Spelling suggestions: "subject:"autophagy"" "subject:"autophagy7""
241 |
The relationship between HIF-1α and autophagy activity in the hypoxic environment of breast cancerMills, Justin 03 1900 (has links)
Thesis (MSc)--Stellenbosch University, 2013. / ENGLISH ABSTRACT: Introduction:
Among the cancers that afflict females world-wide, neoplastic disease of breast tissue is the most frequently diagnosed form and the leading cause of cancer-related death. Conventional treatment entails the use of doxorubicin, an anticancer agent belonging to the anthracycline family of chemotherapeutic drugs. Cancer cells are becoming increasingly resistant to doxorubicin therapy. The existence of hypoxic zones, which is a common feature of solid tumours, has been shown to promote the selection of therapy resistant clones in proliferating cancer cells. By modifying cellular homeostasis, neoplastic cells are capable of tolerating the hypoxic insult and thriving within the hostile microenvironment of the tumour. This adaptation is known as ‘the hypoxic response’ and is mediated through the action of the transcriptional regulator, HIF-1. Its expression in cancer tissue has been associated with a dismal prognosis as it promotes the degree of malignancy to an advanced stage.
Hypothesis & Aims:
We hypothesized that the targeting of HIF-1α would circumvent the ‘protective’ hypoxic response conferred upon breast cancer and improve the cytotoxicity of doxorubicin treatment. In this study, the first aim was to identify the hypoxic conditions at which the MCF-7 breast cancer cell line manifests a doxorubicin-resistant phenotype. This was followed by examination of the molecular pathways contributing to the hypoxic resistance by elucidating the potential relationship with the hypoxic regulator HIF-1α. Once the involvement of HIF-1α was established, the next aim was to evaluate whether the attenuation of HIF-1α would terminate the resistant phenotype and sensitize the neoplastic MCF-7 cells to doxorubicin treatment. Finally, the reproducibility of the in vitro experiment and efficacy of treatments within an animal model was evaluated. 2-Methoxyestradiol is a naturally occurring metabolite originating from 17β-estradiol. It has recently been exploited as an anticancer agent due to its anti-proliferative and anti-angiogenic properties. Among its various mechanisms of action, this compound has been shown to inhibit the expression of HIF-1α. It is for this reason that this study employed 2-methoxyestradiol in the adjuvant therapeutic treatment, along with doxorubicin. Methods:
The in vitro experimental model employed the use of the breast adenocarcinoma estrogen receptor (ER-positive cell line, MCF-7. These neoplastic cells were propagated under standard culture conditions until reaching ~70-80% confluency, after which treatment commenced. The treatment regime comprised a 12 hour exposure to the doxorubicin (1 μM) chemotherapeutic agent, either alone or in combination with HIF-1α inhibitors, 2-methoxyestradiol (10 μM) or siRNA duplex (400 nM), with parallel incubations under normoxic (21%) and hypoxic (~0.1%) conditions. To serve as a positive control for HIF-1α expression, cells were treated with CoCl2 (100 μM). Molecular techniques employed included the Caspase-Glo® 3/7 Assay, western blotting, and the bioreductive MTT Assay. Mitochondrial integrity was assessed by live cell imaging/fluorescent microscopy. Cellular viability was monitored at all times. The experiment was then translated into a pre-clinical in vivo model where C57BL/6 mice bearing E0771 xenografts (4 week growth) were allocated into the following treatment groups: (1) control (2) doxorubicin (5 mg.kg-1), (3) 2-methoxyestradiol (45 mg.kg-1), and (4) the combination of the two previously mentioned groups. Body weight and the rate of tumour growth were monitored throughout the experiment. Results:
Treatment with CoCl2 effectively stabilized HIF-1α under normoxic conditions. 2-Methoxyestradiol was capable of attenuating HIF-1α expression under both normoxia and hypoxia as compared with siRNA transfection, which was only effective under normoxia. HIF-1α stabilization was accompanied by an increase in autophagy along with the morphological transformation of mitochondria from an elongated network to shorter disc-like forms. On the other hand, HIF-1α attenuation caused an induction in the expression of the apoptotic markers, cleaved caspase 3 and cleaved PARP, as well as the restoration of the normoxic morphology. The exposure of MCF-7 cells to 1 μM doxorubicin for 12 hours produced a differential effect in the bioreductive MTT assay between normoxic and hypoxic conditions (42.97 ± 3.095% vs. normoxic dox, p<0.01), while stimulating the apoptotic and autophagic pathways. Compared to the control, a significant expression of phospho-AMPK became evident at 21% O2, while the levels remained stable at ~0.1% O2 after doxorubicin exposure. Furthermore, chemotherapeutic treatment caused the morphology of the mitochondria to appear dot-like. Although the combination of the two drugs removed the differential effect witnessed in the MTT assay, there was no significant change when compared to doxorubicin. Levels of apoptotic cell death decreased under both oxygen conditions. While HIF-1α and autophagy decreased under normoxia, they remained elevated under hypoxia.
In the in vivo component of the study, the administration of doxorubicin and 2-methoxyestradiol, alone or in combination, did not affect the rate of tumour growth or induce systematic toxicity in any of the experimental mice. When drugs were administered separately, a decrease in apoptosis along with a concomitant increase in autophagy and p-AMPK expression became noticeable while neither treatment had any significant effect on the expression of HIF-1α. Adjuvant administration, however, was capable of attenuating HIF-1α along with autophagy.
Discussion:
By inducing (CoCl2) and inhibiting (2-methoxyestradiol; siRNA duplex) HIF-1α, it was established that the autophagic pathway in the in vitro experimental setting of this study was dependent on the expression of HIF-1α. The bioreductive MTT assay measures the metabolic state of a cell, which is an indirect indication of cellular viability. Based on this, hypoxia was shown to confer survival to neoplastic MCF-7 cells based on the differential effect witnessed after doxorubicin treatment.
Apart from the induction of apoptosis and its associated mitochondrial fragmentation, the chemotherapeutic drug increased the activation of the metabolic sensor, AMPK, which upregulated autophagy during normoxia. While this autophagic process may assist in the killing mechanism, we speculate that the autophagy upregulated under hypoxia may be responsible for the survival effect and is most likely dependent on HIF-1α.
In contrast to eliciting a synergistic cytotoxic effect, the combination of doxorubicin with 2-methoxyestradiol produced an antagonistic effect on cellular viability instead. We propose that under normoxia, the combined treatment may stimulate the MCF-7 neoplastic cells to enter a state of growth arrest, or senescence, since the results indicate that the decrease in HIF-1α-dependent autophagy did not significantly affect cellular viability. Under hypoxia, despite the incorporation of the pharmacological HIF-1α inhibitor (2-methoxyestradiol), the expression levels of HIF-1α remained unaffected. We speculate that this could be the result of a potentiated stabilization of HIF-1α caused by the build-up of ROS and TCA intermediates which may be the outcome of mitochondrial dysfunction inflicted upon adjuvant therapy under hypoxia. Furthermore, it is also likely that the slight mitogenic effect observed within the MTT assay may be caused by the conversion of 2-methoxyestradiol to a chemically-reactive estrogen derivative, possibly by the action of doxorubicin, and the fact that an ER-positive cancer cell line was employed in this study.
With regards to the in vivo experimental model, we speculated that the failure of the molecular changes to manipulate the growth of the tumour could have been the result of an ineffective time- and/or dose regime.
Conclusion:
We therefore reject our hypothesis based on the fact that an antagonistic rather than synergistic effect was witnessed when the tumorigenic MCF-7 cell line was treated with adjuvant therapy. The results warrant the need for extensive testing on the pharmacodynamics of 2-methoxyestradiol, and more informative techniques to compliment the study. / AFRIKAANSE OPSOMMING: Inleiding:
Borskanker is die mees algemeen gediagnoseerde kanker asook die hoof oorsaak van kanker-verwante sterftes in vrouens wêreldwyd. Konvensionele behandeling behels die toediening van doxorubicin, ‘n anti-kankermiddel wat aan die antrasiklien-familie van chemoterapeutiese middels behoort. Kankerselle begin egter toenemend weerstandbiedend raak teen doxorubicin behandeling. Daar is al bewys dat die voorkoms van hipoksiese sones, wat ‘n algemene eienskap van soliede tumore is, die seleksie vir weerstandbiedende klone van prolifererende kankerselle, veroorsaak. Neoplastiese selle kan hierdie hipoksiese toestande weerstaan en in hierdie ongunstige mikro-omgewing floreer deur sellulêre homeostase te modifiseer. Hierdie aanpassing staan bekend as die ‘hipoksiese respons’ en word bemiddel deur die aksies van die transkripsiefaktor reguleerder, HIF-1. Die verhoogde uitdrukking van HIF-1 in kankerweefsel word oor die algemeen geassosieer met ‘n swak prognose omdat dit die maligniteit vehoog.
Hipotese en Doelwitte:
Die hipotese van hierdie studie behels dus die volgende: Deur HIF-1α te inhibeer, sal die ‘beskermende’ hipoksiese respons wat in borskankerselle voorkom omseil kan word en sodoende die sitotoksisiteit van doxorubicin terapie verhoog. Die eerste doelwit van hierdie studie was dus om die hipoksiese kondisies te identifiseer waar MCF-7 selle ‘n doxorubicin-weerstandbiedende fenotipe vertoon. Daarna is die molekulêre paaie wat bydrae tot hierdie hipoksiese weerstand ondersoek asook hul moontlike verwantskap met die hipoksiese reguleerder, HIF-1α. Nadat die rol van HIF-1α bevestig is, was die volgende doelwit om te bepaal of die inhibisie van HIF-1α die weerstandbiedende fenotipe sal onderdruk en neoplastiese MCF-7 selle sal sensitiseer vir doxorubicin behandeling. Laastens is die herhaalbaarheid en effektiwiteit van behandeling in die in vitro eksperimente ook in ‘n diermodel getoets.
2-Methoxyestradiol is ‘n metaboliet van 17β-estradiol wat natuurlik in die liggaam voorkom. Dit is ook onlangs as ‘n anti-kanker middel geïdentifiseer as gevolg van die anti-verdelende en anti-angiogeniese eienskappe. Een van die eienskappe van 2-methoxyestradiol is dat dit ook die uitdrukking van HIF-1α kan onderdruk. Dit is dan ook vir hierdie rede dat 2-methoxyestradiol in hierdie studie as bykomende terapie saam met doxorubicin gebruik is. Metodes:
Die in vitro eksperimentele model behels die gebruik van ‘n borsadenokarsinoom, estrogeenreseptor (ER)- positiewe sellyn, MCF-7. Hierdie neoplastiese selle is onder standaard weefselkultuur omstandighede gekweek totdat konfluensie van ~70-80% bereik is, waarna behandeling begin het. Die behandelingsprosedure behels ‘n 12 uur blootstelling aan doxorubicin (1 µM) chemoterapeutiese middel alleen of in kombinasie met die HIF-1α inhibitore, 2-methoxyestradiol (10 µM) of siRNA duplex (400 nM) in normoksiese (21% O2) en hipoksiese (~0.1% O2) toestande. Die selle is ook met CoCl2 behandel wat gedien het as ‘n positiewe kontrole vir HIF-1α uitdrukking. Molekulêre tegnieke wat tydens hierdie studie gebruik is, sluit die “Caspase-Glo® 3/7” bepaling in, asook die westelike kladtegniek en die MTT bepaling. Mitochondriale integriteit is bepaal deur middel van lewende sel afbeeldings/fluoresensie mikroskopie. Sellewensvatbaarheid is ten alle tye gemonitor. Hierdie eksperment is verder ook in ‘n pre-kliniese in vivo model uitgevoer waar C57BL/6 muise met E0771 xenografte (4 weke groei) geïnduseer is en in die volgende behandelingsgroepe verdeel is: (1) kontrole; (2) doxorubicin (5 mg.kg-1); (3) 2-methoxyestradiol (45 mg.kg-1); en (4) die kombinasie van laasgenoemde twee groepe. Die liggaamsgewig en die tempo van tumorgroei is tydens die hele eksperiment gemonitor. Resultate:
CoCl2 behandeling het HIF-1α effektief gestabiliseer tydens normoksiese omstandighede. 2-Methoxyestradiol het HIF-1α uitdrukking tydens normoksiese en hipoksiese toestande onderdruk wanneer dit vergelyk is met siRNA transfeksie wat slegs tydens normoksiese toestande effektief was. HIF-1α stabilisering het gepaardgegaan met ‘n toename in autofagie asook morfologiese veranderinge in die mitochondria vanaf ‘n verlengde netwerk tot korter skyfagtige vorme. Aan die ander kant het HIF-1α onderdrukking ‘n toename in die apoptotiese merkers, nl kliewing in caspase-3 and PARP veroorsaak wat gepaard gegaan het met die herstel van die tubulêre mitochondriale netwerk. Die blootstelling van die MCF-7 selle aan 1 µM doxorubicin vir 12 ure het ‘n differensiële effek in die bioreduktiewe MTT bepaling tot gevolg gehad tussen normoksiese en hipoksiese toestande (42.97 ± 3.095%, p<0.1), terwyl die apoptotiese- en autofagiese paaie in beide toestande gestimuleer is. ‘n Insiggewende toename in fosfo-AMPK uitdrukking was sigbaar tydens normoksiese toestande van 21% O2, terwyl dit onveranderd gebly het tydens hipoksiese toestande van 0.1% ~O2 na doxorubicin behandeling. Die morfologie van die mitochondria het ‘n ‘kollerige’ voorkoms tydens doxorubicin behandeling gehad. Alhoewel die behandeling van die selle met beide middels gelyktydig, die differensiële effek soos weerspieël in die MTT bepaling ophef, is daar geen insiggewende verandering wanneer met doxorubicin behandeling vergelyk word nie. Apoptotiese seldood verminder met gelyktydige behandeling van biede middels tydens normoksiese en hipoksiese toestande. HIF1-α en autofagie het afgeneem tydens normoksiese toestande, maar bly vehoog tydens hipoksie. In die in vivo model, het die toediening van doxorubicin en 2-methoxyestradiol alleen en in kombinasie nie tumorgroei geaffekteer nie en ook nie sistemiese toksisiteit in enige van die eksperimentele muise tot gevolg gehad nie. Die afsonderlike toediening van die middels het ‘n afname in apoptose in ‘n toename in autofagie en p-AMPK uitdrukking tot gevolg gehad, terwyl afsonderlike toediening van die middels nie ‘n effek op HIF-1α uitdrukking gehad het nie. Die gelyktydige toediening van biede middels het egter ‘n onderdrukking van HIF1-α teweeggebring.
Bespreking:
Deur HIF-1α te induseer (CoCl2) en te inhibeer (2-methoxyestradiol en siRNA) in hierdie in vitro eksperimentele omstandighede, bevestig hierdie resultate dat autofagie afhanklik is van die uitdrukking van HIF-1α. Die bioreduktiewe MTT bepaling meet die metaboliese staat van die sel wat indirek sellewensvatbaarheid bepaal. Gebasseer op hierdie bepaling is bewys dat hipoksie ‘n weerstandbiedende fenotipe veroorsaak teen doxorubicin behandeling in neoplastiese MCF-7 selle.
Doxorubicin veroorsaak ‘n toename in apoptose met geassosieerde mitochondriale fragmentering asook ‘n aktivering van die metaboliese sensor, AMPK, wat autofagie stimuleer in normoksiese omstandighede. Alhoewel ‘n toename in autofagie seldood kan stimuleer, spekuleer ons dat ‘n toename in autofagie tydens hipoksie verantwoordelik kan wees vir seloorlewing wat heel moontlik ook afhanklik van HIF-1α is. In kontras met die verwagting dat die kombinasie behandeling ‘n sinergistiese sitotoksiese effek sou teweegbring, dui ons resultate dat daar ‘n antagonistiese effek op sellewensvatbaarheid was. Ons stel voor dat die gekombineerde behandeling tydens normoksiese toestande MCF-7 neoplastiese selle stimuleer om in ‘n toestand van groeistaking in te gaan aangesien die resultate daarop dui dat ‘n afname in HIF-1α afhanklike autofagie nie sellulêre lewensvatbaarheid beïnvloed het nie. Tydens hipoksie, ten spyte van die bykomdende behandeling met die HIF-1α inhibitor (2-methoxyestradiol), het die vlakke van HIF-1α onveranderd gebly. Ons spekuleer dat dat dit die gevolg kan wees van die stabilisering van HIF-1α as gevolg van ‘n toename in ROS en TCA intermediate wat die gevolg van mitochondriale wanfunksie kan wees tydens bykomende terapie onder hipoksiese toestande. Dit is ook moontlik dat die mitogeniese effek wat waargeneem is met die MTT bepaling die gevolg kan wees van die omsetting van 2-methoxyestradiol na ‘n chemiese-reaktiewe estrogeen derivaat; moontlik as gevolg van die aksie van doxorubicin en die feit dat die sellyn wat in hierdie studie gebruik is, ‘n ER-positiewe kankersellyn is.
Met verwysing na die in vivo eksperimentele model, spekuleer ons dat die molekulêre veranderinge wat nie in die tumorgroei weerspieël word nie, die resultaat van oneffektiewe tyds- en dosis behandelingswyses is, of foutiewe toediening van die middel kan wees. Gevolgtrekking:
Ons verwerp dus ons hipotese gebaseer op die feit dat bykomende (adjuvante) behandeling eerder ‘n antogonistiese effek as ‘n sinergistiese effek op seldood in MCF-7 selle het. Hierdie resultate regverdig die nodigheid van intensiewe toetsing op die farmakodinamika van 2-methoxyestradiol asook die gebruik van meer informatiewe tegnieke om hierdie studie te komplimenteer. / CANSA and Marie Stander
|
242 |
Elucidating the role of WDR47 in regulating neuronal migration, autophagy and tubulin dynamicsRoos, Marna 12 1900 (has links)
Thesis (MSc)--Stellenbosch University, 2014. / ENGLISH ABSTRACT: Introduction Normal cerebral cortex development depends on extensive neuronal migration during
embryogenesis, permitting the formation of accurate synaptic circuits and a highly ordered
laminar neocortex. The motility of a migrating neuron is achieved by a dynamic microtubule
cytoskeleton that alternates between states of stabilization/lengthening and
destabilization/shortening. This dynamic instability of the microtubule cytoskeleton is
controlled by numerous microtubule-stabilizing and -destabilising proteins that bind directly
to microtubules.
Autophagy (“self-eating”), a major bulk intracellular degradation system, involves the fusion
of autophagosomes with lysosomes, followed by proteolysis and recycling of cellular
constituents. Like neuronal migration, autophagy is a microtubule-dependent process. The
dynamic microtubule network serves as a track for autophagosomes to be transported to
the lysosomes.
WDR47 is a protein that is expressed in the brain during development, but of which the
function is largely unknown. Novel interactions have recently been identified between
Reelin and WDR47 and between the microtubule-destabilising protein superior cervical
ganglion 10 (SCG10) and WDR47. These findings suggest that WDR47 may be regulating
microtubule-dependent processes such as neuronal migration and autophagy. We
hypothesize that WDR47 may play a role in regulating neuronal migration and/or
autophagy, and that this regulation may be mediated by a tubulin stability-regulating role of
WDR47.
Aims and Methods Our aims are to assess the cellular localization of WDR47 in GT1-7 cells and to determine
whether WDR47 is able to influence neuronal migration, filopodia extension, surface
adhesion, ultra-structure, autophagy, tubulin stability, and tau or SCG10 protein levels.
GT1-7 neuronal cells were cultured under normal conditions and transfected with WDR47
siRNA for 24 hours, followed by western blot verification of the knock-down. A 36 hour neuronal in vitro cell migration assay was performed and images of the wound were
captured every 6 hours; the migration distances and the wound areas for the different time
points were measured and analysed. A 24 hour migration assay was performed, capturing
images every hour, and the direction of migration was determined. Scanning electron
microscopy (SEM) and transmission electron microscopy (TEM) were performed to analyse
neuronal surface morphology and ultra-structure. Western blot analysis of SCG10,
acetylated α-tubulin, Tau, LC3, and Sequestosome 1/p62 (SQTM1) protein levels was
performed. Super-resolution structured Illumination microscopy (SR-SIM) three-dimensional
(3-D) imaging of WDR47-YFP transfected cells, confocal microscopy of LC3 and acetylated
tubulin, co-localization analysis of WDR47 and acetylated tubulin, and fluorescence recovery
after photo-bleaching (FRAP) analysis were performed.
Results
WDR47 siRNA treatment significantly reduced the average migration distance and the
migration velocity, resulted in fewer filopodia-like extensions as well as perturbed surface
adhesion of migrating neurons, and lead to an increased presence of endoplasmic reticulum
(ER) structures as well as an expanded nuclear envelope. LC3-II protein levels were
significantly lower with WDR47 siRNA treatment, but were significantly increased with
WDR47 siRNA treatment in conjunction with Bafilomycin A1 treatment, indicating increased
autophagic flux. SCG10 protein levels were significantly decreased with WDR47 siRNA
treatment. SR-SIM and confocal microscopy of WDR47 siRNA treated cells revealed a robust
presence of highly convoluted acetylated tubulin in the perinuclear region as well as
decreased LC3 fluorescence signal. Confocal microscopy revealed co-localization of WDR47
with acetylated tubulin. - Discussion and Conclusion: The results suggest that WDR47 is involved in regulating neuronal migration, neuronal
surface adhesion and filopodia formation, microtubule dynamics, and likely also autophagic
flux. Taken together, we propose that WDR47 is regulating microtubule dynamics by
facilitating assembly of microtubule-regulating proteins such as SCG10, thereby affecting
microtubule-dependent processes such as neuronal migration and autophagy. / AFRIKAANSE OPSOMMING: Inleiding Normale serebrale korteks ontwikkeling is hoogs afhanklik van neuronale migrasie tydens
embriogenese, en is belanrik vir die vorming van akkurate sinaptiese netwerke en 'n hoogs
geordende laminêre neokorteks. Die vermoё van 'n neuron om te migreer berus op 'n hoogs
dinamiese mikrotubulien sitoskelet wat verleng/stabiliseer of verkort/destabiliseer soos
tubulien-eenhede begevoeg of verwyder word. Hierdie dinamiese onstabiliteit van die
mikrotubulien sitoskelet word beheer deur verskeie mikrotubulien-stabiliserende en -
destabiliserende proteïene wat direk bind aan mikrotubuliene.
Autofagie ("self-eet"), 'n grootmaat intrasellulêre degradasie stelsel, behels die fussie van
autofagosome met lisosome, gevolg deur proteolitiese afbraak van sellulêre organelle en
proteine. Soos neuronale migrasie is autofagie 'n mikrotubulien-afhanklike proses. Die
dinamiese mikrotubulien netwerk dien as 'n spoor vir die vervoer van autofagosome na
lisosome.
WDR47 is 'n proteïen wat voorkom in die brein tydens ontwikkeling, maar waarvan die
funksie grootliks onbekend is. Interaksies was onlangs geïdentifiseer tussen beide Reelin en
WDR47 en die mikrotubulien-destabiliserende proteïen SCG10 en WDR47. Hierdie
bevindinge dui daarop aan dat WDR47 n rol speel in die regulering van tubulienstabiliteit en
sodoende mikrotubulien-afhanklike prosesse. Ons veronderstel dat WDR47 'n rol kan speel
in die regulering van neuronale migrasie en/of autofagie en dat hierdie regulasie moontlik
afhanklik is van 'n tubulien-stabiliteit-regulerende rol van WDR47. - Doelwitte en Metodes: Ons doelwitte is om die sellulêre lokalisering van WDR47 in GT1-7 neurone te evallueer en
om te bepaal of WDR47 n effek het op neuronale migrasie, oppervlak adhesie en filopodia
formasie, ultra-struktuur, autofagie, tubulien-netwerke en -stabiliteit, en Tau of SCG10
proteïenvlakke. GT1-7 neuronale selle is gekweek onder normale omstandighede en vir 24 uur
getransfekteer met WDR47 siRNA, gevolg deur verifikasie met Western-blot analise. 'n 36
uur neuronale in vitro sel migrasie toets is uitgevoer en fotos van die wond is elke 6 uur
geneem. Die migrasie afstande en die wondareas vir die verskillende tydpunte is gemeet en
ontleed. 'N 24-uur-migrasie toets is uitgevoer, 'n foto van die wond is elke uur geneem, en
die rigting van migrasie is bepaal. Skandering elektronmikroskopie (SEM) en transmissieelektronmikroskopie
(TEM) is uitgevoer om neuronale oppervlakmorfologie en ultrastruktuur
te observeer. Western blot analise van SCG10, geasetieleerde α-tubulien, Tau, LC3
en Sequestosome 1/p62 (SQTM1) proteïenvlakke is uitgevoer. Super-resolusie
gestruktureerde verligting mikroskopie (SR-SIM) driedimensionele (3-D) beelding van
WDR47-YFP getransfekteerde selle, konfokale mikroskopie vir visualisering van LC3 en
tubulien, co-lokalisering analise van beide WDR47 en LC3 en WDR47 en tubulien, asook
fluorescentie hersteling na foto-bleek (FRAP) analise is uitgevoer.
Resultate
Die gemiddelde migrasie-afstand en die migrasiesnelheid (μm/min) het beduidend
afgeneem met WDR47 siRNA behandeling. SEM analise van WD47 siRNA-behandelde
neurone het minder filopodia en veranderde oppervlak adhesie vertoon, en TEM analise het
'n verhoogde teenwoordigheid van endoplasmiese retikulum (ER) strukture, en 'n
uitgebreide kernmembraan vertoon. LC3-II proteïenvlakke was beduidend laer met slegs
WDR47 siRNA behandeling, maar beduidend hoёr met WDR47 siRNA behandeling in
samewerking met Bafilomycin A1 behandeling. Hierdie resultate dui aan op toeneemende
autofagie met WDR47 siRNA behandeling. Verder, beduidend laer vlakke van SCG10
proteïenvlakke is waargeneem met WDR47 siRNA behandeling. SR-SIM en konfokale
mikroskopie van WDR47 siRNA behandelde selle het 'n robuuste teenwoordigheid van
hoogs buigende geasetieleerdetubulien in die area rondom die nukleus, 'n afgeneemde LC3 Bespreking en Gevolgtrekking
Die resultate dui daarop aan dat WDR47 betrokke is by die regulering van neuronale
migrasie, filopodia vormasie, oppervlak adhesie, mikrotubuliendinamika, en waarskynlik ook
autofagie. Ons stel voor dat WDR47 mikrotubuliendinamika afekteer deur die regulering van proteïene soos SCG10, en sodoende mikrotubulienafhanklike prosesse soos neuronale
migrasie en autofagie fasiliteer.
|
243 |
Regulation of osteoclast activation and autophagy through altered protein kinase pathways in Paget's disease of boneMcManus, Stephen January 2016 (has links)
Résumé : La maladie osseuse de Paget (MP) est un désordre squelettique caractérisé par une augmentation focale et désorganisée du remodelage osseux. Les ostéoclastes (OCs) de MP sont plus larges, actifs et nombreux, en plus d’être résistants à l’apoptose. Même si la cause précise de la MP demeure inconnue, des mutations du gène SQSTM1, codant pour la protéine p62, ont été décrites dans une proportion importante de patients avec MP. Parmi ces mutations, la substitution P392L est la plus fréquente, et la surexpression de p62P392L dans les OCs génère un phénotype pagétique partiel. La protéine p62 est impliquée dans de multiples processus, allant du contrôle de la signalisation NF-κB à l’autophagie. Dans les OCs humains, un complexe multiprotéique composé de p62 et des kinases PKCζ et PDK1 est formé en réponse à une stimulation par Receptor Activator of Nuclear factor Kappa-B Ligand (RANKL), principale cytokine impliquée dans la formation et l'activation des OCs. Nous avons démontré que PKCζ est impliquée dans l’activation de NF-κB induite par RANKL dans les OCs, et dans son activation constitutive en présence de p62P392L. Nous avons également observé une augmentation de phosphorylation de Ser536 de p65 par PKCζ, qui est indépendante d’IκB et qui pourrait représenter une voie alternative d'activation de NF-κB en présence de la mutation de p62. Nous avons démontré que les niveaux de phosphorylation des régulateurs de survie ERK et Akt sont augmentés dans les OCs MP, et réduits suite à l'inhibition de PDK1. La phosphorylation des substrats de mTOR, 4EBP1 et la protéine régulatrice Raptor, a été évaluée, et une augmentation des deux a été observée dans les OCs pagétiques, et est régulée par l'inhibition de PDK1. Également, l'augmentation des niveaux de base de LC3II (associée aux structures autophagiques) observée dans les OCs pagétiques a été associée à un défaut de dégradation des autophagosomes, indépendante de la mutation p62P392L. Il existe aussi une réduction de sensibilité à l’induction de l'autophagie dépendante de PDK1. De plus, l’inhibition de PDK1 induit l’apoptose autant dans les OCs contrôles que pagétiques, et mène à une réduction significative de la résorption osseuse. La signalisation PDK1/Akt pourrait donc représenter un point de contrôle important dans l’activation des OCs pagétiques.
Ces résultats démontrent l’importance de plusieurs kinases associées à p62 dans la sur-activation des OCs pagétiques, dont la signalisation converge vers une augmentation de
leur survie et de leur fonction de résorption, et affecte également le processus autophagique. / Abstract : Paget’s disease of bone (PDB) is a skeletal disorder characterized by focal and
disorganized increases in bone turnover. In PDB, osteoclasts are larger, more active, more numerous, and resistant to apoptotic stimuli. While no single root cause has been identified, mutations to the gene encoding the p62 protein, SQSTM1, have been described in a significant population of patients with PDB. Among these mutations, the P392L substitution is the most prevalent, and overexpression of p62P392L in osteoclasts generates at least a partial pagetic phenotype in vitro. Normally this protein mediates a number of cell functions, from control of NF-κB signaling to autophagy. In human osteoclasts, a multiprotein complex containing p62 and protein kinases PKCζ and PDK1 (the principal kinase of Akt), form in response to stimulation by receptor activator of nuclear factor kappa-B ligand (RANKL), the principal osteoclastogenic-signaling cytokine. We found that PKCζ is involved in RANKL-induced activation of NF-κB, and that it contributed to a basal activation of NF-κB observed in p62P392L mutants. This may be regulated in part by a PKCζ dependent increase in p65 phosphorylation at Ser536 which we characterized, independent of IκB. This could represent one alternative pathway by which mutant p62 leads to increased NF-κB activation.
We observed increased basal phosphorylation of survival regulators ERK and Akt in
PDB that was reduced upon PDK1 inhibition. The activity of 4EBP1 and Raptor, associated with mTOR activity, were also altered in pagetic osteoclasts and regulated by PDK1 inhibition. We then identified autophagic defects common to pagetic osteoclasts; with higher basal levels of LC3II (associated with autophagic structures), regardless of p62 mutation, and reduced sensitivity to autophagy induction in PDB. These results suggest an accumulation of non-degradative autophagosomes. Inhibition of PDK1 not only induced apoptosis in PDB and controls, but significantly reduced resorption in PDB, and with regards to autophagy, PDK1 inhibition was more potent in PDB than in controls. Therefore PDK1/Akt signaling represents an important checkpoint to PDB osteoclast activation.
In sum, these results demonstrate the importance of several p62-associated kinases
in the over-activation of pagetic osteoclasts, through increased survival and altered
signaling. As p62 mutations alone do not account for most cases of PDB, the
characterization of these pathways may identify a common factor linking pagetic
osteoclasts. Therefore these studies represent a novel approach to osteoclast apoptosis,
activation, and autophagy associated with PDB.
|
244 |
The role of novel protein-protein interactions in the function and mechanism of the sarcomeric protein, myosin binding protein H (MyBPH)Mouton, Jacoba Martina 04 1900 (has links)
Thesis (PhD)--Stellenbosch University, 2014. / ENGLISH ABSTRACT: Left ventricular hypertrophy (LVH) is a major risk factor for cardiovascular morbidity and mortality, and is a feature of common diseases, such as hypertension and diabetes. It is therefore vital to understand the underlying mechanisms influencing its development. However, investigating the mechanisms underlying LVH in such complex disorders can be challenging. For this reason, many researchers have focused their attention on the autosomal dominant cardiac muscle disorder, hypertrophic cardiomyopathy (HCM), since it is considered a model disease in which to study the causal molecular factors underlying isolated cardiac hypertrophy.
HCM is a heterogeneous disease that manifests with various phenotypes and clinical symptoms, even in families with the same genetic defects, suggesting that additional factors contribute to the disease phenotype. Despite the identification of several HCM-causing genes, the genetic factors that modify the extent of hypertrophy in HCM patients remain relatively unknown.
The gene encoding the sarcomeric protein, cardiac myosin binding protein C, cMyBPC (MyBPC3) is one of the most frequently implicated genes in HCM. Identification of proteins that interact with cMyBPC has led to improved insights into the function of this protein and its role in cardiac hypertrophy. However, very little is known about another member of the myosin binding protein family, myosin binding protein H (MyBPH). Given the sequence homology and similarity in structure between cMyBPC and MyBPH, we propose that MyBPH, like cMyBPC, may play a critical role in the structure and functionality of the cardiac sarcomere and could therefore be involved in HCM pathogenesis.
The present study aimed to identify MyBPH-interacting proteins by using yeast two-hybrid (Y2H) analysis and to verify these interactions using three-dimensional (3D) co-localisation and co-immunoprecipitation (Co-IP) analyses. We further hypothesized that both MyBPH and cMyBPC may be involved in autophagy. To test this hypothesis, both MyBPH and cMyBPC were analysed for co-localisation with a marker for autophagy, LC3b-II. The role of MyBPH and cMyBPC in cardiac cell contractility were analysed by measuring the planar cell surface area of differentiated H9c2 rat cardiomyocytes in response to β-adrenergic stress after individual and concurrent siRNA-mediated knockdown of MyBPH and cMyBPC. In the present study we employed a family-based genetic association analysis approach to investigate the contribution of genes encoding the novel MyBPH-interacting proteins in modifying the hypertrophy phenotype. This study investigated the hypertrophy modifying effects of 38 SNPs and haplotypes in four candidate HCM modifier genes, in 388 individuals from 27 HCM families, in which three unique South African HCM-causing founder mutations segregate.
Yeast two-hybrid analysis identified three putative MyBPH-interacting proteins namely, cardiac β-myosin heavy chain (MYH7), cardiac α-actin (ACTC1) and the SUMO-conjugating enzyme UBC9 (UBC9). These interactions were verified using both 3D co-localisation and Co-IP analyses. Furthermore, MyBPH and cMyBPC were implicated in autophagy, since both these proteins were being recruited to the membrane of autophagosomes. In addition, a cardiac contractility assay demonstrated that the concurrent siRNA-mediated knockdown of MyBPH and cMyBPC resulted in a significant reduction in cardiomyocyte contractility, compared to individual protein and control knockdowns under conditions of β-adrenergic stress. These results indicated that MyBPH could compensate for cMyBPC, and vice versa, further confirming that both these proteins are required for efficient sarcomere contraction.
Results from genetic association analyses found a number of SNPs and haplotypes that had a significant effect on HCM hypertrophy. Single SNP and haplotype analyses identified SNPs and haplotypes within genes encoding MyBPH, MYH7, ACTC1 and UBC9, which contribute to the extent of hypertrophy in HCM. In addition, we found that several variants and haplotypes had markedly different statistical significant effects in the presence of each of the three HCM founder mutations.
The results of this study ascribe novel functions to MyBPH. Cardiac MyBPC and MyBPH play a critical role in sarcomere contraction and have been implicated in autophagy. This has further implications for understanding the patho-etiology of HCM-causing mutations in the gene encoding MyBPH and its interacting proteins.
This is to our knowledge the first genetic association analysis to investigate the modifying effect of interactors of MyBPH, as indication of the risk for developing LVH in the context of HCM. Our findings suggest that the hypertrophic phenotype of HCM is modulated by the compound effect of a number of variants and haplotypes in MyBPH, and genes encoding protein interactors of MyBPH. These results provide a basis for future studies to investigate the risk profile of hypertrophy development in the context of HCM, which could consequently lead to improved risk stratification and patient management. / AFRIKAANSE OPSOMMING: Linker ventrikulêre hipertrofie (LVH) is 'n primêre risikofaktor vir kardiovaskulêre morbiditeit en mortaliteit asook 'n kenmerk van algemene siektes soos hipertensie en diabetes. Daarom is dit van kardinale belang om te verstaan wat die onderliggende meganismes is wat die ontwikkeling van LVH beïnvloed. Die ondersoek na die onderliggende meganismes wat lei tot LVH in sulke komplekse siektes is ‟n uitdaging. Om hierdie rede fokus baie navorsers hul aandag op die autosomaal dominante hartspier siekte, hipertrofiese kardiomiopatie (HKM), wat beskou word as 'n model siekte om die molekulêre oorsake onderliggend tot geïsoleerde kardiovaskulêre hipertrofie te ondersoek.
HKM is 'n heterogene siekte wat manifesteer met verskeie fenotipes en kliniese simptome, selfs in families met dieselfde genetiese defekte, wat impliseer dat addisionele faktore bydra tot die modifisering van die siekte fenotipe. Ten spyte van die identifisering van verskeie HKM-versoorsakende gene, bly die genetiese faktore wat die mate van hipertrofie in HKM pasiente modifiseer relatief onbekend.
Die geen wat kodeer vir die sarkomeriese proteïen, kardiale miosien-bindingsproteïen C (kMyBPC) is die algemeenste betrokke in HKM. Die identifisering van proteïene wat bind met kMyBPC het gelei tot verbeterde insigte tot die funksie van hierdie proteïen en die rol wat hierdie proteïen in hipertrofie speel. Ten spyte hiervan, is daar baie min inligting beskikbaar oor 'n ander lid van die miosien-bindingsproteïen families, miosien-bindingsproteïen H (MyBPH). Gegewe die ooreenstemming tussen die DNA basispaar-volgorde en struktuur tussen hierdie twee proteïene, stel ons voor dat MyBPH, net soos kMyBPC, 'n kritiese rol in die struktuur en funksie van die kardiale sarkomeer speel en kan daarom betrokke wees in die patogenese van HKM.
Die huidige studie het beoog om proteïene wat met MyBPH bind te identifiseer deur die gebruik van gis-twee-hibried (G2H) kardiale biblioteek sifting en om hierdie interaksies te verifieer met behulp van drie-dimensionele (3D) ko-lokalisering en ko-immunopresipitasie eksperimente. Ons het verder gehipotiseer dat beide MyBPH and kMyBPC betrokke kan wees in outofagie. Om hierdie hipotese te toets is beide MyBPH en kMyBPC geanaliseer vir ko-lokalisering met 'n merker vir outofagie, LC3b-II. Verder het ons beplan om die rol van MyBPH en kMyBPC in kardiale spiersel-sametrekking te ondersoek deur die oppervlak van gedifferensieerde H9c2 rot kardiomiosiete in reaksie op β-adrenergiese stres te meet, na individuele en gesamentlike siRNA-bemiddelde uitklopping van MyBPH en kMyBPC.
In hierdie studie het ons 'n familie-gebaseerde genetiese assosiasie analise benadering gevolg om vas te stel of MyBPH en gene wat kodeer vir die geverifieerde bindingsgenote van MyBPH bydra tot die modifisering van die hipertrofiese fenotipe. Die doel van hierdie studie was om die hipertrofiese effek van 38 enkel nukleotied polimorfismes (SNPs) en haplotipes in vier kandidaat HKM modifiserende gene in 388 individue van 27 HCM families te toets, waarin drie unieke Suid-Afrikaanse HKM-stigters mutasies segregeer.
G2H analise het drie verneemde MyBPH bindingsgenote geidentifiseer, naamlik miosien (MYH7), alfa kardiale aktien (ACTC1) en die SUMO-konjugerende ensiem UBC9 (UBC9). Hierdie interaksies is geverifieer deur middel van 3D ko-lokalisering en ko-immunopresipitasie analises. Verder is bewys dat MyBPH en kMyBPC betrokke is in outofagie, siende dat beide proteïene gewerf is tot die membraan van die outofagosoom. 'n Kardiale sametrekkings eksperiment het gevind dat die gesamentlike siRNA-bemiddelde uitklopping van MyBPH en kMyBPC 'n merkwaardige vermindering in die kardiomiosiet sametrekking veroorsaak het in reaksie op β-adrenergiese stres kondisies, in vergelyking met die individuele proteïen en kontrole uitkloppings eksperimente. Hierdie resultate bevestig dat MyBPH vir kMyBPC kan instaan en ook andersom, wat verder bevestig dat beide proteïene benodig word vir effektiewe sarkomeer sametrekking.
Resultate van die genetiese assosiasie studie het gevind dat 'n aantal SNPs en haplotipes 'n beduidende effek of HKM hipertrofie het. Enkel SNP en haplotipe analises in gene wat kodeer vir MyBPH, MYH7, ACTC1 en UBC9 het SNPs en haplotipes geidentifiseer wat bydra tot die omvang van hipertrofie in HKM. Verder het ons gevind dat sekere SNPs en haplotipes kenmerkend verskillende statisties beduidende effekte in die teenwoordigheid van elk van die drie HKM-stigter mutasies gehad het.
Die resultate van hierdie studie skryf twee nuwe funksies aan MyBPH toe. Kardiale MyBPC en MyBPH speel 'n kritiese rol in sarkomeer sametrekking en is betrokke in outofagie. Hierdie resultate het verdere implikasies vir die verstaan van die pato-etiologie van die HKM-veroorsakende mutasies in die MyBPH, MYH7, ACTC1 en UBC9 gene.
So vêr dit ons kennis strek is dit die eerste genetiese assosiasie studie wat die modifiserende effek van bindingsgenote van MyBPH ondersoek as risiko aanduiding vir die ontwikkeling van LVH in die konteks van HKM. Ons bevindinge bewys dat die hipertrofiese fenotipe van HKM gemoduleer word deur die komplekse effekte van SNPs en haplotipes in die MyBPH geen en gene wat MyBPH proteïen-bindingsgenote enkodeer. Hierdie resultate verskaf dus 'n basis vir toekomstige studies om die risiko profiel van hipertrofie ontwikkeling met betrekking tot HKM te ondersoek, wat gevolglik kan bydra tot die verbeterde risiko stratifikasie en pasiënte bestuur.
|
245 |
Cell death in hyppxic injury : signaling mechanisms and dynamics in the decision making processLoos, Benjamin 12 1900 (has links)
Thesis (PhD (Physiological Sciences))--University of Stellenbosch, 2009 / ENGLISH ABSTRACT: Three main morphologies of cell death have been described in the diseased
myocardium, type I, better known as apoptotic cell death, which is characterized by
cell shrinkage and chromatin condensation, type II, or cell death with autophagy,
presents a morphology with intracellular accumulation of autophagic vacuoles and
type III, better known as necrosis, is characterized by cellular swelling and rapid
loss in cellular membrane integrity. However, recent literature strongly argues
against rigid classifications in the context of cell death mechanisms but rather
suggests to adopt a view of cell death as a dynamic and integrative cellular
response. Furthermore, the contribution of autophagy in cell death or cell survival
is still poorly understood. Therefore the aims of this study were twofold: (i) to
characterize the contribution of each cell death type in context of the severity and
duration of an ischaemic insult and (ii) to determine whether manipulation of the
autophagic pathway affects the contribution of cell death and translates into
protection of the heart.
Rodent derived cardiac myoblast cells were grown in Dulbecco’s Modified Eagle’s
Medium (DMEM) supplemented with 10% fetal bovine serum (FBS), and incubated
under 5% CO2 conditions. Cells were submitted to protocols of 2, 4 and 8 hrs of
simulated ischaemia (SI) under hypoxic conditions in a humidified environment
containing 0.1% O2, 5% CO2 and the balance N2, followed by 1 hr of reperfusion
respectively. We employed a modified ischaemic buffer containing either 2-deoxy-
D-glucose, sodium dithionate or both, with the aim to create an ischaemic insult of
mild (mild SI), moderate (moderate SI) and severe (severe SI) character
respectively.
We evaluated the contribution of each cell death mode using a combination of
viability- and ATP assays. Molecular markers for each cell death process such as
LC3, PARP and HMGB1 were evaluated using 3-dimensional fluorescence
techniques as well as western blot analysis and flow cytometry. Next, autophagy was induced or inhibited prior to the ischaemic insult, using rapamycin and 3MA
respectively, and similar parameters were evaluated after 2 hours of mild or
moderate SI. Propidium Iodide exclusion and Fluorescence Resonance Energy
Transfer (FRET) in combination with mitochondrial inner membrane depolarization
were employed to assess the onset of cell death dynamically. Flow cytometry was
employed to evaluate the degree of protection. In addition, the ATP levels and
reactive oxygen species (ROS) were evaluated.
Our results strongly indicate a differential induction of cell death, which is
dependent on the severity and duration of the ischaemic insult. Mild SI led to the
induction of autophagy and apoptosis, whilst moderate or severe SI induced both
apoptotic and necrotic cell death without an indication of autophagy. Only mild SI,
but not moderate and severe SI, resulted in an ATP surge.
Moreover, our data provide direct evidence that increased autophagy delays the
loss of cellular membrane integrity and delays caspase-3 activation as well as
mitochondrial depolarization in ischaemic cardiomyocytes. Our results show a
profound effect of increased autophagy on the onset of apoptosis as well as
necrosis under simulated ischaemic conditions, providing cellular protection. This
ATP surge observed during mild SI was abolished with increased autophagy.
Furthermore, our results indicate a profound effect of autophagy on ROS
generation. Under normoxic conditions, increased autophagy induced a significant
decrease in ROS while the inhibition of autophagy significantly increased ROS
generation. However, when increasing or decreasing autophagy prior to the
ischaemic insult, ROS increased significantly in both scenarios.
The results suggest that the severity of ischaemia determines the mode of cell
death differentially. An increase in autophagic responsiveness and flux, as induced
through rapamycin treatment, provides a selective advantage for tissue against
injury, possibly by maintaining intracellular ATP levels through the provision of
metabolic substrates. Autophagy is described as an inherent cellular mechanism
v
which affects the onset of cell death and exhibits protective effects in the ischaemic
myocardium when upregulated prior to the ischaemic insult.
The protective effect of increased autophagy was mirrored in the isolated perfused
rat heart model, reflected by improved functional recovery during
ischaemia/reperfusion. / AFRIKAANSE OPSOMMING: Die drie belangrikste morfologiese beskrywings van seldood in die hart sluit die
volgende in: tipe I, beter bekend as apoptose wat gekenmerk word deur
selkrimping en chromatienkondensering, tipe II, of seldood deur middel van
autofagie wat gekenmerk word deur die intrasellulêre versameling van autofagiese
vakuole en tipe III, beter bekend as nekrose wat gekenmerk word deur sel swelling
en ‘n vinnige verlies aan membraanintegriteit. Onlangse literatuur waarsku egter
teen die onbuigsame klassifikasie van seldoodmeganismes en stel voor dat
seldood as ‘n dinamiese proses met integrerende sellulêre meganismes beskou
moet word. Die bydrae van autofagie in seldoodmeganismes word ook nog nie
goed verstaan nie. Die doel van hierdie studie is dus tweevoudig: (i) om die bydrae
van elke tipe seldood te bepaal in konteks van die felheid en tydperk van die
iskemiese ingryping en (ii) om te bepaal of the manupilering van autofagie ‘n
betekenisvolle bydrae lewer in seldoodmeganismes en sodoende tot beskerming
van die hart kan lei.
Kardiale mioblaste wat van rotweefsel afkomstig is, is in Dulbecco se
gemodifiseerde Eagle medium (DMEM), waarby daar 10% fetale kalfserum gevoeg
is en wat onderhewig was aan 5% CO2 toestande, onderhou. Selle was
onderhewig aan protokolle van 2, 4 en 8 ure gesimuleerde iskemie (SI) onder
hipoksiese toestande in ‘n humiditeitsomgewing wat 0.1% O2, 5% CO2 en die
balans N2 bevat. Daarna was die selle onderhewig aan 1 uur reperfusie. ‘n
Gemodifiseerde iskemiese buffer wat óf 2-deoksie-D-glukose óf natriumdithionaat,
of beide bevat, is gebruik om lig, matig en strawwe iskemiese toestande na te
boots. Die bydrae van elke tipe seldood is geëvalueer tydens bogenoemde toestande
deur gebruik te maak van ‘n kombinasie van sellewensvatbaarheid- en ATP
tegnieke. Molekulêre merkers, wat LC3, PARP en HMGB1 insluit, is gebruik om
deur middel van 3-dimensionele fluoresensie tegnieke, westelike kladtegnieke en
vii
vloeisitometrie die verskillende vorme van seldood te ondersoek. Autofagie is ook
geïnduseer en geïnhibeer voor die iskemiese ingryping, deur middel van
rapamycin en 3MA, respektiewelik om die rol van autofagie tydens seldood te
bepaal. Propidium iodite uitluiting en fluoresensie resonansie energie oordrag
(FRET) in kombinasie met ‘n merker vir mitochondriale binneste membraan
depolarisasie is gebruik om die aanvang van seldood dinamies te ondersoek.
Vloeisitometrie is gebruik om die graad van beskerming aan te dui, terwyl
intrasellulêre ATP vlakke en reaktiewe suurstof spesies (ROS) ook gemeet is.
Ons resultate het getoon dat daar ‘n differensiële indusering van seldood plaasvind
wat afhanklik is van die felheid en tydsduur van die iskemiese ingryping. ‘n Ligte
graad van iskemie lei tot die indusering van autofagie en apoptose, terwyl matige
en strawwe iskemie beide apoptose en nekrose induseer sonder autofagie. Verder
het slegs ‘n ligte graad van iskemie ‘n skerp styging in ATP tweeggebring, terwyl
dit nie die geval was tydens matige en strawwe iskemie nie.
Ons data verskaf ook direkte bewyse dat ‘n toename in autofagie die verlies van
sellulêre membraanintegriteit vertraag. Dit het ook ‘n vermindering in caspase-3
aktivering en mitochondriale depolarisasie in iskemiese kardiomiosiete
teweegebring. Die data dui aan dat ‘n toename in autofagie ‘n beduidende effek op
apoptose en nekrose tydens gesimuleerde iskemiese toestande het om sodoende
selbeskerming te verskaf. Die skerp styging in ATP wat tydens die ligte graad van
iskemie teweeggebring is, is opgehef met ‘n toename in autofagie. Ons resultate
dui ook daarop dat autofagie ‘n beduidende rol in ROS generering speel. Onder
normoksiese omstandighede veroorsaak ‘n toename in autofagie ‘n insiggewende
afname in ROS generering, terwyl die inhibisie van autofagie ROS generering
insiggewend laat toeneem. Wanneer autofagie egter voor die iskemiese ingryping
verhoog of verlaag word, vermeerder ROS generering in beide gevalle.
Hierdie resultate bewys dat die graad van iskemie ‘n invloed het op die tipe
seldood wat geïnduseer word. ‘n Toename in autofagie reaksietyd en vloei, soos
viii
bewerkstellig deur rapamycin, verskaf ‘n selektiewe voordeel vir weefsel teen
beskadiging, heel waarskynlik deur die handhawing van intrasellulêre ATP-vlakke
deur die verskaffing van metaboliese substrate. Autofagie word beskryf as ‘n
inherente sellulêre meganisme wat seldood beïnvloed en die iskemiese
miokardium beskerm wanneer dit opgereguleer word voor die iskemiese ingryping.
Hierdie beskermende rol van autofagie wat in die weefselkultuur waargeneem is, is
ook in die geïsoleerde geperfuseerde rot hart model waargeneem, waar
funksionele herstel verbeter is tydens iskemie/reperfusie.
|
246 |
The Role of Phosphatidylinositol-3 Kinases and Phosphatidylinositol Phosphatases in T Cell Intracellular Homeostasis and AutophagyMcLeod, Ian Alexander January 2013 (has links)
<p>The homeostasis of naïve T lymphocytes is maintained by several mechanisms involving basal TCR and cytokine signaling, and nutrient factors. One of the common net results of these input signals is the production and stabilization of anti-apoptotic Bcl-2 family members. A second result of these processes is the induction of autophagy, an intracellular, catabolic, lysosomal targeting pathway. Autophagy induction in most systems involves the class III phosphatidylinositol-3 kinase (PI3K), Vps34, to produce phosphatidylinositol-3-phosphate (PI(3)P). To test this in T lymphocytes, I generated mice specifically lacking Vps34 in T cells (Vps34f/fLck-cre mice). However, Vps34-deficient T lymphocytes have normal levels of basal autophagy, and upregulate autophagy normally in response to cytokine or nutrient withdrawal, or TCR stimulation. Therefore I conclude that Vps34 activity is not required for autophagy induction in T lymphocytes. T lymphocytes lacking Vps34 do have enhanced rates of apoptosis, but this is due to defects in intracellular trafficking, specifically of the Interleukin-7 receptor alpha subunit (IL-7Rα). Additionally, multivesicular body (MVB) maturation is impaired in T cells lacking Vps34 such that extracellular ligands are not efficiently targeted to the lysosome. </p><p>Autophagy induction in Vps34-deficient T lymphocytes is still sensitive to pan-PI3K inhibitors, such as wortmannin and 3-methyladenine (3MA). Therefore, I hypothesized that other classes of PI3K are necessary to induce autophagy in T lymphocytes through the production of PI(3)P. Autophagy induction is sensitive to specific class I PI3K (PI3KI) inhibitors, such as PIK75. Additionally, T cells lacking the p85 regulatory subunit of PI3KI also have severe defects in T cell receptor (TCR) mediated autophagy induction. PI3KI activity results in the production of PI(3,4,5)P3, though, and not PI(3)P. Because of this specificity, I hypothesize that additional inositol polyphosphatases (Inpp) are required for autophagy induction downstream of PI3KI activity. Indeed, utilizing both inhibitors of pharmacological inhibition and siRNA-mediated knockdown of two classes of phosphatidylinositol phosphatases, inositol polyphosphate-4-phosphatase (Inpp4) and SH2 containing inositol phosphatase (SHIP), had dramatic impacts on autophagy induction. Furthermore, exogenous addition of PI(3,4)P2, a hypothesized intermediate in this pathway, positively regulates autophagy induction and leads to enhanced progression of autophagy. These observations indicate that PI3KI activity, linked to Inpp activity, are necessary and positive regulators of autophagy through the production of PI(3)P.</p> / Dissertation
|
247 |
p97 Negatively Regulates NRF2 by Extracting Ubiquitylated NRF2 from the KEAP1-CUL3 E3 ComplexTao, Shasha, Liu, Pengfei, Luo, Gang, Rojo de la Vega, Montserrat, Chen, Heping, Wu, Tongde, Tillotson, Joseph, Chapman, Eli, Zhang, Donna D. 15 April 2017 (has links)
Activation of the stress-responsive transcription factor NRF2 is the major line of defense to combat oxidative or electrophilic insults. Under basal conditions, NRF2 is continuously ubiquitylated by the KEAP1-CUL3-RBX1 E3 ubiquitin ligase complex and is targeted to the proteasome for degradation ( the canonical mechanism). However, the path from the CUL3 complex to ultimate proteasomal degradation was previously unknown. p97 is a ubiquitin-targeted ATP-dependent segregase that extracts ubiquitylated client proteins from membranes, protein complexes, or chromatin and has an essential role in autophagy and the ubiquitin proteasome system ( UPS). In this study, we show that p97 negatively regulates NRF2 through the canonical pathway by extracting ubiquitylated NRF2 from the KEAP1-CUL3 E3 complex, with the aid of the heterodimeric cofactor UFD1/NPL4 and the UBA-UBX containing protein UBXN7, for efficient proteasomal degradation. Given the role of NRF2 in chemoresistance and the surging interest in p97 inhibitors to treat cancers, our results indicate that dual p97/NRF2 inhibitors may offer a more potent and long-term avenue of p97-targeted treatment.
|
248 |
Multiple Approaches to Novel GSD Ia TherapiesLandau, Dustin James January 2016 (has links)
<p>Glycogen storage disease type Ia is an autosomal recessive disorder caused by a mutation in the glucose-6-phosphatase (G6Pase) catalytic subunit, encoded in humans by G6PC. G6Pase dephosphorylates glucose-6-phosphate (G6P) in the liver to generate glucose that can be shuttled to the bloodstream to maintain normoglycemia. Patients with GSD Ia typically present at 6 months of age with sever hypoglycemia, which is lethal if untreated. The current treatment is a strict dietary regimen in which children must be fed every 2 hours overnight or given nasogastric tube feeding, and adults must consume uncooked cornstarch around the clock to maintain normal blood sugar levels. This treatment maintains survival but fails to prevent other symptoms related to metabolism of the excess G6P, and patients develop hepatic adenomas that may become hepatocellular carcinoma later in life, in addition to progressive renal complications.</p><p>To overcome the problems persisting during dietary therapy, the Koeberl lab has sought to develop gene therapy approaches that use adeno-associated virus (AAV) vectors to replace the G6pase activity, restoring normoglycemia and normal metabolic processes. However, the vast majority of AAV-delivered genetic material exists as episomes that do not replicate as cells divide, so the effects of AAV gene therapy on GSD Ia mouse and dog models have proven temporary. We hypothesized that driving integration of therapeutic vector genomes into an affected individual's genome would improve beneficial effects' longevity.</p><p>We tested several approaches to accomplish this, and have found positive effects using a zinc finger nuclease (ZFN) that targets the mouse safe harbor ROSA26 locus to induce homologous recombination of the G6PC donor vector into the mouse genome. We were able to see an improvement in mouse survival to 8 months of age, an increase in G6Pase activity at 3 months of age, and a decrease in glycogen accumulation at 3 months of age, when the ZFN vector is administered alongside the G6PC vector, compared with mice that received the G6PC vector alone.</p><p>We have also taken an alternative approach to overcoming the long-term complications of the current dietary treatment, which would augment rather than replace the current treatment. We have examined several drugs known to induce autophagy in other disease models or cell culture systems, to determine if we could manipulate autophagic activity in G6PC knockdown hepatocytes or GSD Ia mice. We have found positive results using rapamycin, a well-studied MTOR inhibitor, in mice and cells, and have screened several other drugs as well, finding positive effects for bezafibrate, mifepristone, carbamazepin, and lithium chloride, in terms of lipid reduction (which accumulates as a symptom of GSD Ia) and/or LC3-II enhancement, which is reduced in GSD Ia due to downregulation of autophagy during G6P accumulation.</p> / Dissertation
|
249 |
Network pharmacology of the MPP+ cellular model of Parkinson's diseaseKeane, Harriet January 2015 (has links)
Parkinson's disease (PD) is an incurable neurodegenerative motor disorder caused by the inexorable loss of dopamine neurones from the substantia nigra pars compacta. Cell loss is characterised by the perturbation of multiple physiological processes (including mitochondrial function, autophagy and dopamine homeostasis) and much of this pathophysiology can be reproduced in vitro using the mitochondrial toxin MPP+ (1-methyl-4-phenylpyridinium). It was hypothesised that MPP+ toxicity could be modelled using protein-protein interaction networks (PPIN) in order to better understand the interplay of systems-level processes that result in eventual cell death in MPP+ models and PD. Initially, MPP+ toxicity was characterised in the human, dopamine-producing cell line BE(2)-M17 and it was confirmed that the neurotoxin resulted in time and dose dependent apoptosis. A radio-label pulse-chase assay was developed and demonstrated that MPP+ induced decreased autophagic flux preceded cell death. Autophagic dysfunction was consistent with lysosome deacidification due to cellular ATP depletion. Pertinent PPINs were sampled from publically available data using a seedlist of proteins with validated roles in MPP+ toxicity. These PPINs were subjected to a series of analyses to identify potential therapeutic targets. Two topological methods based on betweenness centrality were used to identify target proteins predicted to be critical for the crosstalk between mitochondrial dysfunction and autophagy in the context of MPP+ toxicity. Combined knockdown of a subset of target proteins potentiated MPP+ toxicity and the combined resulted in cellular rescue. Neither of these effects was observed following single knockdown/overexpression confirming the need for multiple interventions. Cellular rescue occurred via an autophagic mechanism; prominent autophagosomes were formed and it was hypothesised that these structures allowed for the sequestration of damaged proteins. This thesis demonstrates the value of PPINs as a model for Parkinson's disease, from network creation through target identification to phenotypic benefit.
|
250 |
The function of NOD2 in antigen presenting cellsAllan, Philip James January 2012 (has links)
Crohn’s disease (CD), a chronic inflammatory condition of the gut affecting 1:1000 of western populations, is thought to arise from a dysregulated immune response in a genetically susceptible individual. Polymorphisms in the ligand recognition domain of an intracellular pattern recognition receptor (PRR), NOD2, remain the strongest genetic risk factor for the development of CD. NOD2 directs autophagy in human DCs to facilitate bacterial destruction and antigen presentation; the CD-variant-NOD2 shows defects in this pathway. Recent work in the laboratory has demonstrated NOD2 signals to control expression induction of microRNA-29, which is impaired in cells from CD patients expressing CD NOD2-variants, and among other immunoregulatory effects, microRNA-29 suppresses IL-12p40/IL-23. Thus NOD2 directs key anti-microbe and immunoregulatory functions whose breakdown in the presence of CD-variant-NOD2 could act as a trigger for inflammation in this disease. In comparison with other PRRs, relatively little is understood of the hardwiring of NOD2 signalling, the mechanism of NOD2-mediated autophagy induction, the means by which NOD2 recruits a signalling platform within the cytosol and the mechanism of synergy with other PRRs and the inflammasome. In this work I used quantitative proteomics to map the NOD2 signalling cascade and its cross-talk with TLR2, demonstrating novel mediators: LCP1, a plastin, reduced phagocytosis of bacteria but did not alter bacterial killing, and aided control of the release of MCP1 and may be involved in IL-12p40 release. SHP1, a phospho-tyrosine phosphatase, is required for the propagation of signalling cascades via p38, p44/42 MAPK and NF-κB. It controls release of MIP1β and IL-12p40. HMGB1, an alarmin, is dephosphorylated on NOD2 stimulation and would result in changes to cellular location of HMGB1. Lastly, DAPK1, a serine/threonine kinase, is associated with HLA-DM loading compartment on NOD2 triggering, but does not alter CLIP levels on the surface of the cells. Thus, defining the hard wiring of NOD2 signalling in healthy donors, in comparison with CD donors expressing variant NOD2, is important to define targets amenable to drug design within this pathway.
|
Page generated in 0.0509 seconds