881 resultados para Akt,AMPK,Glut4
Resumo:
Introduction: Apoptotic cell death of cardiomyocytes is involved in several cardiovascular diseases including ischemia, hypertrophy and heart failure, thus representing a potential therapeutic target. Apoptosis of cardiac cells can be induced experimentally by several stimuli including hypoxia, serum withdrawal or combination of both. Several lines of research suggest that neurohormonal mechanisms play a central role in the progression of heart failure. In particular, excessive activation of the sympathetic nervous system or the renin-angiotensin-aldosterone system is known to have deleterious effects on the heart. Recent studies report that norepinephrine (NE), the primary transmitter of sympathetic nervous system, and aldosterone (ALD), which is actively produced in failing human heart, are able to induce apoptosis of rat cardiomyocytes. Polyamines are biogenic amines involved in many cellular processes, including apoptosis. Actually it appears that these molecules can act as promoting, modulating or protective agents in apoptosis depending on apoptotic stimulus and cellular model. We have studied the involvement of polyamines in the apoptosis of cardiac cells induced in a model of simulated ischemia and following treatment with NE or ALD. Methods: H9c2 cardiomyoblasts were exposed to a condition of simulated ischemia, consisting of hypoxia plus serum deprivation. Cardiomyocyte cultures were prepared from 1-3 day-old neonatal Wistar rat hearts. Polyamine depletion was obtained by culturing the cells in the presence of α-difluoromethylornithine (DFMO). Polyamines were separated and quantified in acidic cellular extracts by HPLC after derivatization with dansyl chloride. Caspase activity was measured by the cleavage of the fluorogenic peptide substrate. Ornithine decarboxylase (ODC) activity was measured by estimation of the release of 14C-CO2 from 14C-ornithine. DNA fragmentation was visualized by the method of terminal transferase-mediated dUTP nick end-labeling (TUNEL), and DNA laddering on agarose gel electophoresis. Cytochrome c was detected by immunoflorescent staining. Activation of signal transduction pathways was investigated by western blotting. Results: The results indicate that simulated ischemia, NE and ALD cause an early induction of the activity of ornithine decarboxylase (ODC), the first enzyme in polyamine biosynthesis, followed by a later increase of caspase activity, a family of proteases that execute the death program and induce cell death. This effect was prevented in the presence of DFMO, an irreversible inhibitor of ODC, thus suggesting that polyamines are involved in the execution of the death program activated by these stimuli. In H9c2 cells DFMO inhibits several molecular events related to apoptosis that follow simulated ischemia, such as the release of cytochrome c from mitochondria, down-regulation of Bcl-xL, and DNA fragmentation. The anti-apoptotic protein survivin is down-regulated after ALD or NE treatement and polyamine depletion obtained by DFMO partially opposes survivin decrease. Moreover, a study of key signal transduction pathways governing cell death and survival, revealed an involvement of AMP activated protein kinase (AMPK) and AKT kinase, in the modulation by polyamines of the response of cardiomyocytes to NE. In fact polyamine depleted cells show an altered pattern of AMPK and AKT activation that may contrast apoptosis and appears to result from a differential effect on the specific phosphatases that dephosphorylate and switch off these signaling proteins. Conclusions: These results indicate that polyamines are involved in the execution of the death program activated in cardiac cells by heart failure-related stimuli, like ischemia, ALD and NE, and suggest that their apoptosis facilitating action is mediated by a network of specific phosphatases and kinases.
Resumo:
Eine wesentliche Voraussetzung für die maligne Transformation von Zellen ist die Inaktivierung des programmierten Zelltodes (Apoptose). Die dabei erworbenen Defekte der Apoptose-Signalwege führen häufig zu Resistenzen gegenüber Radio- und Chemotherapien. Immuntherapeutische Ansätze haben zum Ziel, solche resistenten Tumorzellen spezifisch zu entfernen. Resistenzen gegenüber Immuntherapien können wiederum in einer gestörten Immunerkennung der Tumorzellen oder deren Resistenz gegenüber Immuneffektormechanismen begründet sein. Ziel der vorliegenden Arbeit war, zu überprüfen, ob durch Proteinkinase B (PKB)/Akt Immunresistenz vermittelt werden kann. Hierbei zeigte sich, dass die Aktivierung des PKB/Akt-Signalweges in Tumorzellen einen deutlichen Schutz gegenüber verschiedenen Apoptosestimuli in vitro vermittelt. Die konditionale Aktivierung von PKB/Akt hemmte sowohl die pharmakologisch, als auch die durch ZTL induzierte Apoptose-Signalkaskade über eine posttranskriptionelle Stabilisierung des anti-apoptotischen Proteins MCL-1. Diese Beobachtung konnte auch in einem murinen Tumorimmuntherapiemodell in vivo bestätigt werden. Unstimulierte Splenozyten von C57Bl/6-Mäusen wurden adoptiv in NOD/SCID-Mäuse mit etablierten, PKB/Akt-exprimierenden, murinen Fibrosarkomen transferiert. Die konditionale Aktivierung von PKB/Akt inhibierte den tumorsuppressiven Effekt dieser transplantierten Splenozyten signifikant. Des Weiteren konnte gezeigt werden, dass die PKB/Akt-abhängige Immunresistenz auch in vivo durch anti-apoptotisches MCL-1 vermittelt wird. PKB/Akt-exprimierende Fibrosarkome mit supprimierter endogener MCL-1-Expression verloren ihre Resistenz gegenüber der durch adoptiven Splenozytentransfer vermittelten Tumorsuppression. Dies bestätigte endogenes MCL-1 als entscheidenden Faktor der PKB/Akt-vermittelten Immunresistenz. Ferner konnte gezeigt werden, dass eine Hemmung der PKB/Akt-induzierten Signaltransduktion auf der Ebene der nachgeschalteten Kinase mTOR etablierte Fibrosarkome gegenüber adoptiver Lymphozytentherapie sensitiviert. Der mTOR-Inhibitor Rapamycin verhinderte die PKB/Akt-induzierte Aufregulation von MCL-1 und die damit einhergehende Resistenzentwicklung in vivo. Zusammengefasst wurde erstmalig gezeigt, dass eine Deregulation des PKB/Akt-Signalweges Resistenz gegenüber immunologischer Tumorsuppression vermitteln kann. PKB/Akt stellt somit ein entscheidendes Zielmolekül für die Verbesserung von Krebsimmuntherapien dar.
Resumo:
Le cellule staminali/stromali mesenchimali umane (hMSC) sono attualmente applicate in diversi studi clinici e la loro efficacia è spesso legata alla loro capacità di raggiungere il sito d’interesse. Poco si sa sul loro comportamento migratorio e i meccanismi che ne sono alla base. Perciò, questo studio è stato progettato per comprendere il comportamento migratorio delle hMSC e il coinvolgimento di Akt, nota anche come proteina chinasi B. L’espressione e la fosforilazione della proteinchinasi Akt è stata studiata mediante Western blotting. Oltre al time-lapse in vivo imaging, il movimento cellulare è stato monitorato sia mediante saggi tridimensionali, con l’uso di transwell, che mediante saggi bidimensionali, attraverso la tecnica del wound healing. Le prove effettuate hanno rivelato che le hMSC hanno una buona capacità migratoria. E’ stato osservato che la proteinchinasi B/Akt ha elevati livelli basali di fosforilazione in queste cellule. Inoltre, la caratterizzazione delle principali proteine di regolazione ed effettrici, a monte e a valle di Akt, ha permesso di concludere che la cascata di reazioni della via di segnale anche nelle hMSC segue un andamento canonico. Specifici inibitori farmacologici sono stati utilizzati per determinare il potenziale meccanismo coinvolto nella migrazione cellulare e nell'invasione. L’inibizione della via PI3K/Akt determina una significativa riduzione della migrazione. L’utilizzo di inibitori farmacologici specifici per le singole isoforme di Akt ha permesso di discriminare il ruolo diverso di Akt1 e Akt2 nella migrazione delle hMSC. E’ stato infatti dimostrato che l'inattivazione di Akt2, ma non quella di Akt1, diminuisce significativamente la migrazione cellulare. Nel complesso i risultati ottenuti indicano che l'attivazione di Akt2 svolge un ruolo critico nella migrazione della hMSC; ulteriori studi sono necessari per approfondire la comprensione del fenomeno. La dimostrazione che l’isoforma Akt2 è necessaria per la chemiotassi diretta delle hMSC, rende questa chinasi un potenziale bersaglio farmacologico per modulare la loro migrazione.
Resumo:
Because of its aberrant activation, the PI3K/AKT/mTOR signaling pathway represents a pharmacological target in blast cells from patients with acute myelogenous leukemia (AML). Using Reverse Phase Protein Microarrays (RPMA), we have analyzed 20 phosphorylated epitopes of the PI3K/Akt/mTor signal pathway of peripheral blood and bone marrow specimens of 84 patients with newly diagnosed AML. Fresh blast cells were grown for 2 h, 4 h or 20 h untreated or treated with a panel of phase I or phase II Akt allosteric inhibitors, either alone or in combination with the mTOR kinase inhibitor Torin1 or the broad RTK inhibitor Sunitinib. By unsupervised hierarchical clustering a strong phosphorylation/activity of most of the sampled members of the PI3K/Akt/mTOR pathway was observed in 70% of samples from AML patients. Remarkably, however, we observed that inhibition of Akt phosphorylation, as well as of its substrates, was transient, and recovered or even increased far above basal level after 20 h in 60% samples. We demonstrated that inhibition of Akt induces FOXO-dependent insulin receptor expression and IRS-1 activation, attenuating the effect of drug treatment by reactivation of PI3K/Akt. Consistent with this model we found that combined inhibition of Akt and RTKs is much more effective than either alone, revealing the adaptive capabilities of signaling networks in blast cells and highliting the limations of these drugs if used as monotherapy.
Resumo:
Il trapianto allogenico di cellule staminali emopoietiche è spesso l’unica soluzione per la cura di diverse malattie ematologiche. La aGVHD è la complicanza più importante che si può avere a seguito del trapianto allogenico ed è causata dai linfociti T del donatore che riconoscono gli antigeni del ricevente presentati dalle APC. Eliminare o inattivare la APC del ricevente prima del trapianto potrebbe prevenire la aGVHD. Ad oggi non esistono farmaci specifici diretti contro le APC, sono però noti i meccanismi molecolari coinvolti nella sopravvivenza cellulare come la via di segnale di PI3K. In questo lavoro abbiamo testato l’attività di due farmaci, che colpiscono target molecolari della via di PI3K, la rapamicina e la perifosina, sul differenziamento dei monociti a differenti popolazioni di cellule dendritiche (DC), in vitro. La rapamicina riduceva il recupero cellulare delle DC derivate da monociti coltivate in presenza di IL-4 aumentando l’apoptosi, mentre i monociti coltivati in presenza di GM-CSF con o senza IFN-α risultavano resistenti alla rapamicina. Inoltre la rapamicina riduceva l’espressione della molecola costimolatoria CD86 e incrementava l’espressione della molecola CD1a solo nei monociti coltivati con GM-CSF e IL-4. Nelle DC derivate dai monociti in presenza di IL-4 la rapamicina bloccava la produzione di IL-12 e TNF-α e ne alterava la capacità allostimolatoria. La rapamicina non alterava la sopravvivenza e la funzione delle DC circolanti. Il trattamento con perifosina provocava un incremento di apoptosi nei monociti coltivati sia con GM-CSF che con GM-CSF e IL-4. La perifosina bloccava la produzione di TNF-α nelle DC derivate da monociti coltivati nelle diverse condizioni. Questi risultati dimostrano che l’azione della rapamicina è strettamente dipendente dalla presenza dell’IL-4 nel terreno di coltura, in vitro, rispetto alla perifosina e suggeriscono un possibile ruolo della perifosina nella prevenzione della GVHD prima del trapianto allogenico di cellule staminali.
Resumo:
Die AMPK ist ein ubiquitär exprimiertes, heterotrimeres Enzym, das bei Energiemangel das Überleben der Zelle sichert. Um diese Funktion ausüben zu können fungiert die AMPK als sogenannter „Energie-Sensor“, der durch steigende AMP Mengen aktiviert wird. In diesem Zustand werden ATP verbrauchende Reaktionen inhibiert und gleichzeitig ATP generierende Vorgänge induziert. Im vaskulären System konnte gezeigt werden, dass die endotheliale NOSynthase durch die AMPK aktiviert, die Angiogenese stimuliert, die Endothelzellapoptose und das Wachstum von Gefäßmuskelzellen inhibiert wird. All diese Prozesse sind fundamental in der Entwicklung von kardiovaskulären Krankheiten, was auf eine protektive Funktion der AMPK im vaskulären System hindeutet. In der vorliegenden Arbeit sollten die Effekte der in vivo Modulation der AMPK Aktivität auf Endothelfunktion, oxidativen Stress und Inflammation untersucht werden. Dazu wurden zwei unterschiedliche Mausmodelle genutzt: Einerseits wurde die AMPK Aktivität durch den pharmakologischen AMPK-Aktivator AICAR stimuliert und andererseits die vaskulär vorherrschende AMPK-Isoform durch knock out ausgeschaltet. Zur Induktion von oxidativem Stress wurde ein bereits charakterisiertes Angiotensin II-Modell angewandt. Zur Untersuchung gehörten neben den Superoxid-Messungen auch die Bestimmung der Stickstoffmonoxid-Mengen in Serum und Aortengewebe, die Relaxationsmessungen in isometrischen Tonusstudien sowie HPLC-basierte Assays. Es konnte gezeigt werden, dass durch die Aktivierung der AMPK mittels AICAR die Angiotensin II induzierte Endotheldysfunktion, der oxidative Stress und auch die vaskuläre Inflammation verbessert werden konnte. Weiterhin zeigte sich dass der knock out der vaskulären Isoform (α1) im Angiotensin II Modell eine signifikant verstärkte Endotheldysfunktion, oxidativen Stress und Inflammation nach sich zog. Anhand der erhobenen Daten konnte die NADPH-Oxidase als Hauptquelle des Angiotensin II induzierten oxidativen Stresses identifiziert werden, wobei sich diese Quelle als AMPK sensitiv erwies. Durch die Aktivierung konnte die Aktivität der NADPH-Oxidase verringert und durch die α1AMPK Defizienz signifikant erhöht werden. Auch die mitochondriale Superoxidproduktion konnte durch die Modulation der AMPK Aktivität beeinflusst werden. Die vaskuläre Inflammation, die anhand der Surrogaten VCAM-1, COX-2 und iNOS untersucht wurde, konnte durch Aktivierung der AMPK verringert werden, der knock out der α1AMPK führte so einer sehr starken Expressionssteigerung der induzierbaren NO-Synthase, was in einem starken Anstieg der NO-Produktion und somit der Peroxynitritbildung resultierte.Die dargestellten Daten deuten stark auf eine protektive Funktion der AMPK im vaskulären System hin und sollte als therapeutisches Ziel, nicht nur in Bezug auf diabetische Patienten, in Betracht gezogen werden.
Resumo:
Class I phosphatidylinositol 3-kinases (PI3Ks) are heterodimeric lipid kinases consisting of a regulatory subunit and one of four catalytic subunits (p110α, p110β, p110γ or p110δ). p110γ/p110δ PI3Ks are highly enriched in leukocytes. In general, PI3Ks regulate a variety of cellular processes including cell proliferation, survival and metabolism, by generating the second messenger phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P3). Their activity is tightly regulated by the phosphatase and tensin homolog (PTEN) lipid phosphatase. PI3Ks are widely implicated in human cancers, and in particular are upregulated in T-cell acute lymphoblastic leukemia (T-ALL), mainly due to loss of PTEN function. These observations lend compelling weight to the application of PI3K inhibitors in the therapy of T-ALL. At present different compounds which target single or multiple PI3K isoforms have entered clinical trials. In the present research, it has been analyzed the therapeutic potential of the pan-PI3K inhibitor BKM120, an orally bioavailable 2,6-dimorpholino pyrimidine derivative, which has entered clinical trials for solid tumors, on both T-ALL cell lines and patient samples. BKM120 treatment resulted in cell cycle arrest and apoptosis, being cytotoxic to a panel of T-ALL cell lines and patient T-lymphoblasts. Remarkably, BKM120 synergized with chemotherapeutic agents currently used for treating T-ALL patients. BKM120 efficacy was confirmed in in vivo studies to a subcutaneous xenotransplant model of human T-ALL. Because it is still unclear which agents among isoform-specific or pan inhibitors can achieve the greater efficacy, further analyses have been conducted to investigate the effects of PI3K inhibition, in order to elucidate the mechanisms responsible for the proliferative impairment of T-ALL. Overall, these results indicated that BKM120 may be an efficient treatment for T-ALLs that have aberrant up-regulation of the PI3K signaling pathway and strongly support clinical application of pan-class I PI3K rather than single-isoform inhibitors in T-ALL treatment.
Resumo:
Pneumococcal meningitis causes apoptosis of developing neurons in the dentate gyrus of the hippocampus. The death of these cells is accompanied with long-term learning and memory deficits in meningitis survivors. Here, we studied the role of the PI3K/Akt (protein kinase B) survival pathway in hippocampal apoptosis in a well-characterized infant rat model of pneumococcal meningitis. Meningitis was accompanied by a significant decrease of the PI3K product phosphatidylinositol 3,4,5-trisphosphate (PIP(3)) and of phosphorylated (i.e., activated) Akt in the hippocampus. At the cellular level, phosphorylated Akt was decreased in both the granular layer and the subgranular zone of the dentate gyrus, the region where the developing neurons undergo apoptosis. Protein levels and activity of PTEN, the major antagonist of PI3K, were unaltered by infection, suggesting that the observed decrease in PIP(3) and Akt phosphorylation is a result of decreased PI3K signaling. Treatment with the PTEN inhibitor bpV(pic) restored Akt activity and significantly attenuated hippocampal apoptosis. Co-treatment with the specific PI3K inhibitor LY294002 reversed the restoration of Akt activity and attenuation of hippocampal apoptosis, while it had no significant effect on these parameters on its own. These results indicate that the inhibitory effect of bpV(pic) on apoptosis was mediated by PI3K-dependent activation of Akt, strongly suggesting that bpV(pic) acted on PTEN. Treatment with bpV(pic) also partially inhibited the concentration of bacteria and cytokines in the CSF, but this effect was not reversed by LY294002, indicating that the effect of bpV(pic) on apoptosis was independent of its effect on CSF bacterial burden and cytokine levels. These results indicate that the PI3K/Akt pathway plays an important role in the death and survival of developing hippocampal neurons during the acute phase of pneumococcal meningitis.
Resumo:
Activators of 5'-AMP-activated protein kinase (AMPK) 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR), metformin, and exercise activate atypical protein kinase C (aPKC) and ERK and stimulate glucose transport in muscle by uncertain mechanisms. Here, in cultured L6 myotubes: AICAR- and metformin-induced activation of AMPK was required for activation of aPKC and ERK; aPKC activation involved and required phosphoinositide-dependent kinase 1 (PDK1) phosphorylation of Thr410-PKC-zeta; aPKC Thr410 phosphorylation and activation also required MEK1-dependent ERK; and glucose transport effects of AICAR and metformin were inhibited by expression of dominant-negative AMPK, kinase-inactive PDK1, MEK1 inhibitors, kinase-inactive PKC-zeta, and RNA interference (RNAi)-mediated knockdown of PKC-zeta. In mice, muscle-specific aPKC (PKC-lambda) depletion by conditional gene targeting impaired AICAR-stimulated glucose disposal and stimulatory effects of both AICAR and metformin on 2-deoxyglucose/glucose uptake in muscle in vivo and AICAR stimulation of 2-[(3)H]deoxyglucose uptake in isolated extensor digitorum longus muscle; however, AMPK activation was unimpaired. In marked contrast to AICAR and metformin, treadmill exercise-induced stimulation of 2-deoxyglucose/glucose uptake was not inhibited in aPKC-knockout mice. Finally, in intact rodents, AICAR and metformin activated aPKC in muscle, but not in liver, despite activating AMPK in both tissues. The findings demonstrate that in muscle AICAR and metformin activate aPKC via sequential activation of AMPK, ERK, and PDK1 and the AMPK/ERK/PDK1/aPKC pathway is required for metformin- and AICAR-stimulated increases in glucose transport. On the other hand, although aPKC is activated by treadmill exercise, this activation is not required for exercise-induced increases in glucose transport, and therefore may be a redundant mechanism.
Resumo:
The quassinoid analogue NBT-272 has been reported to inhibit MYC, thus warranting a further effort 7to better understand its preclinical properties in models of embryonal tumors (ET), a family of childhood malignancies sharing relevant biological and genetic features such as deregulated expression of MYC oncogenes. In our study, NBT-272 displayed a strong antiproliferative activity in vitro that resulted from the combination of diverse biological effects, ranging from G(1)/S arrest of the cell cycle to apoptosis and autophagy. The compound prevented the full activation of both eukaryotic translation initiation factor 4E (eIF4E) and its binding protein 4EBP-1, regulating cap-dependent protein translation. Interestingly, all responses induced by NBT-272 in ET could be attributed to interference with 2 main proproliferative signaling pathways, that is, the AKT and the MEK/extracellular signal-regulated kinase pathways. These findings also suggested that the depleting effect of NBT-272 on MYC protein expression occurred via indirect mechanisms, rather than selective inhibition. Finally, the ability of NBT-272 to arrest tumor growth in a xenograft model of neuroblastoma plays a role in the strong antitumor activity of this compound, both in vitro and in vivo, with its potential to target cell-survival pathways that are relevant for the development and progression of ET.
Resumo:
The secretin receptor (SR), a G protein-coupled receptor, mediates the effects of the gastrointestinal hormone secretin on digestion and water homeostasis. Recently, high SR expression has been observed in pancreatic ductal adenocarcinomas, cholangiocellular carcinomas, gastrinomas, and bronchopulmonary carcinoid tumors. Receptor overexpression associates with enhanced secretin-mediated signaling, but whether this molecule plays an independent role in tumorigenesis is currently unknown. We recently discovered that pheochromocytomas developing in rats affected by the MENX (multiple endocrine neoplasia-like) syndrome express at very high-level Sctr, encoding SR. We here report that SR are also highly abundant on the membranes of rat adrenal and extraadrenal pheochromocytoma, starting from early stages of tumor development, and are functional. PC12 cells, the best characterized in vitro pheochromocytoma model, also express Sctr at high level. Thus, we used them as model to study the role of SR in neoplastic transformation. Small interfering RNA-mediated knockdown of Sctr decreases PC12 cells proliferation and increases p27 levels. The proproliferative effect of SR in PC12 cells is mediated, in part, by the phosphatidylinositol 3 kinase (PI3K)/serine-threonine protein kinase (AKT) pathway. Transfection of Sctr in Y1 adrenocortical carcinoma cells, expressing low endogenous levels of Sctr, stimulates cell proliferation also, in part, via the PI3K/AKT signaling cascade. Because of the link between SR and PI3K/AKT signaling, tumor cells expressing high levels of the receptor (MENX-associated primary pheochromocytoma and NCI-H727 human bronchopulmonary carcinoid cells) respond well and in a SR-dependent manner to PI3K inhibitors, such as NVP-BEZ235. The association between SR levels and response to PI3K inhibition might open new avenues for the treatment of tumors overexpressing this receptor.