110 resultados para cytoprotective
Resumo:
Apolipoprotein J (ApoJ) ist ein sezerniertes heterodimeres 80kDa Glykoprotein mit zytoprotektiven und antiinflammatorischen Eigenschaften, das ein nahezu ubiquitäres Expressionsmuster aufweist. Eine stark erhöhte ApoJ-Expression ist mit neurodegenerativen Erkrankungen, Atherosklerose, myokardialem Infarkt sowie einer Vielzahl anderer pathophysiologischer Bedingungen assoziiert. Die potentielle Bedeutung von ApoJ umfasst eine Funktion als extrazelluläres Chaperon, Komplementinhibitor, NF-kB-Inhibitor sowie eine Beteiligung an der Endozytose von nekrotischen Zellfragmenten. Unter Bedingungen, die zu einer massiven Akkumulation von absterbenden Zellen führen, ist eine vermehrte Expression von ApoJ auf die überlebenden Nachbarzellen in den betroffenen Geweben beschränkt. Die molekularen Mechanismen, die dieser gesteigerten ApoJ-Genexpression zugrunde liegen, sind jedoch unbekannt. Untersuchungen unserer Arbeitsgruppe konnten zeigen, dass eine Inkubation mit nekrotischem Zellmaterial in vitro eine Akkumulation von ApoJ-mRNA in Fibroblasten der Zelllinie Rat1 induziert, was darauf hindeutet, dass unter pathophysiologischen Bedingungen von nekrotischen Zellen exponierte bzw. freigesetzte Faktoren zu einer gesteigerten ApoJ-Genexpression in umliegenden vitalen Zellen beitragen können. Die im Rahmen der vorliegenden Arbeit durchgeführten Untersuchungen zeigen eine Korrelation zwischen der Expression von Toll-like Rezeptoren (TLRs) in Fibroblasten (Rat1), glatten Gefäßmuskelzellen (CRL2018) sowie embryonalen Dottersackzellen (10A) und einer durch nekrotische Zellen induzierten ApoJ-mRNA-Expression in diesen Zelllinien. Es wird angenommen, dass TLRs neben pathogenassoziierten Strukturen (PAMPs) auch durch körpereigene Agonisten wie Hitzeschockproteine und Nukleinsäuren aktiviert werden. In weiterführenden Experimenten stellte sich unter anderem heraus, dass neben nekrotischen Zellen auch der TLR3-spezifische Agonist Poly(I:C), eine synthetische doppelsträngige RNA, ausschließlich in den beiden TLR3-exprimierenden Zelllinien CRL2018 und Rat1, nicht jedoch in TLR3-defizienten 10A-Zellen, die ApoJ-mRNA-Expression induziert. Darüber hinaus führt auch die Inkubation mit eukaryotischer RNA (Gesamt-RNA, t-RNA) zu einer Akkumulation von ApoJ-mRNA in CRL2018-Zellen. Die Ergebnisse dieser Arbeit zeigen erstmals, dass die Expression von ApoJ-mRNA durch extrazelluläre Ribonukleinsäuren in TLR3-abhängiger Weise induziert wird, was darauf hindeutet, dass in verletzten Geweben aus post-apoptotischen oder nekrotischen Zellen freigesetzte Ribonukleinsäuren zu einer vermehrten ApoJ-Genexpression in vitalen Nachbarzellen beitragen.
Resumo:
Alzheimer's disease (AD) is a fatal neurodegenerative condition characterized clinically by progressive memory loss and irreversible cognitive deterioration. It has been shown that there is a progressive degeneration of the brain cholinergic neurons which leads to the appearance of cognitive symptoms of the disease. The aim of this work was the formulation of multifunctional nanocarriers for nasal administration of tacrine-HCl (THA). This route has many advantages; in particular is possible to convey the drug directly to the Central Nervous System, through the olfactory bulb. In particular, were prepared Albumin nanoparticles carrying beta cyclodextrin and two different beta cyclodextrin derivatives (hydroxypropyl beta cyclodextrin and sulphobutylether beta cyclodextrin), and Multifunctional liposomes, prepared using traditional excipients (cholesterol and phosphatidylcholine), partly enriched with α-tocopherol (Toc) and/or polyunsaturated fatty acids (eicosapentaenoic acid and docosahexaenoic acid) (Ω3). Both nanosystems were characterized in terms of size, Zeta potential and encapsulation efficiency. Were also evaluated their functional properties such as mucoadhesion and permeability, using an ex-vivo assay based on nasal sheep mucosa. On Liposomes were also assessed drug neuronal uptake, cell toxicity, antioxidant and, cytoprotective activity in the human neuronal cell line SH-SY5Y and finally tocopherol trans-membrane diffusion. Both the nanocarriers produced presented excellent properties and a high potential as new systems for CNS-delivery of anti-Alzheimer drugs via the nasal route.
Resumo:
Oxidativer Stress ist seit über 25 Jahren als ein Charakteristikum vieler pathologischer Prozesse bekannt. Helmut Sies beschrieb bereits in den 1980er Jahren oxidativen Stress als Störung in der prooxidativ – antioxidativen Balance zugunsten der prooxidativen Seite, wodurch es potentiell zu Schäden in verschiedenen Geweben kommt. Oxidativer Stress tritt sowohl bei neurodegenerativen Erkrankungen wie Morbus Alzheimer, Morbus Parkinson und zerebraler Ischämie, bei peripheren Erkrankungen wie Arteriosklerose, als auch beim Alterungsprozess per se auf und wird als Ursache oder zumindest als ein krankheitsfördernder Faktor diskutiert. Die in in vitro-Experimenten als vielversprechend antioxidativ getesteten Substanzen (meist phenolhaltig) ergaben in mehreren klinischen Studien keinen signifikanten Vorteil. Um die Ursachen dieser Ergebnisse näher zu analysieren, wurde in der vorliegenden Arbeit auf Basis des cytoprotektiven Phenothiazins, einem aromatischen trizyklischen Amin, der Einfluss von verschiedenen Substituenten im Hinblick auf Lipophilie, Radikalstabilisierung und Löslichkeit des Moleküls chemisch vorhergesagt. Anhand dieser in silicio Struktur-Wirkungs-Beziehung wurden anschließend neue Modellsubstanzen synthetisiert, welche sich systematisch in den drei zuvor genannten Parametern unterschieden. Dies wurde durch Substitution von unterschiedlich langen Fettsäureketten, von löslichkeitsbeeinflussenden funktionellen Gruppen, oder durch Anellierung zusätzlicher aromatischer Ringe erreicht. In den folgenden Versuchen zu antioxidativer Kapazität, zellulärem Überleben, Lipidperoxidation und Proteinoxidation zeigte sich, dass mit gesteigerter Stabilität der korrespondierenden Radikale und mit wachsender Lipophilie die antioxidativ cytoprotektive Aktivität der neuen Derivate bis zu einer gewissen Grenze (logP ≈ 7) signifikant zunahm; über diesen Wert hinaus sank die Effektivität wieder ab. Benzanellierte Phenothiazine entwickelten mit EC50-Werten von ungefähr 8-10 nM die höchste mittlere effektive Wirkkonzentration in oxidativ geschädigten, klonalen hippocampalen Neuronen (HT-22 Zellen). Dies entspricht einer etwa 20-fachen Verbesserung gegenüber α-Tocopherol, welches bisher als bestes natürliches lipophiles Antioxidans angesehen wurde. Im Vergleich zu Phenothiazin erreichen die neuen Antioxidantien immerhin eine höhere Effektivität um den Faktor 4. Folglich sind es sowohl Aspekte der Löslichkeit und der Distribution, welche die Potenz der gegenwärtigen Antioxidantien limitieren als auch Aspekte der Radikalstabilisierung, die Einfluss auf die primäre Wirksamkeit nehmen. Dieses Wissen sollte beim zukünftigen Design neuer, antioxidativ potenter Moleküle im Hinblick auf ihren langfristigen Einsatz bei neurodegenerativen Erkrankungen von Nutzen sein.
Resumo:
Der Ginkgo biloba-Extrakt EGb 761 besteht aus einer Reihe pharmakologisch wirksamer Substanzen, welche gut beschriebene Wirkungen auf verschiedene potentiell zytoprotektive Signalwege ausüben und u.a. antioxidative Wirksamkeit haben. Folglich wurde EGb 761 bisher als eine natürliche Behandlung bei neurodegenerativen Erkrankungen mit zellulärem oxidativen Stress angewendet, einschließlich der Alzheimer-Krankheit (AD). Aufgrund von vielen gemeinsamen Merkmalen zwischen der AD und der Huntington-Krankheit (HD) wurde vermutet, dass EGb 761 eventuell auch positive Wirksamkeit bei der HD aufweisen könnte. rnDie Neuropathologie der HD wird durch pathologische Verlängerung an Glutamin-Wiederholungen im Huntingtin-Protein (polyQ-Protein) verursacht, wodurch es zu Fehlfaltungen im Protein kommt und hierdurch der proteasomale Abbau aberranter Proteine erschwert wird. Somit sollten in der vorliegenden Arbeit die EGb 761-Wirkungen auf die Proteasom-Aktivität und die Proteinaggregation in zellulären Modellen der HD untersucht werden. rnWie die ersten Untersuchungen in nativen HEK293-Zellen ergaben, bewirkte die Behandlung der Zellen mit EGb 761 eine Steigerung der basalen Proteasom-Aktivität sowie des proteasomalen Proteinabbaus und erhöhte die Transkription proteasomaler Gene. Hieraus ergaben sich Untersuchungen in Zellen mit Expressionen pathologischer Varianten von polyQ-Proteinen als zelluläre Modelle der HD. Hierbei konnte festgestellt werden, dass die Expression aberranter polyQ-Proteine eine verminderte zelluläre Proteasom-Aktivität bewirkte. Interessanterweise verursachte EGb 761 eine Abmilderung der pathologisch-induzierten verminderten Proteasom-Aktivität, in dem die EGb 761-Behandlung der Zellen zu einer erhöhten Proteasom-Aktivität, einem verbesserten proteasomalen Proteinabbau, sowie zu einer erhöhten Transkription proteasomaler Gene führte. Da diese EGb 761-Effekte unabhängig von der Expression aberranter polyQ-Proteine waren, demonstrierten diese Ergebnisse eine allgemeine EGb 761-Wirkungen auf die Proteasom-Aktivität. Anhand dieser Ergebnisse sollten anschließend weitere Untersuchungen mit zellulären Modellen der HD die genau Wirkung von EGb 761 auf die Degradation von abnormal verlängerten polyQ-Proteinen sowie auf die Bildung von polyQ-Aggregaten klären. rnHier konnte gezeigt werden, dass die Expression aberranter polyQ-Proteinen zu einer Akkumulation von SDS-resistenten bzw. SDS-unlöslichen, aggregierten polyQ-Proteinen führte, sowie die Bildung von sichtbaren polyQ-Aggregaten in Zellen bewirkte. Hierbei verursachte eine EGb 761-Behandlung der Zellen eine signifikante Verminderung im Gehalt an SDS-resistenten polyQ-Proteinen sowie eine Reduzierung von Aggregat-tragenden Zellen. Zudem konnte gezeigt werden, dass eine pharmakologische Inhibition des Proteasoms in EGb 761-behandelten Zellen, den Gehalt an SDS-unlöslichen polyQ-Proteinaggregate wieder erhöhte und somit den Effekt von EGb 761 aufhob. Folglich zeigten diese Ergebnisse, dass die EGb 761-induzierte Reduzierung der polyQ-Proteinaggregate durch einen effizienteren proteasomalen Abbau von fehlgefalteten, aberranten polyQ-Proteinen bewirkte wurde. rnAufbauend auf diesen Ergebnissen wurde eine experimentell-therapeutische Anwendung von EGb 761 in Modellen der HD in vitro und in vivo überprüft und hierzu primäre humane Fibroblasten sowie transgene C. elegans Würmer mit Expressionen aberranter polyQ-Proteine untersucht. Interessanterweise konnte in vitro und in vivo gezeigt werden, dass die EGb 761-Behandlung auch hier eine Reduzierung von SDS-unlöslichen polyQ-Proteinen bewirkte und zudem eine Reduzierung des pathologisch erhöhten Gehalts an Polyubiquitin-Proteinen bewirkte. Folglich wurde auch hier vermutet, dass EGb 761 einen verbesserten proteasomalen Abbau von polyQ-Proteinen induzierte und dies eine Verminderung der polyQ-Proteinaggregate verursachte. Darüber hinaus führte die EGb 761-Behandlung von seneszenten Fibroblasten zur Reduzierung von altersabhängig erhöhten Mengen von polyQ-Aggregaten, wodurch ein therapeutischer Effekt auf den proteasomalen Abbau der polyQ-Proteine verdeutlicht wurde. Zusätzlich konnte in polyQ-transgenen C. elegans demonstriert werden, dass eine EGb 761-Behandlung die Abmilderung eines typischen pathologischen Phänotyps bewirkte, indem eine polyQ-induzierte verminderte Motilität der Nematoden verbessert wurde und hierdurch eine positive EGb 761-Wirkung auf die Pathologie der HD in vivo dargestellt wurde. rnZusammenfassend konnten in dieser Arbeit neue Wirkungen von EGb 761 in der HD demonstriert werden. Hierbei wurde gezeigt, dass EGb 761 die Aggregation von pathogenen aberranten polyQ-Proteinen in vitro und in vivo reduziert, indem eine effizientere Degradation von polyQ-Proteinen erfolgt. Somit könnte diese Wirkungen von EGb 761 eine potentiell therapeutische Anwendung in der HD und ähnliche neurodegenerativen Erkrankungen darstellen.
Resumo:
The transcription factor PU.1 is a master regulator of myeloid differentiation and function. On the other hand, only scarce information is available on PU.1-regulated genes involved in cell survival. We now identified the glycolytic enzyme hexokinase 3 (HK3), a gene with cytoprotective functions, as transcriptional target of PU.1. Interestingly, HK3 expression is highly associated with the myeloid lineage and was significantly decreased in acute myeloid leukemia patients compared with normal granulocytes. Moreover, HK3 expression was significantly lower in acute promyelocytic leukemia (APL) compared with non-APL patient samples. In line with the observations in primary APL patient samples, we observed significantly higher HK3 expression during neutrophil differentiation of APL cell lines. Moreover, knocking down PU.1 impaired HK3 induction during neutrophil differentiation. In vivo binding of PU.1 and PML-RARA to the HK3 promoter was found, and PML-RARA attenuated PU.1 activation of the HK3 promoter. Next, inhibiting HK3 in APL cell lines resulted in significantly reduced neutrophil differentiation and viability compared with control cells. Our findings strongly suggest that HK3 is: (1) directly activated by PU.1, (2) repressed by PML-RARA, and (3) functionally involved in neutrophil differentiation and cell viability of APL cells.
Resumo:
OBJECTIVE: To determine whether a specifically designed bispecific (Bcl-2/Bcl-xL) antisense oligonucleotide (ASO) induces apoptosis and enhances chemosensitivity in human prostate cancer LNCaP cells, as Bcl-2 and Bcl-xL are both anti-apoptotic genes associated with treatment resistance and tumour progression in many malignancies, including prostate cancer. MATERIALS AND METHODS: Inhibition of Bcl-2 and Bcl-xL expression by the bispecific ASO was evaluated using real-time reverse transcription-polymerase chain reaction and Western blotting, while growth inhibition and induction of apoptosis were analysed by a crystal violet assay, flow cytometry and Western blotting of apoptosis-relevant proteins. The effect of combined treatment with bispecific ASO and chemotherapy or small-interference RNA (siRNA) targeting the clusterin gene was also investigated. RESULTS: Bispecific ASO reduced Bcl-2 and Bcl-xL expression in LNCaP cells in a dose-dependent manner. There was cell growth inhibition, increases in the sub-G0-G1 fraction, and cleavage of caspase-3 and poly(ADP-Ribose) polymerase proteins in LNCaP cells after bispecific ASO treatment. Interestingly, Bcl-2/Bcl-xL bispecific ASO treatment also resulted in the down-regulation of Mcl-1 and up-regulation of Bax. The sensitivity of LNCaP cells to mitoxantrone, docetaxel or paclitaxel was significantly increased, reducing the 50% inhibitory concentration by 45%, 80% or 90%, respectively. Furthermore, the apoptotic induction by Bcl-2/Bcl-xL bispecific ASO was synergistically enhanced by siRNA-mediated inhibition of clusterin, a cytoprotective chaperone that interacts with and inhibits activated Bax. CONCLUSIONS: These findings support the concept of the targeted suppression of Bcl-2 anti-apoptotic family members using multitarget inhibition strategies for prostate cancer, through the effective induction of apoptosis.
Resumo:
Complement is an essential part of the innate immune system and plays a crucial role in organ and islet transplantation. Its activation, triggered for example by ischemia/reperfusion (I/R), significantly influences graft survival, and blocking of complement by inhibitors has been shown to attenuate I/R injury. Another player of innate immunity are the dendritic cells (DC), which form an important link between innate and adaptive immunity. DC are relevant in the induction of an immune response as well as in the maintenance of tolerance. Modulation or inhibition of both components, complement and DC, may be crucial to improve the clinical outcome of solid organ as well as islet transplantation. Low molecular weight dextran sulfate (DXS), a well-known complement inhibitor, has been shown to prevent complement-mediated damage of the donor graft endothelium and is thus acting as an endothelial protectant. In this review we will discuss the evidence for this cytoprotective effect of DXS and also highlight recent data which show that DXS inhibits the maturation of human DC. Taken together the available data suggest that DXS may be a useful reagent to prevent the activation of innate immunity, both in solid organ and islet transplantation.
Resumo:
BACKGROUND/AIMS: Adipokines and hepatocellular apoptosis participate in the pathogenesis of nonalcoholic steatohepatitis (NASH). In a randomized trial ursodeoxycholic acid (UDCA) with vitamin E (VitE) improved serum aminotransferases and hepatic histology. The present work evaluates the effect of this combination on adipokines and hepatocellular apoptosis. METHODS: Circulating levels of adiponectin, resistin, leptin, interleukin (IL)-6, IL-8, retinol binding protein-4, monocyte chemoattractant protein-1 and tumour necrosis factor-alpha were measured by enzyme-linked immunoassays at the beginning and after 2 years of treatment with either UDCA+VitE, UDCA+placebo (P) or P+P. Apoptosis was assessed by immunohistochemistry for activated caspase-3 and circulating levels of apoptosis-associated cytokeratin 18 fragments (M30). RESULTS: Levels of adiponectin increased in patients treated with UDCA+VitE, whereas they decreased in the two other groups (P<0.04) and correlated with the improvement of liver steatosis (P<0.04). M30 levels worsened in the P/P group and improved in the other two groups. They correlated with hepatocellular apoptosis (P<0.02) and steatosis (P<0.02) as well as negatively with adiponectin levels (P<0.04). CONCLUSIONS: UDCA+VitE improves not only aminotransferase levels and liver histology of patients with NASH, but also decreases hepatocellular apoptosis and restores circulating levels of adiponectin. These results suggest that the UDCA+VitE combination has metabolic effects in addition to its beneficial cytoprotective properties.
Resumo:
MET, also known as hepatocyte growth factor receptor (HGFR), is a receptor tyrosine kinase with an important role, both in normal cellular function as well as in oncogenesis. In many cancer types, abnormal activation of MET is related to poor prognosis and various strategies to inhibit its function, including small molecule inhibitors, are currently in preclinical and clinical evaluation. Autophagy, a self-digesting recycling mechanism with cytoprotective functions, is induced by cellular stress. This process is also induced upon cytotoxic drug treatment of cancer cells and partially allows these cells to escape cell death. Thus, since autophagy protects different tumor cells from chemotherapy-induced cell death, current clinical trials aim at combining autophagy inhibitors with different cancer treatments. We found that in a gastric adenocarcinoma cell line GTL-16, where MET activity is deregulated due to receptor overexpression, two different MET inhibitors PHA665752 and EMD1214063 lead to cell death paralleled by the induction of autophagy. A combined treatment of MET inhibitors together with the autophagy inhibitor 3-MA or genetically impairing autophagy by knocking down the key autophagy gene ATG7 further decreased cell viability of gastric cancer cells. In general, we observed the induction of cytoprotective autophagy in MET expressing cells upon MET inhibition and a combination of MET and autophagy inhibition resulted in significantly decreased cell viability in gastric cancer cells.
Resumo:
Recent studies have shown that sulforaphane, a naturally occurring compound that is found in cruciferous vegetables, offers cellular protection in several models of brain injury. When administered following traumatic brain injury (TBI), sulforaphane has been demonstrated to attenuate blood-brain barrier permeability and reduce cerebral edema. These beneficial effects of sulforaphane have been shown to involve induction of a group of cytoprotective, Nrf2-driven genes, whose protein products include free radical scavenging and detoxifying enzymes. However, the influence of sulforaphane on post-injury cognitive deficits has not been examined. In this study, we examined if sulforaphane, when administered following cortical impact injury, can improve the performance of rats tested in hippocampal- and prefrontal cortex-dependent tasks. Our results indicate that sulforaphane treatment improves performance in the Morris water maze task (as indicated by decreased latencies during learning and platform localization during a probe trial) and reduces working memory dysfunction (tested using the delayed match-to-place task). These behavioral improvements were only observed when the treatment was initiated 1h, but not 6h, post-injury. These studies support the use of sulforaphane in the treatment of TBI, and extend the previously observed protective effects to include enhanced cognition.
Resumo:
Members of the WD-repeat protein interacting with phosphoinositides (WIPI) family are phosphatidylinositol 3-phosphate (PI3P) effectors that are essential for the formation of autophagosomes. Autophagosomes, unique double-membraned organelles, are characteristic for autophagy, a bulk degradation mechanism with cytoprotective and homeostatic function. Both, WIPI-1 and WIPI-2 are aberrantly expressed in several solid tumors, linking these genes to carcinogenesis. We now found that the expression of WIPI-1 was significantly reduced in a large cohort of 98 primary acute myeloid leukemia (AML) patient samples (complex karyotypes; t(8;21); t(15,17); inv(16)). In contrast, the expression of WIPI-2 was only reduced in acute promyelocytic leukemia (APL), a distinct subtype of AML (t(15,17)). As AML cells are blocked in their differentiation, we tested if the expression levels of WIPI-1 and WIPI-2 increase during all-trans retinoic acid (ATRA)-induced neutrophil differentiation of APL. According to the higher WIPI-1 expression in granulocytes compared with immature blast cells, WIPI-1 but not WIPI-2 expression was significantly induced during neutrophil differentiation of NB4 APL cells. Interestingly, the induction of WIPI-1 expression was dependent on the transcription factor PU.1, a master regulator of myelopoiesis, supporting our notion that WIPI-1 expression is reduced in AML patients lacking proper PU-1 activity. Further, knocking down WIPI-1 in NB4 cells markedly attenuated the autophagic flux and significantly reduced neutrophil differentiation. This result was also achieved by knocking down WIPI-2, suggesting that both WIPI-1 and WIPI-2 are functionally required and not redundant in mediating the PI3P signal at the onset of autophagy in NB4 cells. In line with these data, downregulation of PI3KC3 (hVPS34), which generates PI3P upstream of WIPIs, also inhibited neutrophil differentiation. In conclusion, we demonstrate that both WIPI-1 and WIPI-2 are required for the PI3P-dependent autophagic activity during neutrophil differentiation, and that PU.1-dependent WIPI-1 expression is significantly repressed in primary AML patient samples and that the induction of autophagic flux is associated with neutrophil differentiation of APL cells.
Resumo:
Autophagy, a fundamental cellular catabolic process, is involved in the development of numerous diseases including cancer. Autophagy seems to have an ambivalent impact on tumor development. While increasing evidence indicates a cytoprotective role for autophagy that can contribute to resistance against chemotherapy and even against the adverse, hypoxic environment of established tumors, relatively few publications focus on the role of autophagy in early tumorigenesis. However, the consensus is that autophagy is inhibitory for the genesis of tumors. To understand this apparent contradiction, more detailed information about the roles of the individual participants in autophagy is needed. This review will address this topic with respect to autophagy-related protein 5 (ATG5), which in several lines of investigation has been ascribed special significance in the autophagic pathway. Furthermore, it was recently shown that an ATG5 deficiency in melanocytes interferes with oncogene-induced senescence, thus promoting melanoma tumorigenesis. Similarly, an ATG5 deficiency resulted in tumors of the lung and liver in experimental mouse models. Taken together, these findings indicate that ATG5 and the autophagy to which it contributes are essential gatekeepers restricting early tumorigenesis in multiple tissues.
Resumo:
Bortezomib (VELCADE™, formerly known as PS-341) is a selective and potent inhibitor of the proteasome that was recently FDA-approved for the treatment of multiple myeloma. Despite its success in multiple myeloma and progression into clinical trials for other malignancies, bortezomib's exact mechanism of action remains undefined. The major objective of this study was to evaluate the anticancer activity of this drug using in vitro and in vivo pancreatic cancer models and determine whether bortezomib-induced apoptosis occurs via induction of endoplasmic reticular (ER) stress. The investigation revealed that bortezomib inhibited tumor cell proliferation via abrogation of cdk activity and induced apoptosis in pancreatic cancer cell lines. I hypothesized that bortezomib-induced apoptosis was triggered by a large accumulation ubiquitin-conjugated proteins that resulted in ER stress. My data demonstrated that bortezomib induced a unique type of ER stress in that it inhibited PKR-like ER kinase (PERK) and subsequent phosphorylation of eukaryotic initiation factor 2α (eif2α), a key event in translational suppression. The combined effects of proteasome inhibition and the failure to attenuate translation resulted in an accumulation of aggregated proteins (proteotoxicity), JNK activation, cytochrome c release, caspase-3 activation, and DNA fragmentation. Bortezomib also enhanced apoptosis induced by other agents that stimulated the unfolded protein response (UPR), demonstrating that translational suppression is a critical cytoprotective mechanism during ER stress. Tumor cells attempt to survive bortezomib-induced ER stress by sequestering aggregated proteins into large structures, termed aggresomes. Since histone deacetylase 6 (HDAC6) is essential for aggresome formation, tumor cells may be sensitized to bortezomib-induced apoptosis by blocking HDAC function. My results demonstrated that HDAC inhibitors disrupted aggresome formation and synergized with bortezomib to induce apoptosis in pancreatic cancer or multiple myeloma cells in vitro and in orthotopic pancreatic tumors in vivo. Taken together, my data establish a mechanistic link between bortezomib-induced aggresome formation, ER stress, and apoptosis and identify a novel therapeutic strategy for the treatment of pancreatic cancer and other hematologic and solid malignancies. ^
Resumo:
Cells govern their activities and modulate their interactions with the environment to achieve homeostasis. The heat shock response (HSR) is one of the most well studied fundamental cellular responses to environmental and physiological challenges, resulting in rapid synthesis of heat shock proteins (HSPs), which serve to protect cellular constituents from the deleterious effects of stress. In addition to its role in cytoprotection, the HSR also influences lifespan and is associated with a variety of human diseases including cancer, aging and neurodegenerative disorders. In most eukaryotes, the HSR is primarily mediated by the highly conserved transcription factor HSF1, which recognizes target hsp genes by binding to heat shock elements (HSEs) in their promoters. In recent years, significant efforts have been made to identify small molecules as potential pharmacological activators of HSF1 that could be used for therapeutic benefit in the treatment of human diseases relevant to protein conformation. However, the detailed mechanisms through which these molecules drive HSR activation remain unclear. In this work, I utilized the baker's yeast Saccharomyces cerevisiae as a model system to identify a group of thiol-reactive molecules including oxidants, transition metals and metalloids, and electrophiles, as potent activators of yeast Hsf1. Using an artificial HSE-lacZ reporter and the glucocorticoid receptor system (GR), these diverse thiol-reactive compounds are shown to activate Hsf1 and inhibit Hsp90 chaperone complex activity in a reciprocal, dose-dependent manner. To further understand whether cells sense these reactive compounds through accumulation of unfolded proteins, the proline analog azetidine-2-carboxylic acid (AZC) and protein cross-linker dithiobis(succinimidyl propionate) (DSP) were used to force misfolding of nascent polypeptides and existing cytosolic proteins, respectively. Both unfolding reagents display kinetic HSP induction profiles dissimilar to those generated by thiol-reactive compounds. Moreover, AZC treatment leads to significant cytotoxicity, which is not observed in the presence of the thiol-reactive compounds at the concentrations sufficient to induce Hsf1. Additionally, DSP treatment has little to no effect on Hsp90 functions. Together with the ultracentrifugation analysis of cell lysates that detected no insoluble protein aggregates, my data suggest that at concentrations sufficient to induce Hsf1, thiol-reactive compounds do not induce the HSR via a mechanism based on accumulation of unfolded cytosolic proteins. Another possibility is that thiol-reactive compounds may influence aspects of the protein quality control system such as the ubiquitin-proteasome system (UPS). To address this hypothesis, β-galactosidase reporter fusions were used as model substrates to demonstrate that thiol-reactive compounds do not inhibit ubiquitin activating enzymes (E1) or proteasome activity. Therefore, thiol-reactive compounds do not activate the HSR by inhibiting UPS-dependent protein degradation. I therefore hypothesized that these molecules may directly inactivate protein chaperones, known as repressors of Hsf1. To address this possibility, a thiol-reactive biotin probe was used to demonstrate in vitro that the yeast cytosolic Hsp70 Ssa1, which partners with Hsp90 to repress Hsf1, is specifically modified. Strikingly, mutation of conserved cysteine residues in Ssa1 renders cells insensitive to Hsf1 activation by cadmium and celastrol but not by heat shock. Conversely, substitution with the sulfinic acid and steric bulk mimic aspartic acid led to constitutive activation of Hsf1. Cysteine 303, located in the nucleotide-binding/ATPase domain of Ssa1, was shown to be modified in vivo by a model organic electrophile using Click chemistry technology, verifying that Ssa1 is a direct target for thiol-reactive compounds through adduct formation. Consistently, cadmium pretreatment promoted cells thermotolerance, which is abolished in cells carrying SSA1 cysteine mutant alleles. Taken together, these findings demonstrate that Hsp70 acts as a sensor to induce the cytoprotective heat shock response in response to environmental or endogenously produced thiol-reactive molecules and can discriminate between two distinct environmental stressors.
Resumo:
The mechanisms underlying cellular response to proteasome inhibitors have not been clearly elucidated in solid tumor models. Evidence suggests that the ability of a cell to manage the amount of proteotoxic stress following proteasome inhibition dictates survival. In this study using the FDA-approved proteasome inhibitor bortezomib (Velcade®) in solid tumor cells, we demonstrated that perhaps the most critical response to proteasome inhibition is repression of global protein synthesis by phosphorylation of the eukaryotic initiation factor 2-α subunit (eIF2α). In a panel of 10 distinct human pancreatic cancer cells, we showed marked heterogeneity in the ability of cancer cells to induce eIF2α phosphorylation upon stress (eIF2α-P); lack of inducible eIF2α-P led to excessive accumulation of aggregated proteins, reactive oxygen species, and ultimately cell death. In addition, we examined complementary cytoprotective mechanisms involving the activation of the heat shock response (HSR), and found that induction of heat shock protein 70 kDa (Hsp72) protected against proteasome inhibitor-induced cell death in human bladder cancer cells. Finally, investigation of a novel histone deacetylase 6 (HDAC6)-selective inhibitor suggested that the cytoprotective role of the cytoplasmic histone deacetylase 6 (HDAC6) in response to proteasome inhibition may have been previously overestimated.