• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 5
  • 2
  • Tagged with
  • 8
  • 8
  • 6
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Mechanismen der Apoptoseresistenz der Tumorzellen des klassischen Hodgkin Lymphoms / Mechanisms of resistance to apoptosis in classical Hodgkin Lymphoma tumor cells

Lietz, Andreas January 2006 (has links)
Apoptose, der programmierte Zelltod, spielt eine wichtige Rolle für das Gleichgewicht zwischen Proliferation und Sterben von Zellen und ist außerdem an der Beseitigung von infizierten und geschädigten Zellen beteiligt. Apoptose kann durch Stimulation von Rezeptoren aus der Familie der TNF-Rezeptoren wie CD95, ausgelöst werden. Nach Liganden-induzierter Trimerisierung der Rezeptoren bindet FADD an den zytoplasmatischen Teil des Rezeptors und rekrutiert Caspase-8 und/oder -10. Die räumliche Nähe der Caspasen in diesem als DISC bezeichneten Komplex führt zu ihrer auto- und transkatalytischen Spaltung und damit Aktivierung. Dadurch wird das apoptotische Programm gestartet, welches zum Tod der Zelle führt. Kontrolliert wird dieser Vorgang von einer Vielzahl anti-apoptotischer Proteine. Störungen in diesem System sind an der Entstehung einer Reihe von Krankheiten beteiligt. Die Blockade der Apoptoseinduktion kann zur Entstehung von Tumoren beitragen. Das klassische Hodgkin Lymphom ist eine maligne Erkrankung des lymphatischen Systems. Die Tumorzellen sind große, einkernige Hodgkin- oder mehrkernige Reed/Sternbergzellen (HRS-Zellen). Sie leiten sich von Keimzentrum-B-Zellen ab. In HRS-Zellen fehlt die Expression einer Vielzahl von typischen B-Zellmarkern, darunter die des B-Zellrezeptors. Solche B-Zellen sterben normalerweise während der Keimzentrumsreaktion durch Apoptose. An diesem Prozess ist CD95 beteiligt. In einer Reihe von malignen Erkrankungen wurden eine Herunterregulation der CD95-Expression oder Mutationen im CD95-Gen beobachtet. Es wird daher vermutet, dass CD95-induzierte Apoptose zur Entfernung von Tumorzellen beiträgt. Im Gegensatz dazu exprimieren sowohl primäre HRS-Zellen als auch etablierte HRS-Zelllinien in der Regel Wildtyp-CD95, sind aber trotzdem CD95-resistent. In dieser Arbeit konnte gezeigt werden, dass Komponenten des CD95-Systems, im Gegensatz zu anderen malignen Erkrankungen, in den HRS-Zellen hochreguliert sind, darunter CD95 selbst. In immunpräzipitierten DISCs von CD95-stimulierten HRS-Zellen wurde neben FADD und Caspase-8/-10 auch c-FLIP nachgewiesen. c-FLIP ist ein Caspase-8/-10-Homolog, das ebenfalls an FADD bindet, aber aufgrund fehlender katalytischer Aktivität die Aktivierung der Caspasen im DISC und damit die Apoptoseinduktion verhindert. Eine starke c-FLIP-Expression konnte in allen HRS-Zelllinien und in den HRS-Zellen nahezu aller untersuchter primärer Hodgkinfälle (55/59) gezeigt werden. Durch siRNA-vermittelte Herunterregulation von c-FLIP war es möglich, HRS-Zelllinien gegenüber CD95-induzierter Apoptose zu sensitivieren. Dies zeigt, dass die CD95-Rezeptor-induzierte Apoptose in den HRS-Zellen nicht strukturell, sondern funktionell inhibiert ist und c-FLIP stark zu dieser Inhibition beiträgt. Darüber hinaus konnte gezeigt werden, dass die c-FLIP-Expression in den HRS-Zellen von der konstitutiven Aktivität des Transkriptionsfaktors NF-κB abhängt, die charakteristisch für diese Zellen ist. Normalerweise wird NF-κB von Inhibitorproteinen, den IκBs, im Zytoplasma zurückgehalten. Diverse Stimuli können den IKK-Komplex aktivieren, der die IκBs an bestimmten Serinresten phosphoryliert. Dies hat die Ubiquitinylierung und den Abbau der IκBs zur Folge, wodurch NF-κB frei wird, in den Kern wandert und dort seine Zielgene aktiviert. Es wird angenommen, dass in HRS-Zellen ein konstitutiv aktiver IKK-Komplex und teilweise Mutationen der IκB-Proteine zur konstitutiven NF-κB-Aktivität beitragen. Zu den NF-κB-abhängigen Genen in den HRS-Zellen gehören solche mit anti-apoptotischer und Zellzyklus-treibender Wirkung. Die Inhibition der NF-κB-Aktivität in den HRS-Zellen führt zu Apoptose und eingeschränkter Proliferation. Von dreiwertigem Arsen ist bekannt, dass es die Induzierbarkeit des IKK-Komplexes inhibieren kann und damit letztendlich die Aktivierung von NF-κB. In dieser Arbeit konnte gezeigt werden, dass Arsen in HRS-Zellen den konstitutiv aktiven IKK-Komplex inhibiert. In Zelllinien mit intakten IκB-Proteinen führte dies zur NF-κB-Inhibition und Apoptoseinduktion. Die Reduktion der NF-κB-Aktivität ging mit der Herunterregulation von anti-apoptotischen und Proliferations-fördernden Zielgenen einher. Die ektope Überexpression von NF-κB hob die Apoptose-induzierende Wirkung von Arsen teilweise auf. Durch Arsen-Behandlung von Mäusen konnte das Tumorwachstum xenotransplantierter HRS-Zellen stark verlangsamt werden. In explantierten Tumorzellen konnte ebenfalls eine NF-κB-Inhibition nachgewiesen werden. Die NF-κB-Inhibition durch Arsen trägt also stark zur Apoptoseinduktion in den HRS-Zellen bei. Zusammengefasst zeigen die Ergebnisse dieser Arbeit, dass die Modulation der Apoptoseresistenz neue therapeutische Ansätze für die Behandlung des Hodgkin Lymphoms bieten könnte. Der Einsatz von Arsen ist dabei besonders interessant, da Arsen schon für die Behandlung anderer maligner Erkrankungen eingesetzt wird. / Apoptosis, the programmed cell death, is important for the balance between proliferation and dying of cells. It is also involved in the removal of infected and damaged cells. Apoptosis can be induced by stimulation of receptors of the TNF-receptor family like CD95. After ligand-induced trimerisation of these receptors, FADD binds to the cytoplasmic part of the receptor and recruits Caspase-8 and/or -10. The induced proximity of the caspases in this complex, called DISC, leads to their auto- and transcatalytic cleavage and subsequently to their activation. This starts the apoptotic program which leads to the death of the cell. A number of anti-apoptotic proteins control this process. The deregulation of this system is involved in a variety of diseases. The disruption of the apoptotic program can contribute to the development of tumors. Classical Hodgkin Lymphoma is a malignant disease of the lymphatic system, characterized by mononucleated Hodgkin or multinucleated Reed/Sternberg (HRS) cells. These tumor cells are derived from germinal-center B-cells. However, HRS cells lack the expression of typical B cell markers, such as the B-cell receptor. Usually, B-cells without B-cell receptor expression die by apoptosis during the germinal-center reaction. CD95 is involved in this process. It has been shown previously that in many malignant diseases CD95 is down-regulated or mutated, indicating that CD95 is involved in the removal of tumor cells. In opposite to these findings, primary HRS cells and Hodgkin-derived cell lines usually express wild type CD95, but are resistant to CD95 induced apoptosis. In this work, it could be demonstrated that in contrast to other malignant diseases components of the CD95 system are up-regulated in HRS cells, including CD95 itself. By immunoprecipitation it was shown that, in addition to FADD and Caspase-8/-10, c-FLIP is a component of the DISC in CD95-stimulated cells. c-FLIP is a caspase homolog which, like caspases, binds to FADD, but lacks proteolytic activity. It inhibits the activation of caspases in the DISC and thus prevents apoptosis induction. A strong c-FLIP expression was shown in all HRS cell lines and in HRS cells of nearly all investigated primary cases of Hodgkin Lymphoma (55/59). siRNA-mediated (small interfering RNA) down-regulation of c-FLIP sensitized HRS cell lines to CD95-induced apoptosis. This shows that the CD95 receptor-induced apoptosis in HRS cells is not structurally but functionally inhibited and that c-FLIP strongly contributes to this inhibition. In addition, it was shown that c-FLIP expression depends on the constitutive activity of the transcription factor NF-κB which is characteristic for HRS cells. Usually, NF-κB is sequestered in the cytoplasm by inhibitor proteins, the IκBs. A variety of stimuli can activate the IKK-complex which subsequently phosphorylates the IκBs, leading to their ubiquitinylation and degradation. The released NF-κB translocates to the nucleus where it activates the transcription of target genes. It is supposed that a constitutively activated IKK complex and, in some cases, mutated IκB proteins contribute to the constitutive NF-κB activity in HRS cells. To the NF-κB dependent genes in HRS cells belong those with anti-apoptotic and cell cycle promoting activities. Inhibition of the NF-κB activity in HRS cells leads to apoptosis and decreased proliferation. Trivalent arsenic is known to inhibit the induction of the IKK complex and thus the activation of NF-κB. In this work, it was shown that arsenic inhibits the constitutively active IKK complex in HRS cells. This led to an inhibition of NF-κB and induction of apoptosis in HRS cell lines with non-mutated IκB proteins. The NF-κB inhibition was accompanied by the down-regulation of anti-apoptotic and cell cycle promoting genes. Ectopic overexpression of NF-κB partially reverted the apoptotic effect of arsenic. Treatment of mice with arsenic reduced the growth of subcutaneously xenotransplanted HRS cells. In explanted tumor cells, a reduced NF-κB activity could be demonstrated following treatment with arsenic. Thus, the inhibition of NF-κB by arsenic contributes to the induction of apoptosis in HRS cells. Taken together, the results indicate that modulation of the apoptosis resistance may offer new strategies for the treatment of Hodgkin Lymphoma. Of particular interest is the application of arsenic because it is already used in the treatment of other malignant disorders.
2

The Role of c-FLIP in the Regulation of Apoptosis, Necroptosis and Autophagy in T Lymphocytes

He, Ming-Xiao January 2013 (has links)
<p>To maintain homeostasis, T lymphocytes die through caspase&ndash;dependent apoptosis. However, blockage of caspase activity in T lymphocytes does not increase cell survival. The loss of caspase 8 activity leads to programmed necrosis (necroptosis) upon T cell receptor (TCR) stimulation in T lymphocytes. Necroptosis is correlated with excessive macroautophagy, an intracellular catabolic process characterized by the sequestration of cytoplasmic compartments through double&ndash;membrane vacuoles. Meanwhile, the proper induction of macroautophagy is required for T lymphocyte survival and function. Cellular caspase 8 (FLICE)&ndash;like inhibitory protein (c&ndash;FLIP) promotes survival in T lymphocytes. c&ndash;FLIP suppresses death receptor&ndash;induced apoptosis by modulating caspase 8 activation. Whether this modulation plays a role in the regulation of necroptosis has yet to be studied. Additionally, overexpression of c&ndash;FLIP reduces autophagy induction and promotes cell survival in cell lines. It remains unclear whether c&ndash;FLIP protects primary T lymphocytes by regulating the threshold at which autophagy occurs. In this study, c&ndash;FLIP isoform&ndash;specific conditional deletion models were used to study the role of c&ndash;FLIP in necroptosis and autophagy in primary T lymphocytes.</p><p>Our results showed that the long isoform of c&ndash;FLIP (c&ndash; FLIP<sub>L</sub>) regulates necroptosis by inhibiting receptor interacting protein 1 (RIP&ndash;1). Upon TCR stimulation, c&ndash;FLIP<sub>L</sub>&ndash;deficient T cells underwent RIP&ndash;1&ndash;dependent necroptosis. Interestingly, though previous studies have generally described necroptosis in the absence of caspase 8 activity and apoptosis, pro&ndash;apoptotic caspase 8 activity and the rate of apoptosis were also increased in c&ndash;FLIPL&ndash;deficient T lymphocytes. Moreover, c&ndash; FLIP<sub>L</sub>&ndash;deficient T cells exhibited enhanced autophagy, which served a cytoprotective function. </p><p>Apoptosis can be induced by either death receptors on the plasma membrane (extrinsic pathway), or the damage of the genome and/or cellular organelles (intrinsic pathway). Previous studies in c&ndash;FLIP&ndash;deficient T lymphocytes suggested that c&ndash;FLIP promotes cell survival in the absence of death receptor signals. Independent of death receptor signaling, mitochondria sense apoptotic stimuli and mediate the activation of caspases. Whether c&ndash;FLIP regulates mitochondrion&ndash;dependent apoptotic signaling remains unknown. Here, by deleting the <italic>c&ndash;Flip <italic> gene in mature T lymphocytes, we showed a role for c&ndash;FLIP in the intrinsic apoptosis pathway. In naïve T cells stimulated with the apoptosis inducer, c&ndash;FLIP suppressed cytochrome c release from mitochondria. Bim&ndash;deletion rescued the enhanced apoptosis in c&ndash;FLIP&ndash;deficient T cells, while inhibition of caspase 8 did not. Different from activated T cells, there were no signs of necroptosis in c&ndash;FLIP&ndash;deficient naïve T cells. Together, our findings indicate that c&ndash;FLIP is a key regulator of apoptosis, necroptosis and autophagy in T lymphocytes.</p> / Dissertation
3

Smac Mimetic Compound Treatment Induces Tumour Regression and Skeletal Muscle Wasting

Vineham, Jennifer January 2014 (has links)
Of all of the cancer patients throughout the world, approximately 50% of them are affected to some degree by cachexia. This syndrome involves significant skeletal muscle wasting, loss of adipose tissue and overall decrease in body weight in patients, particularly those with lung, pancreatic and gastric cancers. Cancer-induced cachexia is characterized by the presence of increased cytokines, notably TNF-α, IL-1β and IL-6. Most patients suffering of cancer-induced cachexia experience increased toxicity in response to chemotherapy, leading to fewer rounds of treatment and thus impeding the patients’ chances for recovery. More research into effective treatments for cancer-induced cachexia would therefore be indispensable. The inhibitor of apoptosis proteins (IAPs) have emerged as important cancer targets, primarily because of their roles as caspase inhibitors and regulators of NF-κB signalling. Small molecule IAP antagonists known as Smac mimetic compounds (SMCs) are currently in stage I/II clinical trials. They function by targeting cIAP1 and cIAP2 (and to a lesser extent, XIAP) resulting in a cytokine mediated death response in cancer cells. SMCs induce the production of TNF-α, a cytokine with which SMCs can potently synergize. However, limited efficacy occurs in some cancer cell lines (presumably because TNF-α cannot be induced in an autocrine fashion) and an exogenous source of the cytokine, such as that induced by using an oncolytic virus, is required. Notably, TNF-α (initially known as “cachectin”) is known to play a significant role in the induction of skeletal muscle atrophy. We therefore wanted to examine the effects of TNF-α induction by SMC and oncolytic virus co-treatment on both tumour regression and skeletal muscle in tumour bearing mice. We investigated the effects of SMC treatment on Lewis Lung Carcinoma (LLC) and B16F10 melanoma cell lines, both of which have been shown to be established cachectic cancer cell lines. Our in-vitro analysis of LLC and B16F10 cells revealed that LLC cells are sensitive to SMC and TNF-α co-treatment whereas B16F10 cancer cells remain resistant. SMC treatment, in combination with an oncolytic virus, VSVΔ51, increased tumour regression and survival time in LLC tumour bearing mice. Based on findings from previous studies, we investigated the role of cellular FLICE-like inhibitory protein (c-FLIP) in the resistance of the B16F10 melanoma cell line to SMC treatment. We were able to determine that the down-regulation of c-FLIP sensitizes the B16F10 cells to SMC and TNF-α induced cell death. In extending these findings, we found that SMC treatment alone can cause skeletal muscle wasting in the tibialis anterior muscle of LLC tumour bearing mice. However, the atrophic response was observed to be minimal as documented by a slight but significant decrease (approximately 10%) in muscle fibre cross-sectional area. Moreover, no biochemical evidence of muscle atrophy, as visualized by changes in the expression of myosin heavy chain (MHC) and Muscle RING Finger protein 1 (MuRF1), was found. Regardless, we speculate that the impact of SMC treatment on muscle wasting would be transient and reversible, and propose that the benefits of such a combination immunotherapy would greatly outweigh the risks.
4

c-FLIP as a potent anticancer target : Enhancement of cancer cell apoptosis by compounds identified through virtual screening / c-FLIP comme une cible anticancéreuse : Restauration de la voie apoptotique des cellules cancéreuses par des nouveaux composés identifiés par criblage virtuel

Yaacoub, Katherine 27 April 2017 (has links)
FLIP (FLICE inhibitory protein) est une protéine anti-apoptotique qui a des identités de séquence partagées avec la protéine pro-apoptotique caspase-8. FLIP se trouve en compétence avec caspase-8 pour se fixer sur la protéine adaptatrice FADD, ce qui empêche l’activation de caspase-8 bloquant ainsi l'apoptose. Lors du développement des molécules interférant avec des protéines anti-apoptotiques, la recherche d'inhibiteurs de la protéine FLIP qui est surexprimée dans un très grand nombre de cancers, a échoué. Cela s'explique en partie par le fait que peu d'information structurelle de FLIP est actuellement disponible TRAIL est une cytokine de la famille TNFα. Elle est décrite pour activer des voies de signalisation conduisant à la mort cellulaire par apoptose. TRAIL a montré un grand intérêt dans la thérapie anticancéreuse, grâce à sa capacité d’induire la mort des cellules tumorales sans aucun effet sur les cellules normales. Cependant, l’efficacité de TRAIL est limitée par plusieurs mécanismes moléculaires. Un de ces mécanismes est la surexpression de FLIP qui fait compromettre l’utilisation thérapeutique de TRAIL. Le but principal de ce projet est de développer des nouvelles molécules capables d’inhiber la protéine FLIP dans les cellules tumorales, sans aucun effet sur la protéine homologue caspase-8. Après modélisation des protéines FLIP et caspase-8 sur la base de la structure cristallographique de FLIP viral et FADD respectivement, des premières expériences d’ancrage ou “docking” utilisant une banque de composés chimiques du «National Cancer Institute NCI » ont été réalisées. Les 9 molécules les plus intéressantes, étant comme sélectives pour FLIP et non caspase 8, ont été sélectionnées et testées sur des lignées de cancer de poumons surexprimant la protéine FLIP. Une co-administration de chacune des molécules inhibitrices de FLIP avec TRAIL a été faite pour vérifier la restauration de la voie apoptotique dans les cellules cancéreuses. Un test moléculaire de « Pull down assay » a été fait afin de confirmer l’inhibition de l’interaction de FLIP avec FADD. Finalement, l’évaluation de l’activité enzymatique des caspases a été étudiée pour vérifier la réactivation de la voie apoptotique après la combinaison de TRAIL avec les inhibiteurs de c-FLIP. En conclusion, la combinaison de TRAIL avec les inhibiteurs de FLIP aboutit à la restauration de la voie apoptotique dans des cellules cancéreuses. Ces composés nouvellement identifiés, peuvent servir ultérieurement comme des potentiels éléments des stratégies utilisées dans le domaine du traitement du cancer. / FLIP (FLICE Inhibitory Protein) is an anti-apoptotic protein which shares sequencesimilarity with the pro-apoptotic protein caspase-8. FLIP competes with caspase-8 for binding to the adaptor protein FADD (Fas-associated death domain), thus it inhibits caspase-8 activation, thereby blocking apoptosis. During the development of molecules interfering with anti-apoptotic proteins, searching for inhibitors of FLIP protein which is overexpressed in a very large number of cancers, has failed. This is partly due to the fact that little FLIP structural information is available at present. TRAIL is a member of TNFα superfamily. It has been described to activate the apoptotic signaling pathways. TRAIL showed great interest in anti-cancer therapy, due to its ability to induce tumor cell death without any effect on normal cells. However, the efficacy of TRAIL is limited by several molecular mechanisms. One of these mechanisms is the overexpression of FLIP which is able to compromise the therapeutic use of TRAIL. The main goal of this project is to develop novel inhibitory molecules able to interfere with FLIP in tumor cells without any effect on the homologous protein caspase 8. After the construction of FLIP and caspase-8 proteins on the basis of the crystallographic structure of the viral FLIP and FADD respectively, the first docking experiments using a chemical library of the National Cancer Institute NCI have been carried out. The most interesting molecules, being selective for FLIP versus caspase 8, were selected and tested on lung cancer cell lines that overexpress FLIP protein. Co-administration of FLIP inhibitors with TRAIL was performed to verify the restoration of the apoptotic pathway in cancer cells. A molecular test of "Pull down assay" was done in order to confirm the inhibition of the FLIP/FADD interaction. Finally, the evaluation of caspases activity was carried out to confirm the reactivation of the apoptotic machinery after TRAIL/FLIP-inhibitors combination. In conclusion, the combination of TRAIL with FLIP inhibitors resulted in apoptosis restoration in resistant tumor cells. These newly identified compounds may serve later as potential elements in cancer treatment field.
5

Klärung der Ursachen der Apoptoseresistenz von kutanen T-Zel-Lymphomen und Entwicklung therapeutischer konzepte

Braun, Frank Karl Horst 14 September 2011 (has links)
Kutane T-Zell-Lymphome (CTCL) stellen eine heterogene Gruppe von non-Hodgkin-Lymphomen dar. Die häufigsten Entitäten sind die Mycosis fungoides, das Sézary-Syndrom sowie CD30+ lymphoproliferativen Erkrankungen (cALCL). Todesliganden-vermittelte Apoptose ist auch für die Lymphozyten-Homöostase von essentieller Bedeutung. Zunächst wurden die CTCL-Zelllinien mit systemischen T-Zell-Lymphomzellen hinsichtlich ihrer Apoptosesensitivität verglichen. Hierbei zeigte sich eine ausgesprochene Resistenz aller kutanen Zelllinien gegenüber TRAIL- und TNF-α-induzierter Apoptose. Anhand der Aktivierung der Caspasensignalkaskade war eine frühe Blockierung der Apoptose nachweisbar. Überdies konnte der Verlust von CD95, TNF-R1, Caspase-10 sowie von Bid in verschiedenen CTCL-Zelllinien nachgewiesen werden. Die Apoptoseresistenz konnte hingegen mit einer generellen und starken Expression des anti-apoptotischen Proteins c-FLIP korreliert werden. Des Weiteren wurde der Einfluss von CD30-Stimulation auf die durch CD95-induzierte Apoptose in CD30+-Zelllinien untersucht. Hierbei zeigte sich, dass CD30 Stimulation zur Aktivierung von NF-kB und zu erhöhter c-FLIP Expression führt, was mit Apoptoseresistenz korrelierte. Die Bedeutung der in vitro-Ergebnisse zeigte sich auch durch eine weitgehende Parallelität dieser zu untersuchten Biopsieproben von CTCL-Patienten. Schließlich wurden verschiedene NSAIDs auf ihre Fähigkeit hin untersucht, die Expression von c-FLIP zu vermindern und Apoptose zu induzieren. Dies wurde sowohl in CTCL-Zelllinien als auch in Tumor-T-Zellen von CTCL-Patienten untersucht. NSAIDs aktivierten beide Apoptosesignalwege und Diclofenac sensitiviert überdies für TRAIL-vermittelte Apoptose. In Ihrer Gesamtheit vermittelt die vorliegende Arbeit einen Einblick in die komplexen Apoptoseregulationsmechanismen in kutanen T-Zell-Lymphomen und identifiziert c-FLIP als einen zentralen Resistenzfaktor sowie NSAIDs als mögliche CTCL-Therapieoption. / Cutaneous T cell lymphomas (CTCL) form a heterogeneous group of non-Hodgkin lymphomas. Its most frequent forms are Mycosis fungoides, Sézary syndrome and CD30-positive cutaneous anaplastic large-cell lymphoma. Death ligands critically contribute to lymphocyte homeostasis by induction of apoptosis and may further represent safeguard mechanisms to prevent lymphoma development and tumor growth. First, we analyzed induction of apoptosis by death ligands in CTCL cell lines and compared it to that in systemic T-cell lymphoma cells. This revealed for CTCL cells a pronounced resistance to death ligands. In particular, there was no caspase activation in non-responsive CTCL cells, indicating an early blockade of the apoptosis signal. Furthermore a loss of CD95, TNF-R1, caspase-10, as well as of Bid was found in several cell lines. Changes at the receptor expression level were largely ruled out, whereas, consistent and strong expression of c-FLIP was correlated with resistance. In addition, we investigated the CD30/CD95 crosstalk in CD95-sensitive cALCL cell lines. Experiments showed that CD30 ligation leads to NF-κB-mediated c-FLIP upregulation in cALCL cells, which in turn conferred resistance to CD95-induced apoptosis. Parallels with regard to expression of apoptosis regulators were seen in peripheral blood mononuclear cells and biopsies of CTCL patients. Finally, we evaluated non-steroidal anti-inflammatory drugs (NSAIDs) for their capacity to downregulate c-FLIP expression and apoptosis induction in CTCL cells as well as in tumor T cells from CTCL patients. NSAID treatment resulted in an activation of both apoptosis signaling pathways and, furthermore, Diclofenac pre-treatment sensitized for TRAIL-induced apoptosis. In conclusion, this study elucidates defects in apoptosis regulation, proved the significance of c-FLIP for the survival of CTCL cells and provides a rationale for the use of NSAIDs as a potentially new therapeutic option for CTCL patients.
6

Charakterizace vlivu senescence na indukci a regulaci smrti nádorových buněk / Charakterizace vlivu senescence na indukci a regulaci smrti nádorových buněk

Nováková, Gita January 2014 (has links)
4 Abstract Senescence is a specific cell state distinquished by cessation of cell division and proliferation and changes in gene expression. Normal cells enter senescence after distinct number of cell divisions or in case of an unrepairable damage. Senescence in cancer cells can be induced by subliminal stress as sublethal treatment with certain drugs. Senescent cancer cells persist in the tissue and may secrete a number of factors and nutrients affecting surrounding cells. Senescence can thus change the response of cancer cells to various apoptogens during cancer therapy. In this study, we focused on the elucidation of presumed differences between normal proliferating and senescent cancer cells in their response to selected apoptogens. Implementing bromodeoxyuridine (BrdU)-mediated replication stress in cancer cells derived from pancreatic (PANC-1) or mesothelioma (H28) tumors, we efficiently forced these cells to acquire senescent phenotype. We document that these senescent cells gain higher resistance to combined TRAIL and homoharringtonine (HHT) treatment and enhance sensitivity to other apoptogens such as FasL, camptothecin and mVES. These cells also showed increased expression of anti-apoptotic protein c-FLIP in senescent cells and changes in the expression of some Bcl-2 family proteins....
7

Pro- and antiapoptotic events in Herpes simplex virus type 1 (HSV-1) infection of immature dendritic cells

Kather, Angela 13 February 2012 (has links)
Herpes simplex virus Typ 1 (HSV-1) ist ein humanpathogenes Virus der Familie Herpesviridae. Für eine erfolgreiche Virusreplikation besitzt HSV-1 mehrere Gene, die in den meisten infizierten Zelltypen Apoptose verhindern. Im Gegensatz dazu führt die HSV-1 Infektion eines zentralen Zelltyps des Immunsystems, den unreifen dendritischen Zellen (iDCs), zu Apoptose. Dies könnte ein Aspekt der HSV-1 Immunevasion sein. Bisher waren die Ursachen der Apoptose von HSV-1 infizierten iDCs unzureichend aufgeklärt. Es wurde jedoch gezeigt, dass das antiapoptotische zelluläre Protein c-FLIP in HSV-1 infizierten iDCs reduziert ist. In dieser Arbeit wurde die c-FLIP Menge in iDCs erstmalig mit Hilfe von RNA Interferenz erfolgreich reduziert. Dies bestätigte die Bedeutung von c-FLIP für die Lebensfähigkeit von iDCs. Folglich könnte auch die Reduktion der c-FLIP Menge nach HSV-1 Infektion iDCs für Apoptose empfindlich machen. Die HSV-1 induzierte c-FLIP Reduktion erfolgte in späten Stadien der Infektion, abhängig von der ordnungsgemäßen Expression viraler „early“ und „leaky late“ Gene. Sie fand nicht auf RNA Ebene statt und war unabhängig vom Proteasom und der Bindung an den „death inducing signaling complex“. Stattdessen wurde c-FLIP wahrscheinlich von einer viralen oder zellulären Protease abgebaut. In dieser Arbeit wurde erstmals gezeigt, dass zusätzlich zu Veränderungen im zellulären Apoptosesignalnetzwerk der Mangel an einem antiapoptotischen viralen Faktor zur Apoptose von HSV-1 infizierten iDCs beiträgt. Eine Microarray Analyse der HSV-1 Genexpression ergab, dass HSV-1 Latenz-assoziierte Transkripte (LATs) in apoptotischen iDCs signifikant geringer exprimiert waren als in nicht-apoptotischen epithelialen Zellen. LATs besitzen in Neuronen und epithelialen Zellen eine antiapoptotische Aktivität. Diese könnte den Mangel an c-FLIP kompensieren. Übereinstimmend mit dieser Hypothese induzierte eine HSV-1 LAT-Deletionsmutante mehr Apoptose in iDCs im Vergleich zum Wildtyp-Virus. / Herpes simplex virus type 1 (HSV-1) is a human pathogen which belongs to the family Herpesviridae. HSV-1 encodes several genes, which serve to efficiently prevent apoptosis in most infected cell types, thereby ensuring successful virus replication. In contrast, HSV-1 infection of one central cell type of the immune system, immature dendritic cells (iDCs), results in apoptosis. This could be one aspect of HSV-1 immunevasion. So far, the mechanisms underlying apoptosis of HSV-1 infected iDCs were poorly defined. However, it has been shown that the antiapoptotic cellular protein c-FLIP is reduced in HSV-1 infected iDCs. In this work, the amount of c-FLIP was for the first time successfully reduced in iDCs by RNA interference. This confirmed the importance of c-FLIP for viability of iDCs. Therefore, it is likely that c-FLIP reduction after HSV-1 infection also sensitizes iDCs to apoptosis. HSV-1 induced c-FLIP reduction occurred at late stages of infection and was dependent on proper expression of early and leaky late virus genes. Furthermore, it was not operative at the RNA level and was independent from the proteasome and binding to the death inducing signaling complex. Rather, c-FLIP was presumably degraded by a viral or cellular protease. In this work it was shown for the first time, that in addition to changes in the cellular apoptosis signaling network, the lack of one antiapoptotic viral factor contributes to apoptosis of HSV-1 infected iDCs. HSV-1 latency-associated transcripts (LATs) were significantly lower expressed in apoptotic iDCs compared to non-apoptotic epithelial cells, determined by microarray analysis of HSV-1 gene expression. It is known that in neurons and epithelial cells, LATs possess a potent antiapoptotic activity. This could compensate the lack of c-FLIP. Consistent with this hypothesis, a LAT deletion mutant of HSV-1 induced more apoptosis in iDCs compared to the respective wild type virus.
8

Targeting breast cancer with natural forms of vitamin E and simvastatin

Gopalan, Archana 13 July 2012 (has links)
Breast cancer is the second leading cause of death due to cancer in women. A number of effective therapeutic strategies have been implemented in clinics to cope with the disease yet recurrent disease and toxicity reduce their effectiveness. Hence, there is a need to identify and develop more effective therapies with reduced toxic side effects to improve overall survival rates. This dissertation investigates the mechanisms of action of two natural forms of vitamin E and a cholesterol lowering drug, simvastatin, as a therapeutic strategy in human breast cancer cells. Vitamin E in nature consists of eight distinct forms which are fat soluble small lipids. Until recently, vitamin E was known as a potent antioxidant but emerging work suggests they may be resourceful agents in managing a number of chronic diseases including cancer. Anticancer properties of vitamin E have been identified to be limited to the γ- and δ- forms of both tocopherols and tocotrienols. Gamma-tocopherol ([gamma]T) and gamma-tocotrienol ([gamma]T3) have both already been identified to induce death receptor 5 (DR5) mediated apoptosis in breast cancer cells. Studies here show that similar to [gamma]T3, [gamma]T induced DR5 activation is mediated by c-Jun N-terminal kinase/C/EBP homologous protein (JNK/CHOP) proapoptotic axis which in part contributed to [gamma]T mediated dowregulation of c-FLIP, Bcl-2 and Survivin. Also, both agents activate de novo ceramide synthesis pathway which induces JNK/CHOP/DR5 proapoptotic axis and downregulates antiapoptotic factors FLICE inhibitory protein (c-FLIP), B-cell lymphoma 2 (Bcl-2) and Survivin leading to apoptosis. Simvastatin (SVA) has been identified to display pleiotropic effects including anticancer effects but mechanisms responsible for these actions have yet to be fully understood. In this dissertation, it was observed that simvastatin induced apoptosis in human breast cancer cells via activation of JNK/CHOP/DR5 proapoptotic axis and down regulation of antiapoptotic factors c-FLIP and Survivin which are in part dependent on JNK/CHOP/DR5 axis. The anticancer effects mediated by simvastatin can be reversed by exogenously added mevalonate and geranylgeranyl pyrophosphate (GGPP), implicating the blockage of mevalonate as a key event. Furthermore, work has been done to understand the factors responsible for drug resistance and identify therapeutic strategies to counteract the same. It was observed that development of drug resistance was associated with an increase in the percentage of tumor initiating cells (TICs) in both tamoxifen and Adriamycin resistant cells compared to their parental counterparts which was accompanied by an increase in phosphorylated form of Signal transducer and activator of transcription 3 (Stat3) proteins as well as its downstream mediators c-Myc, cyclin D1, Bcl-xL and Survivin. Inhibition of Stat3 demonstrated that Stat3 and its downstream mediators play an important role in regulation of TICs in drug resistant breast cancer. Moreover, SVA, [gamma]T3 and combination of SVA+[gamma]T3 has been observed to target TICs in drug resistant human breast cancer cells and downregulate Stat3 as well as its downstream mediators making it an attractive agent to overcome drug resistance. From the data presented here, the mechanisms responsible for the anticancer actions of [gamma]T, [gamma]T3 and SVA have been better understood, providing the necessary rationale to test these agents by themselves or in combination in pre-clinical models. / text

Page generated in 0.1042 seconds