S.japonicum and S.mansoni cause hepatic fibrosis associated with portal blood hypertension, which can be lethal. In previous studies, our laboratory had shown that worm burden in S.haematobium infections were aggravated by IL13 variants and that severe hepatic fibrosis (HF) was controlled by gene(s) located on 6q23. The present study is to further evaluate other IL-13 pathway genes (STAT6) in the control of infection in Malian farmers and to test candidate genes in the 6q23 region in hepatic fibrosis (HF) in S.japonicum infected Chinese fishermen and farmers. First we have developped an improved FTA® technology technique to perform SNP genotyping. This technique allows us to use saliva samples for genotyping SNPs. Subsequently, this improved FTA® technology was used in our study on HF.Our work on a Malian sample infected with S. haematobium indicated that a polymorphism (rs324013) in the promoter of STAT6 gene was associated with the control of S. haematobium infection levels and has an additive effect with IL13rs1800925, a polymorphism previously associated with infection in this same population. Both SNPs modify the binding of nuclear factors to the promoter regions of their respective genes. Thus, both SNPs may play a crucial role in controlling S. haematobium infection levels. In order to study HF in S.japonicum infections, we have participated actively in the study that recruited of a large sample of Chinese fishermen and farmers who had been exposed to the infection for most of their life. HF was evaluated by ultrasound and covariates that could affect HF were evaluated by interviews. Then, we tested two genes (IFNGR1, CTGF) of the 6q23 region that were good candidates for the control of HF on these samples. Both genes encode molecules that were shown in animal and human studies to have strong effect on extracellular matrix proteins deposition and turnover. We found that two polymorphisms (rs17066192 and rs673156) in IFNGR1 promoter were associated with HF: the rs673156A/A genotype was associated with a 7.3-fold increased risk of advanced HF; and rs17066192C/C genotype with a 1.5-fold increased risk of HF. These results must now be confirmed in another population sample. We also found that variants of CTGF rs9402373 and rs12526196 were independently associated with HF in Chinese fishermen and farmers, in Sudanese, and in Brazilians infected with either S. japonicum or S. mansoni. Our results provide additional evidence for a protective role of IL-13 in schistosome infections, and they also demonstrate that TGFβ / CTGF pathway plays a key role in HF and should be targeted by chemotherapy. Ongoing studies evaluate whether CTGF variants could be used in the prognosis of the HF caused by schistosomes and also by other infectious agents.
2

Étude de la voie de signalisation IL-4/IL-13 dans les lymphomes B primitifs du médiastin

Guiter, Chrystelle 19 December 2008 (has links)
Les lymphomes B primitifs du médiastin (LBPMs) constituent une entité anatomo-clinique particulière au sein des lymphomes diffus à grandes cellules B. Les analyses du transcriptome des LBPMs ont montré une forte expression des gènes induits par l’IL-4 ou l’IL-13 et des effecteurs de cette voie de signalisation. L’objectif de ce travail a été d’étudier la voie IL-4/IL-13 dans les LBPMs. Dans une 1ere partie, nous avons montré que le facteur de signalisation et de transcription 6 (STAT6) est constitutivement phosphorylé et possède une activité de liaison à l’ADN dans les lignées dérivées de LBPM (MedB1, Karpas1106). Le STAT6 phosphorylé (-P) est présent dans les noyaux des cellules tumorales de LBPMs (73% des cas). Cette activation est due en partie à l’activité de la kinase JAK2 et aux altérations du gène régulant négativement cette voie de signalisation, SOCS1. Dans une 2eme partie, nous avons étudié le rôle de STAT6 dans la physiopathologie de ces lymphomes en inhibant son expression par un siRNA dans les lignées. Nous avons montré une diminution de la prolifération et une augmentation de la mort cellulaire, ainsi qu’une diminution du taux du mRNA Bcl-xL dans les cellules MedB1. Nous avons observé une corrélation entre l’accumulation nucléaire de STAT6-P et l’expression cytoplasmique de Bcl-xL dans les cellules tumorales. Enfin, nous avons mis récemment en évidence des mutations du domaine de liaison à l’ADN de STAT6, dans 35% des LBPMs. L’étude des mécanismes oncogéniques activés dans la voie IL-4/IL-13 devrait permettre de comprendre le dysfonctionnement cellulaire à l’origine des LBPMs et pourrait aussi donner de nouvelles cibles pour le diagnostic et la thérapie / Primary mediastinal large B-cell lymphomas (PMBCLs) are a particular anatomo-clinical entity among diffuse large B-cell lymphomas (DLBCLs). The transcriptome analyses of PMBCLs showed high expression of genes activated by IL-4 or IL-13 and effectors of this signaling pathway. The objective of this work was study the IL-4 / IL-13 signaling pathway in PMBCLs. In a 1st part, we demonstrated that signal transducer and activator of transcription 6 (STAT6) is constitutively phosphorylated and exhibits DNA binding activity in PMBCL derived cell lines (MedB1, Karpas1106). This phosphorylated STAT6 (P-) is present in nuclei of PMBCL neoplastic cells (73 % of cases). This activation is partially due to the activity of JAK2 kinase and to the alteration of a gene which regulates negatively this signalling pathway, SOCS1. In a 2nd part, we studied the STAT6 role in physiopathology of these lymphomas by inhibiting its expression with a siRNA in cell lines. We showed proliferation decrease and cell death increase, as well as diminution of Bcl-xL mRNA in MedB1 cells. We observed a correlation between P-STAT6 nuclear accumulation and Bcl-xL cytoplasmic expression in neoplastic cells. In a last part, we recently demonstrate mutations of STAT6 DNA binding domain, in 35 % of PMBCLs. The study of oncogenic mechanisms activated in IL-4/IL-13 signaling pathway could allow to understand the cellular dysfunction at the origin of the PMBCLs and could also identify new targets for the diagnostic and the therapy
3

Biological functions and molecular mechanisms of the interleukin-4 signaling pathways in autoimmune exocrinopathy using the nod.b10.h2b mouse model of sjogren's syndrome

Gao, Juehua, January 2004 (has links)
Thesis (Ph. D.)--University of Florida, 2004. / Typescript. Title from title page of source document. Document formatted into pages; contains 152 pages. Includes Vita. Includes bibliographical references.
4

Molecular Mechanisms of Synergy Between IL-13 and IL-17A in Severe Asthma

Hall, Sara L., M.S. January 2017 (has links)
No description available.
5

Influência da anexina A1 sobre a fagocitose e a expressão de receptor ativado por proliferador de peroxissomo gama em células da microglia / Influence of annexin A1 upon phagocytosis and expression of peroxissome proliferator activated receptor gamma in microglial cells.

Rocha, Gustavo Henrique Oliveira da 13 March 2017 (has links)
A inflamação é fundamental para a manutenção da homeostasia e para a resposta do organismo à injúria. A resposta inflamatória deve ser adequada aos estímulos agressores; no sistema nervoso central, sua inadequação conduz à gênese de diferentes doenças neurodegenerativas. A proteína anexina A1 (ANXA1) e os receptores ativados por proliferadores de peroxissomo (PPAR) controlam a inflamação, pois ambos inibem o desenvolvimento da inflamação e aceleram sua resolução. Nosso grupo de pesquisa tem mostrado que a ANXA1 modula a expressão de PPARγ em macrófagos. Assim, o presente trabalho investigou a modulação da expressão do PPARγ e das suas funções em células da microglia pela ANXA1. Foram empregadas células imortalizadas da linhagem BV2 (microglia murina), inalteradas ou transfectadas para redução da expressão de ANXA1, tratadas com ANXA1 exógena (recombinante - rANXA1) ou com agonista ou antagonista de PPARγ (pioglitazona e GW9662, respectivamente). Os resultados obtidos mostraram que: 1) tratamento com rANXA1 aumenta as expressões gênica (RT-PCR) e proteica (Western Blotting) de PPARγ, e ambas as expressões estão reduzidas em células com deficiência endógena de ANXA1, sendo que tal efeito foi revertido pela ação da rANXA1; 2) tratamento com rANXA1 não induz a expressão dos fatores de transcrição ligados a expressão de PPARγ: proteínas ligantes de elementos de resposta ao cAMP - CREB - e transdutores de sinais e ativadores de transcrição - STAT6 - (Western Blotting), mas os níveis de ambos os fatores estão reduzidos em células transfectadas, e tal efeito não foi revertido pelo tratamento com rANXA1; 3) tratamento com pioglitazona ou com rANXA1 individualmente aumenta a fagocitose de células PC12 apoptóticas (citometria de fluxo), mas o tratamento simultâneo não altera a fagocitose induzida por pioglitazona ou rANXA1; no entanto, tratamento com GW9662 inibiu a fagocitose induzida pelo tratamento com rANXA1; 4) o tratamento com rANXA1 aumenta a expressão de CD36 (citometria de fluxo); a expressão de CD36 está reduzida em células transfectadas e tal expressão não é revertida pelo tratamento com rANXA1. Em conjunto, os dados obtidos mostram a modulação da ANXA1 sobre PPARγ em células da micróglia, com possível ação sobre a fagocitose de células apoptóticas, e que a redução da expressão de ANXA1 reduz acentuadamente a expressão dos fatores de transcrição STAT6 e CREB, bem como a expressão de CD36. A elucidação dos efeitos resultantes destas alterações desencadeadas pela deficiência de ANXA1 endógena poderá contribuir para compreensão da fisiopatologia da neuroinflamação. / Inflammation is a key process in maintaining homeostasis and is essential for the body\'s response to injury. The inflammatory response must be proportional to the aggressor stimuli; in the central nervous system, a failed proper modulation leads to the development of different neurodegenerative diseases. Protein annexin A1 (ANXA1) and peroxisome proliferated-activated receptors (PPAR) control inflammation, as both inhibit development of inflammation and accelerate its resolution. Our research group has demonstrated that ANXA1 modulates PPARγ expression in macrophages. Thus, the present work investigated the modulation of PPARγ expression and its functions in microglia cells by ANXA1. In order to assess such, immortalized cells from cell line BV2 (murine microglia), either unadulterated or transfected for reduced expression of ANXA1, were treated with exogenous ANXA1 (recombinant protein - rANXA1) or either with PPARγ agonist or antagonist (pioglitazone and GW9662, respectively). The obtained results demonstrated that: 1) treatment with rANXA1 increases both gene (RT-PCR) and protein (Western Blotting) expressions of PPARγ, and also that both expressions are reduced in cells with endogenous deficiency of ANXA1, and such effect was reversed by the actions of rANXA1; 2) treatment with rANXA1 does not promote the expression of transcription factors associated with PPARγ expression: cAMP response element binding protein - CREB - and signal transductor and activator of transcription 6 - STAT6 (Western Blotting), but the expression levels of both factors are reduced in transfected cells, and such effect was not reversed by treatment with rANXA1; 3) individual treatment with pioglitazone or rANXA1 increases phagocytosis of apoptotic PC12 cells (flow cytometry), but simultaneous treatment does not affect pioglitazone/rANXA1-induced phagocytosis; however, treatment with GW9662 inhibited rANXA1-induced phagocytosis; 4) treatment with rANXA1 increases CD36 expression (flow cytometry); the expression of CD36 is reduced in transfected cells, and such expression is not reversed by treatment with rANXA1. The obtained data demonstrate the modulation ANXA1 exerts upon PPARγ in microglia cells, with a possible action upon phagocytosis of apoptotic cells, and that reduction of ANXA1 expression greatly reduces the expression of transcription factors STAT6 and CREB, as well as the expression of CD36. Elucidation of such effects that arise from a deficiency of endogenous ANXA1 will contribute to a better comprehension of the pathophysiology of neuroinflammation.
6

Influência da anexina A1 sobre a fagocitose e a expressão de receptor ativado por proliferador de peroxissomo gama em células da microglia / Influence of annexin A1 upon phagocytosis and expression of peroxissome proliferator activated receptor gamma in microglial cells.

Gustavo Henrique Oliveira da Rocha 13 March 2017 (has links)
A inflamação é fundamental para a manutenção da homeostasia e para a resposta do organismo à injúria. A resposta inflamatória deve ser adequada aos estímulos agressores; no sistema nervoso central, sua inadequação conduz à gênese de diferentes doenças neurodegenerativas. A proteína anexina A1 (ANXA1) e os receptores ativados por proliferadores de peroxissomo (PPAR) controlam a inflamação, pois ambos inibem o desenvolvimento da inflamação e aceleram sua resolução. Nosso grupo de pesquisa tem mostrado que a ANXA1 modula a expressão de PPARγ em macrófagos. Assim, o presente trabalho investigou a modulação da expressão do PPARγ e das suas funções em células da microglia pela ANXA1. Foram empregadas células imortalizadas da linhagem BV2 (microglia murina), inalteradas ou transfectadas para redução da expressão de ANXA1, tratadas com ANXA1 exógena (recombinante - rANXA1) ou com agonista ou antagonista de PPARγ (pioglitazona e GW9662, respectivamente). Os resultados obtidos mostraram que: 1) tratamento com rANXA1 aumenta as expressões gênica (RT-PCR) e proteica (Western Blotting) de PPARγ, e ambas as expressões estão reduzidas em células com deficiência endógena de ANXA1, sendo que tal efeito foi revertido pela ação da rANXA1; 2) tratamento com rANXA1 não induz a expressão dos fatores de transcrição ligados a expressão de PPARγ: proteínas ligantes de elementos de resposta ao cAMP - CREB - e transdutores de sinais e ativadores de transcrição - STAT6 - (Western Blotting), mas os níveis de ambos os fatores estão reduzidos em células transfectadas, e tal efeito não foi revertido pelo tratamento com rANXA1; 3) tratamento com pioglitazona ou com rANXA1 individualmente aumenta a fagocitose de células PC12 apoptóticas (citometria de fluxo), mas o tratamento simultâneo não altera a fagocitose induzida por pioglitazona ou rANXA1; no entanto, tratamento com GW9662 inibiu a fagocitose induzida pelo tratamento com rANXA1; 4) o tratamento com rANXA1 aumenta a expressão de CD36 (citometria de fluxo); a expressão de CD36 está reduzida em células transfectadas e tal expressão não é revertida pelo tratamento com rANXA1. Em conjunto, os dados obtidos mostram a modulação da ANXA1 sobre PPARγ em células da micróglia, com possível ação sobre a fagocitose de células apoptóticas, e que a redução da expressão de ANXA1 reduz acentuadamente a expressão dos fatores de transcrição STAT6 e CREB, bem como a expressão de CD36. A elucidação dos efeitos resultantes destas alterações desencadeadas pela deficiência de ANXA1 endógena poderá contribuir para compreensão da fisiopatologia da neuroinflamação. / Inflammation is a key process in maintaining homeostasis and is essential for the body\'s response to injury. The inflammatory response must be proportional to the aggressor stimuli; in the central nervous system, a failed proper modulation leads to the development of different neurodegenerative diseases. Protein annexin A1 (ANXA1) and peroxisome proliferated-activated receptors (PPAR) control inflammation, as both inhibit development of inflammation and accelerate its resolution. Our research group has demonstrated that ANXA1 modulates PPARγ expression in macrophages. Thus, the present work investigated the modulation of PPARγ expression and its functions in microglia cells by ANXA1. In order to assess such, immortalized cells from cell line BV2 (murine microglia), either unadulterated or transfected for reduced expression of ANXA1, were treated with exogenous ANXA1 (recombinant protein - rANXA1) or either with PPARγ agonist or antagonist (pioglitazone and GW9662, respectively). The obtained results demonstrated that: 1) treatment with rANXA1 increases both gene (RT-PCR) and protein (Western Blotting) expressions of PPARγ, and also that both expressions are reduced in cells with endogenous deficiency of ANXA1, and such effect was reversed by the actions of rANXA1; 2) treatment with rANXA1 does not promote the expression of transcription factors associated with PPARγ expression: cAMP response element binding protein - CREB - and signal transductor and activator of transcription 6 - STAT6 (Western Blotting), but the expression levels of both factors are reduced in transfected cells, and such effect was not reversed by treatment with rANXA1; 3) individual treatment with pioglitazone or rANXA1 increases phagocytosis of apoptotic PC12 cells (flow cytometry), but simultaneous treatment does not affect pioglitazone/rANXA1-induced phagocytosis; however, treatment with GW9662 inhibited rANXA1-induced phagocytosis; 4) treatment with rANXA1 increases CD36 expression (flow cytometry); the expression of CD36 is reduced in transfected cells, and such expression is not reversed by treatment with rANXA1. The obtained data demonstrate the modulation ANXA1 exerts upon PPARγ in microglia cells, with a possible action upon phagocytosis of apoptotic cells, and that reduction of ANXA1 expression greatly reduces the expression of transcription factors STAT6 and CREB, as well as the expression of CD36. Elucidation of such effects that arise from a deficiency of endogenous ANXA1 will contribute to a better comprehension of the pathophysiology of neuroinflammation.
7

Die Analyse der Rolle von STAT6 im klassischen Hodgkin-Lymphom / Analysis of the role of STAT6 in classical Hodgkin´s lymphoma

Matthias, Kathrin 21 November 2011 (has links)
No description available.
8

Regulation of STAT6, STAT3 and STAT1 by the Cytoplasmic Tail of Polycystin-1, the Protein Affected in Polycystic Kidney Disease

Shivakumar, Vasanth 01 May 2007 (has links)
No description available.
9

Transcriptional regulation of 12/15-lipoxygenase expression and the implication of the enzyme in hepoxilin biosynthesis and apoptosis

Pattabhiraman, Shankaranarayanan 03 November 2003 (has links)
Die 12/15-Lipoxygenasen (12/15-LOXs) gehören zu einer heterogenen Klasse Lipid-peroxidierender Enzyme, deren biologische Rolle noch nicht vollständig geklärt ist. Eine Reihe experimenteller Daten deuten darauf hin, dass diese Enzym an Reifungs- und Differenzierungsprozessen beteiligt sind und auch für die Pathogenese verschiedener Erkrankungen (Asthma bronchiale, Entzündung, Atherosklerose) bedeutsam zu sein scheinen. Die Expression von 12/15-LOXs wird in vielen Säugetierzellen durch TH2-Zytokine reguliert und die Zytokin-induzierte Signaltransduktionskaskade verläuft über die Aktivierung van JAK-Kinasen und STAT6. Nach einer Stimulation von A549 Lungenkarzinomzellen mit Interleukin-4 (IL-4) kommt es erst nach 12 Stunden zu einer Hochregulation der 12/15-LOX mRNA Expression. Untersuchungen zum Induktionsmechanismus haben gezeigt, dass Genistein, ein Hemmstoff von Tyrosinkinasen, die Phosphorylierung von STAT6 und dessen Bindung an den Promoter des 12/15-LOX Gens verhinderte. Damit konnte die Induktion der katalytisch aktiven LOX geblockt werden. In Gegensatz dazu verhinderte Zykloheximid, ein spezifischer Hemmstoff der Proteinbiosynthese, die Expression der 12/15-LOX mRNA nicht, Dieses Ergebnis deutet darauf hin, dass die Neusynthese eines Transkriptionsfaktors im Rahmen der IL-4 induzierten Transduktionskaskade unwahrscheinlich ist. Weiterhin wurde beobachtet, dass IL-4 die zelluläre Histonacetyltransferase-Aktivität stark erhöhte und dass dieser Effekt überwiegend auf die enzymatische Aktivität des (CREB-bindenden Protein)-bindenden Proteins (CBP) zurückzuführen ist. Transfektion der Zellen mit E1A, einem viralen Protein, welches als Hemmstoff der Histonacetyltransferase-Aktivität von CBP/p300 bekannt ist, führte zu einer Unterdrückung der 12/15-LOX Expression. Andererseits stimuliert Natriumbutyrat, ein Hemmstoff der Histondeacetylase, die 12/15-LOX Synthese. Damit konnte gezeigt werden, dass die Acetylierung von Histonproteinen und von STAT6 ein essentieller Prozesse bei der IL-4 induzierten 12/15-LOX Expression in A549 Zellen ist. Weiterhin belegen diese Daten, dass sowohl die Phosphorylierung als auch die Acetylierung von STAT6 an der transkriptionellen Aktivierung des 12/15-LOX Gens beteiligt sind, obwohl beide Prozesse eine unterschiedliche Kinetik aufweisen. STAT6 Phosphorylierung erfolgt innerhalb der ersten Stunde nach IL-4 Stimulation, während die Acetylierungsreaktion zeitlich verzögert abläuft. Zusammenfassend kann die Signaltransduktionskaskade, die in A549 Zellen zur Expression der 12/15-LOX führt, wie folgt beschrieben werden: IL-4 induziert über die Aktivierung von JAK-Kinasen eine Phosphorylierung von STAT6, dessen Bindung an den 12/15-LOX Promotor jedoch zunächst durch nicht-acetylierte Histonproteine verhindert wird. Nach 9-11 Stunden werden Histone und der phosphorylierte STAT6 durch die Acetyltransferase-Aktivität von CBP/p300 acetyliert. Diese Reaktion führt zu einer Veränderung der Histonstruktur, wodurch die Bindung von modifizierten STAT6 und damit die Expression des 12/15-LOX Gens ermöglicht wird. Als wesentliche zellphysiologische Konsequenz der IL-4 induzierten 12/15-LOX Expression in A549 Zellen, wurde eine Apoptoseinduktion beobachtet. Das endogene 12/15-LOX Produkt 15-HETE bindet an den Kernrezeptor PPARg und induziert damit den programmierten Zelltod. Vorinkubation von A549 Zellen mit dem LOX-Hemmstoff NDGA oder der Einsatz von PPARg Dominant Negativ Vektor verhinderten die Apoptose. Mechanistische Untersuchungen zum Ablauf des durch IL-4 induzierten Zelltodes zeigten, dass der Prozess überwiegend dem extrinsischen Mechanismus folgt, bei dem Kaspasen-8 direkt zu einer Aktivierung der Effektorkaspase-3 führt. Der mitochondriale Mechanismus (Spaltung von Bid bzw. initiale Cytochrom C Freisetzung) scheint dabei nicht involviert zu sein. Die IL-4 induzierte Apoptose könnte von patho-physiologischer Bedeutung für den Verlauf von Lungenerkrankungen sein, bei denen Zellen mit hoher konstitutiver 12/15-LOX Expression, z.B. eosinophile Granulozyten, beteiligt sind. Hepoxiline sind bioaktive Mediatoren des 12/15-LOX Weges der Arachidonsäurekaskade, die durch Isomerisierung des primären Oxygenierungsproduktes 12S-HpETE gebildet werden. Zu Beginn unserer Untersuchungen war überwiegend unklar, welche Enzyme an der Isomerisierungsreaktion beteiligt sind. Bei der Suche nach geeigneten zellulären Modellen für die Untersuchung dieser Fragestellung fanden wir heraus, dass in den Ratteninsulinom-Zellen Rinm5F, die wegen ihres Mangels an Glutathionperoxidasen eine geringe Kapazität zur Reduktion von 12S-HpETE aufweisen, die Synthese von Hepoxilin A3 (HXA3) besonders hoch ist. Da wir vermuteten, dass 12/15-LOXs für die Isomerisierung von 12S-HpETE zu HXA3 verantwortlich sein könnten, verfolgten wir eine duale Forschungsstrategie um experimentelle Hinweise für unsere Arbeitshypothese zu finden. In den 12/15-LOX exprimierenden Rinm5F Zellen führte eine Immunopräzipitation mit 12/15-LOX spezifischen Antikörper zu einen vollständigen Verlust der 12/15-LOX- und der Hepoxilinsynthase-Aktivität eines Zelllysates. Beide Aktivitäten wurden fast vollständig im Immunopräzipitat wiedergefunden. 2. Transfektion von HeLa Zellen, die selbst keine 12/15-LOX exprimieren, mit 12/15-LOX und gleichzeitige Hemmung der zellulären Glutathionperoxidasen (Depletion von GSH mit Diethlmaleat) führte zu einer deutlichen zellulären Hepoxilinsynthese. Bei entsprechenden Kontrolltransfektanten wurde diese Aktivität nicht beobachtet. Weiterhin konnte festgestellt werden, dass rekombinante 12/15-LOXs (Expression in E. coli) in vitro eine intrinsische Hepoxinsynthase-Aktivität aufweisen, wenn 12S-HpETE als Substrat angeboten wird. Diese Daten belegen, dass 12/15-LOXs neben den bisher beschriebenen katalytischen Aktivitäten (Oxygenase, Hydroperoxidase, Leukotrienesynthase) auch Hepoxilinsynthase-Aktivität aufweisen. / 12/15-Lipoxygenases (human 15-LOX-1, rat 12/15-lipoxygenase) belong to family of lipid peroxidising enzymes. The enzyme has been implicated with roles in a variety of pathological conditions such as asthma, atherosclerosis, inflammation and in cellular differentiation. The enzyme expression in most human cell types is tightly regulated by Th2 cytokines, interleukin-4 (IL-4) and interleukin-13 (IL-13). Interleukin-4 (IL-4) induces expression of reticulocyte-type 15-lipoxygenase-1 (15-LOX-1) in various mammalian cells via the Janus kinase/signal transducer and activator of transcription 6 (STAT6) signaling system. 15-LOX-1 mRNA expression was first observed only 12h post IL-4 stimuation and required a minimum of 11h exposure to the cytokine. The mechanism of 15-LOX-1 induction in A549 lung epithelial cells and the observed delay was studied and it was found that genistein, a potent tyrosine kinase inhibitor, prevented phopsphorylation of STAT6, its binding to the 15-LOX-1 promoter, and the expression of catalytically active enzyme. In contrast, cycloheximide did not prevent 15-LOX-1 induction. Surprisingly, it was observed that IL-4 up-regulated the histone acetyltransferase activity of CREB-binding protein (CBP)/p300, which is responsible for acetylation of nuclear histones and STAT6. The acetylation of both proteins appears to be essential for the IL-4-induced signal transduction cascade, because inhibition of CBP/p300 by the viral wild-type E1A oncoprotein abrogated acetylation of both histones and STAT6 and strongly suppressed transcriptional activation of the 15-LOX-1 gene. Moreover, the inhibition by sodium butyrate of histone deacetylases, which apparently suppress 15-LOX-1 gene transcription, synergistically enhanced the IL-4-stimulated 15-LOX-1 expression. These data suggest that both phosphorylation and acetylation of STAT6 as well as acetylation of nuclear histones are involved in transcriptional activation of the 15-LOX-1 gene, although these reactions follow differential kinetics. STAT6 phosphorylation proceeds within the first hour of IL-4 stimulation. In contrast, CBP/p300-mediated acetylation requires 9-11 h, and similar kinetics were observed for the expression of the active enzyme. Thus, the results suggest that in the absence of IL-4, nuclear histones may be bound to regulatory elements of the 15-LOX-1 gene, preventing its transcription. IL-4 stimulation causes rapid phosphorylation of STAT6, but its binding to the promoter appears to be prevented by nonacetylated histones. After 9-11 h, when histones become acetylated, STAT6 binding sites may be demasked so that the phosphorylated and acetylated transcription factor can bind to activate gene transcription. The proinflammatory cytokine IL-4 is secreted in large amounts during allergic inflammatory response in asthma and plays a pivotal role in the airway inflammation. IL-4 has been shown to up-regulate 15-lipoxygenase and produce 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) in A549 cells via the Janus kinase/STAT6 pathway under coactivation of CREB binding protein/p300. IL-4 has also been shown to up-regulate peroxisome proliferator-activated receptor (PPARg ) nuclear receptors in macrophages and A549 cells. In this study it is observed that 15(S)-HETE binds to PPARg nuclear receptors and induces apoptosis in A549 cells. Moreover, pre-treatment of cells with nordihydroguaiaretic acid, a 15-lipoxygenase inhibitor, prevented PPARg activation and apoptosis. The latter was accomplished by the interaction of the 15(S)-HETE/PPARg complex with the adapter protein Fas-associating protein with death domain and caspase-8, as shown by transfection of Fas-associating protein with death domain dominant negative vector and cleavage of caspase 8 to active subunits p41/42 and p18. Whereas IL-4 and PPARg ligands failed to induce cleavage of Bid and release of cytochrome c from mitochondria, they caused translocation of the proapoptotic protein Bax from cytoplasm to mitochondria with a concomitant decrease in the Bcl-XL level. The cells were, thereofre, observed to follow the extrinsic pathway of apoptosis where caspase-8 directly activates the effector caspase-3, bypassing the mitochondria. The data also suggests that in IL-4-stimulated cells the 15(S)-HETE/PPARg complex down-regulates Bcl-XL, and the translocation of Bax to the mitochondria commits the cell to apoptosis. The IL-4-induced apoptosis may contribute to severe loss of alveolar structures and infiltration of eosinophils, mononuclear phagocytes, etc., into the lung tissue as observed in chronic asthma patients. The 12(S)-lipoxygenase (12-LOX) pathway of arachidonic acid (AA) metabolism after dioxygenation to 12(S)-hydroperoxy-eicosatetraenoic acid is bifurcated in a reduction route to formation of 12(S)-hydroxy-eicosatetraenoic acid (12-HpETE) and an isomerization route to formation of hepoxilins. Interestingly, rat insulinoma RINm5F cells, which are devoid of cytoplasmic glutathione peroxidase (cGPx)/phospholipid hydroperoxide glutathione peroxidase (PHGPx), were observed to produce solely hepoxilin A3 (HXA3). Since HXA3 synthesis was abolished in heat-denatured or cGPx- or PHGPx-transfected cells, suggesting that a HXA3 synthase activity regulated by cGPx/PHGPx is present. To confirm this assumption AA was incubated with HeLa cells overexpressing the rat 12/15-LOX. Neither HXA3 nor 12(S)-HETE were detected due to abundance of cGPx/PHGPx. But, pretreatment of transfected cells with diethyl maleate, an inhibitor of glutathione and PHGPx, restored HXA3 synthase and 12-LOX activities. Moreover, recombinant rat 12/15-LOX produced HXA3 when incubated with 12-HpETE. Further confirmation was obtained by immunoprecipitation with 12/15-LOX specific antibodies. Immunoprecipitation of Rinm5F lysates results in the depletion of hepoxilin synthase activity. The hepoxilin synthase activity was localised in the immunoprecipitated protein. Thus, cells containing rat 12/15-LOX also possess an intrinsic HXA3 synthase activity, which is activated by inhibition of cGPx/PHGPx. In normal cells HXA3 is down-regulated by cGPx/PHGPx, but, it is persistently activated in oxidatively stressed cells deficient in cGPx/PHGPx, such as Rinm5F. Furthermore, formation of corresponding epoxyhydroxy products was observed when 15-HpETE was used as substrate, indicating a broad range of specificity for the enzyme.
10

A Novel Mechanism for Prostate Cancer Progression: from Polo-like Kinase 1 to Epigenetics

Ruixin Wang (8082788) 05 December 2019 (has links)
<p>Prostate cancer is (PCa) the second leading cause of cancer death in males in the United State, with 174,650 new cases and 31,620 deaths estimated in 2019. Polo-like kinase 1 (PLK1) has been postulated to have a pro-tumorigenesis function, besides its critical role in regulation of cell cycle, and to be overexpressed in various types of human cancer, including prostate cancer (PCa). However, our understanding remains unclear regarding the pro-tumor properties of PLK1 partially due to a lack of proper animal model. Integrating our recently generated prostate-specific PLK1 knock-in genetically engineered mouse model (GEM) and the transcriptome data of human PCa patients, we identify an oncogenic role of PLK1 in the prostate adenocarcinoma progression, castration resistance and metastatic dissemination. To elucidate the underlying mechanism, we investigate the link between PLK1 and tumor microenvironment in PCa using the transgenic mouse model, and find that PLK1overexpression enable the macrophages polarization towards M2 phenotype via driving the activation of IL4/IL13/STAT6 pathway. These findings first validates PLK1 as a critical oncogene closely associated with PCa progression in vivo, and uncover a novel function of PLK1 to facilitate IL4/STAT6 signaling and M2 macrophage polarization. Importantly, these findings suggest an efficient therapeutic strategy targeting STAT6 for treatment of advanced PCa which usually possessing a high level of PLK1 expression. To further explore the molecular mechanism underlying PLK1-induced PCa progression and resistance to therapy, we turned our eyes to epigenetic modifications. It has been documented that epigenetic deregulation such as histone modification and DNA methylation contributes to PCa initiation and progression. Enhancer of zeste homologue 2 (EZH2), the catalytic subunit of Polycomb-repressive complex 2 (PRC2), plays a critical role in repressing gene expression by tri-methylation of histone 3 at lysine 27 (H3K27me3). Emerging data have demonstrated that there is a link between EZH2 and oncogenesis as EZH2-mediated methylation acts as an important factor in epigenetic silencing of tumor suppressor genes in cancer. Expression of EZH2 is often upregulated in castration-resistant prestate cancer (CRPC), thus EZH2 has been proposed as a target for CRPC. Importantly, it has been demonstrated that EZH2 becomes hyperphosphorylated in CPRC cells. Further, it has been shown that the oncogenic function of EZH2 is usually regulated by the post-translational modifications. PLK1 acting as a serine/threonine kinase to regulate multiple signaling pathways in human cancer, however, whether PLK1 is involved in EZH2 phosphorylation is not known. Herein, we show that Plk1 physically interacts with EZH2 and negatively regulates H3K27 trimethylation (H3K27me3). Furthermore, Plk1 can phosphorylate EZH2 at T144, and Plk1-mediated phosphorylation of EZH2 is involved in inhibiting EZH2 activity toward H3K27me3. More importantly, EZH2 phosphorylation by Plk1 is inhibitory for PRC2-mediated gene repression but required for transcriptional activation toward oncogenesis. Finally, by combination with Plk1 inhibitor BI2536, we show a robust sensitization of EZH2 inhibitors in CRPC cell lines, as well as in CRPC xenograft tumors. Our findings provide a new mechanism to define the oncogenic activity of EZH2 and suggest that inhibition of Plk1-mediated EZH2 activity may provide a promising therapeutic approach for CRPC.</p>

Page generated in 0.0243 seconds