954 resultados para tumor systemic effects
Resumo:
Il medulloblastoma (MB) è il tumore più comune del sistema nervoso centrale. Attualmente si riesce a curare solo il 50-60% dei pazienti che tuttavia presentano gravi effetti collaterali dovuti alle cure molto aggressive. Una recente classificazione molecolare ha ridistribuito le varianti più comuni di MB in quattro distinti gruppi. In particolare, il gruppo SHH e D sono caratterizzati da un’ alta espressione di MYCN ed associati a prognosi sfavorevole. MYCN è coinvolto nella regolazione della proliferazione cellulare, differenziazione, apoptosi, angiogenesi e metastasi. L’obiettivo di questo lavoro è la valutazione dell’attività antitumorale di oligonucleotidi diretti contro MYCN, sia in vitro su diverse linee cellulari di MB che in vivo in modelli murini xenograft ortotopici di MB. I risultati hanno dimostrato un’ottima inibizione della crescita in linee cellulari di MB, accompagnata da una riduzione della trascrizione genica e dei livelli proteici di MYCN. Inoltre, sono stati confermati tramite RT-PCR alcuni dei geni trovati significativamente variati nell’esperimento di microarray , dopo il trattamento. Molto interessanti sono stati geni quali BIRC5, che risulta down regolato mentre il gene p21 risulta up regolato in tutte le linee cellulari di MB utilizzate. Inoltre, sono stati generati modelli murini di MB utilizzando cellule precedentemente trasfettate per esprimere il gene della luciferasi e valutarne così la crescita tumorale tramite imaging bioluminescente in vivo (BLI), al fine di poter testare l’attività antitumorale data dagli oligonucleotidi. I risultati hanno dimostrato una buona risposta al trattamento come rilevato dalla tecnica di BLI. I dati preliminari prodotti, dimostrano che MYCN potrebbe esser un buon target per la terapia del MB nei casi in cui è overespresso. In particolare, una sua inibizione potrebbe presentare effetti indesiderati moderati in quanto è poco espresso dopo la nascita.
Resumo:
Cross Reacting Material 197(CRM197) is a Diphteria toxin non toxic mutant that had shown anti-tumor activity in mice and humans. CRM197 is utilized as a specific inhibitor of heparin-binding epidermal growth factor (HB-EGF), that competes for the epidermal growth factor receptor (EGFR), overexpressed in colorectal cancer and implicated in its progression. We evaluated the effects of CRM197 on HT-29 human colon cancer cell line behaviour and, for CRM197 recognized ability to inhibit HB-EGF, its possible effects on EGFR activation. In particular, while HT-29 does not show any reduction of viability after CRM197 treatment, or changes in cell cycle distribution, in EGFR localization or activation, they show a change in gene expression profile analyzed by microarray. This is the first study where the CRM197 treatment on HT-29 show the alteration of a specific and selected number of genes.
Analyse der Beteiligung von Toll-like-Rezeptoren an der DC-Aktivierung nach Parvovirus-H-1-Infektion
Resumo:
Ziel dieser Dissertation war es die funktionelle Rolle der Toll-like Rezeptoren (TLRs) und ihrer Signalwege bei der Aktivierung von dendritischen Zellen (DC) durch Parvovirus H-1- rn(H-1PV) induzierte Tumorzelllysate (TCL) zu untersuchen. rnDas angeborene Immunsystem bekämpft die Bildung und das Wachstum von Tumoren, insbesondere durch Interaktion von Effektor-Immunzellen mit Tumorzellen. Die Aktivierung dieser Immunreaktionen in der Antitumortherapie ist wünschenswert, aber in vielen Situationen nicht zufriedenstellend, da sie durch klassische systemische Therapie allein nicht immer erreicht werden kann. Die therapeutische Anwendung von onkolytischen Viren bei Patienten mit malignen Erkrankungen (Virotherapie) ist ein vielversprechendes Gebiet der Forschung. Die onkosuppressive und immunstimulierende Wirkung von H-1PV auf humane Tumor- und Immunzellen spricht für eine Verwendung in der Krebstherapie. Ein Aktivierung des Immunsystems durch H-1PV konnte bereits in unserer Arbeitsgruppe gezeigt werden.rnIn dieser Arbeit wurden wichtige Aspekte bezüglich der Aktivierung von Toll-like Rezeptoren bei einer H-1PV Infektion untersucht. Zunächst wurde die Rolle von TLRs nach der H-1PV Infektion untersucht. Humane embryonale Nierenzellen (HEK293) wurden stabil mit humanen TLRs transfiziert, um die Rolle spezifischer TLRs während der Aktivierung des Immunsystems zu untersuchen. TLR3 und TLR9 wurden durch eine H-1PV Infektion, die mit der NFκB-Translokation in den Zellkern korreliert, aktiviert. Mit Hilfe eines Reporterplasmides (pNiFty-Luc), wurde durch erhöhte Expression eines NFκB-induzierbaren Reportergens die NFκB-Aktivität im Anschluss an eine H-1PV Infektion nachgewiesen. Zudem wurde die immunologische Wirkung von H-1PV-induzierten Tumorzelllysaten (TCL) auf die humane antitumor-gerichtete Immunantworten analysiert. Ein humanes ex vivo-Modell, bestehend aus einer HLA-A2-positiven humanen Melanom-Zelllinie (SK29Mel) wurde verwendet, um Immunreaktionen mit entsprechenden HLA-restringierten humanen DCs zu untersuchen. DCs die mit H-1PV-infizierten SK29Mel Zellen koinkubiert wurden, zeigten eine erhöhte TLR3- und TLR9-Expression. Diese Daten deuten darauf hin, dass H-1PV-induzierte TCLs humane DCs stimulieren und dies zumindest teilweise durch TLR-abhängige Signalwege geschieht. Demnach wird eine DC-Reifung durch Kokultur mit H-1PV-induzierten TCLs über den TLR-Signalweg erreicht und führte u.a. zu einer NFκB-abhängigen Aktivierung des adaptiven Immunsystems. Die onkolytischen Virotherapie mit H-1PV erhöht so durch unterschiedliche Auswirkungen auf DCs die Immunreaktion und verstärkt die Anti-Tumor-Immunität. Diese Ergebnisse zeigen einen neuen potenziellen Ansatz für den Einsatz onkolytischer Viren für TLR-zielgerichtete Therapieoptionen und stellen eine ideale Möglichkeit zur Erweiterung der Krebsbehandlung dar.rn
Resumo:
Inflammatory bowel diseases are associated with increased risk of developing colitis-associated colorectal cancer (CAC). Epidemiological data show that the consumption of ω-3 polyunsaturated fatty acids (ω-3 PUFAs) decreases the risk of sporadic colorectal cancer (CRC). Importantly, recent data have shown that eicosapentaenoic acid-free fatty acid (EPA-FFA) reduces polyps formation and growth in models of familial adenomatous polyposis. However, the effects of dietary EPA-FFA are unknown in CAC. We tested the effectiveness of substituting EPA-FFA, for other dietary fats, in preventing inflammation and cancer in the AOM-DSS model of CAC. The AOM-DSS protocols were designed to evaluate the effect of EPA-FFA on both initiation and promotion of carcinogenesis. We found that EPA-FFA diet strongly decreased tumor multiplicity, incidence and maximum tumor size in the promotion and initiation arms. Moreover EPA-FFA, in particular in the initiation arm, led to reduced cell proliferation and nuclear β-catenin expression, whilst it increased apoptosis. In both arms, EPA-FFA treatment led to increased membrane switch from ω-6 to ω-3 PUFAs and a concomitant reduction in PGE2 production. We observed no significant changes in intestinal inflammation between EPA-FFA treated arms and AOM-DSS controls. Importantly, we found that EPA-FFA treatment restored the loss of Notch signaling found in the AOM-DSS control, resulted in the enrichment of Lactobacillus species in the gut microbiota and led to tumor suppressor miR34-a induction. In conclusion, our data suggest that EPA-FFA is an effective chemopreventive agent in CAC.
Resumo:
Die akute myeloische Leukämie (AML) ist eine heterogene Erkrankung der hämatopoetischen Vorläuferzelle, die durch unkontrollierte Vermehrung und ein reduziertes Differenzierungsverhalten gekennzeichnet ist. Aufgrund von Therapieresistenzen und häufig vorkommenden Rückfällen ist die AML mit einer schlechten Langzeitprognose verbunden. Neue Studienergebnisse zeigen, dass leukämische Zellen einer hierarchischen Ordnung unterliegen, an deren Spitze die leukämische Stammzelle (LSC) steht, welche den Tumor speist und ähnliche Charakteristika besitzt wie die hämatopoetische Stammzelle. Die LSC nutzt den Kontakt zu Zellen der hämatopoetischen Nische des Knochenmarks, um die erste Therapie zu überdauern und Resistenzen zu erwerben. Neue Therapieansätze versuchen diese Interaktion zwischen leukämischen Zellen und supportiv wirkenden Stromazellen anzugreifen. rnrnIn dieser Arbeit sollte die Bedeutung des CXC-Motiv Chemokinrezeptors Typ 4 (CXCR4) und des Connective Tissue Growth Factors (CTGF) innerhalb der AML-Stroma-Interaktion untersucht werden. CXCR4, der in vivo dafür sorgt, dass AML-Zellen in der Nische gehalten und geschützt werden, wurde durch den neuwertigen humanen CXCR4-spezifischen Antikörper BMS-936564/MDX-1338 in AML-Zelllinien und Patientenzellen in Zellkulturversuchen blockiert. Dies induzierte Apoptose sowie Differenzierung und führte in Kokulturversuchen zu einer Aufhebung des Stroma-vermittelten Schutzes gegenüber der Chemotherapie. Für diese Effekte musste teilweise ein sekundärer Antikörper verwendet werden, der die CXCR4-Moleküle miteinander kreuzvernetzt.rnDie Auswertung eines quantitativen Real time PCR (qPCR)-Arrays ergab, dass CTGF in der AML-Zelllinie Molm-14 nach Kontakt zu Stromazellen hochreguliert wird. Diese Hochregulation konnte in insgesamt drei AML-Zelllinien sowie in drei Patientenproben in qPCR- und Western Blot-Versuchen bestätigt werden. Weitere Untersuchungen zeigten, dass diese Hochregulation (i) unabhängig von der Stromazelllinie ist, (ii) den direkten Kontakt zum Stroma benötigt und (iii) auch unter hypoxischen Bedingungen, wie sie innerhalb des Knochenmarks vorherrschen, stattfindet. Der durch Zell-Zell- oder Zell-Matrix-Kontakt gesteuerte Hippo-Signalweg konnte aus folgenden Gründen als möglicher upstream-Regulationsmechanismus identifiziert werden: (i) Dessen zentraler Transkriptions-Kofaktor TAZ wurde in kokultivierten Molm-14-Zellen stabilisiert, (ii) der shRNA-gesteuerte Knockdown von TAZ führte zu einer reduzierten CTGF-Hochregulation, (iii) CTGF wurde in Abhängigkeit von der Zelldichte reguliert, (iv) Cysteine-rich angiogenic inducer 61 (Cyr61), ein weiteres Zielgen von TAZ, wurde in kokultivierten AML-Zellen ebenfalls verstärkt exprimiert. Der Knockdown von CTGF führte in vitro zu einer partiellen Aufhebung der Stroma-vermittelten Resistenz und die Blockierung von CTGF durch den Antikörper FG-3019 wirkte im AML-Mausmodell lebensverlängernd. rn rnDie Rolle von CTGF in der AML ist bisher nicht untersucht. Die vorliegenden Ergebnisse zeigen, dass CTGF ein interessantes Therapieziel in der AML darstellt. Es bedarf weiterer Untersuchungen, um die Bedeutung von CTGF in der Tumor-Stroma-Interaktion näher zu charakterisieren und nachgeschaltete Signalwege zu identifizieren.
Resumo:
Patienten, die an Osteosarkom leiden werden derzeit mit intravenös applizierten krebstherapeutischen Mitteln nach Tumorresektion behandelt, was oftmals mit schweren Nebenwirkungen und einem verzögerten Knochenheilungsprozess einhergeht. Darüber hinaus treten vermehrt Rezidive aufgrund von verbleibenden neoplastischen Zellen an der Tumorresektionsstelle auf. Erfolgreiche Knochenregeneration und die Kontrolle von den im Gewebe verbleibenden Krebszellen stellt eine Herausforderung für das Tissue Engineering nach Knochenverlust durch Tumorentfernung dar. In dieser Hinsicht scheint der Einsatz von Hydroxyapatit als Knochenersatzmaterial in Kombination mit Cyclodextrin als Medikamententräger, vielversprechend. Chemotherapeutika können an Biomaterial gebunden und direkt am Tumorbett über einen längeren Zeitraum freigesetzt werden, um verbliebene neoplastische Zellen zu eliminieren. Lokal applizierte Chemotherapie hat diverse Vorteile, einschließlich der direkten zytotoxischen Auswirkung auf lokale Zellen, sowie die Reduzierung schwerer Nebenwirkungen. Diese Studie wurde durchgeführt, um die Funktionsfähigkeit eines solchen Arzneimittelabgabesystems zu bewerten und um Strategien im Bereich des Tissue Engineerings zu entwickeln, die den Knochenheilungsprozess und im speziellen die Vaskularisierung fördern sollen. Die Ergebnisse zeigen, dass nicht nur Krebszellen von der chemotherapeutischen Behandlung betroffen sind. Primäre Endothelzellen wie zum Beispiel HUVEC zeigten eine hohe Sensibilität Cisplatin und Doxorubicin gegenüber. Beide Medikamente lösten in HUVEC ein tumor-unterdrückendes Signal durch die Hochregulation von p53 und p21 aus. Zudem scheint Hypoxie einen krebstherapeutischen Einfluss zu haben, da die Behandlung sensitiver HUVEC mit Hypoxie die Zellen vor Zytotoxizität schützte. Der chemo-protektive Effekt schien deutlich weniger auf Krebszelllinien zu wirken. Diese Resultate könnten eine mögliche chemotherapeutische Strategie darstellen, um den Effekt eines zielgerichteten Medikamenteneinsatzes auf Krebszellen zu verbessern unter gleichzeitiger Schonung gesunder Zellen. Eine erfolgreiche Integration eines Systems, das Arzneimittel abgibt, kombiniert mit einem Biomaterial zur Stabilisierung und Regeneration, könnte gesunden Endothelzellen die Möglichkeit bieten zu proliferieren und Blutgefäße zu bilden, während verbleibende Krebszellen eliminiert werden. Da der Prozess der Knochengeweberemodellierung mit einer starken Beeinträchtigung der Lebensqualität des Patienten einhergeht, ist die Beschleunigung des postoperativen Heilungsprozesses eines der Ziele des Tissue Engineerings. Die Bildung von Blutgefäßen ist unabdingbar für eine erfolgreiche Integration eines Knochentransplantats in das Gewebe. Daher ist ein umfangreich ausgebildetes Blutgefäßsystem für einen verbesserten Heilungsprozess während der klinischen Anwendung wünschenswert. Frühere Experimente zeigen, dass sich die Anwendung von Ko-Kulturen aus humanen primären Osteoblasten (pOB) und humanen outgrowth endothelial cells (OEC) im Hinblick auf die Bildung stabiler gefäßähnlicher Strukturen in vitro, die auch effizient in das mikrovaskuläre System in vivo integriert werden konnten, als erfolgreich erweisen. Dieser Ansatz könnte genutzt werden, um prä-vaskularisierte Konstrukte herzustellen, die den Knochenheilungsprozess nach der Implantation fördern. Zusätzlich repräsentiert das Ko-Kultursystem ein exzellentes in vitro Model, um Faktoren, welche stark in den Prozess der Knochenheilung und Angiogenese eingebunden sind, zu identifizieren und zu analysieren. Es ist bekannt, dass Makrophagen eine maßgebliche Rolle in der inflammatorisch-induzierten Angiogenese spielen. In diesem Zusammenhang hebt diese Studie den positiven Einfluss THP-1 abgeleiteter Makrophagen in Ko-Kultur mit pOB und OEC hervor. Die Ergebnisse zeigten, dass die Anwendung von Makrophagen als inflammatorischer Stimulus im bereits etablierten Ko-Kultursystem zu einer pro-angiogenen Aktivierung der OEC führte, was in einer signifikant erhöhten Bildung blutgefäßähnlicher Strukturen in vitro resultierte. Außerdem zeigte die Analyse von Faktoren, die in der durch Entzündung hervorgerufenen Angiogenese eine wichtige Rolle spielen, eine deutliche Hochregulation von VEGF, inflammatorischer Zytokine und Adhäsionsmoleküle, die letztlich zu einer verstärkten Vaskularisierung beitragen. Diese Resultate werden dem Einfluss von Makrophagen zugeschrieben und könnten zukünftig im Tissue Engineering eingesetzt werden, um den Heilungsprozess zu beschleunigen und damit die klinische Situation von Patienten zu verbessern. Darüber hinaus könnte die Kombination der auf Ko-Kulturen basierenden Ansätze für das Knochen Tissue Engineering mit einem biomaterial-basierenden Arzneimittelabgabesystem zum klinischen Einsatz kommen, der die Eliminierung verbliebener Krebszellen mit der Förderung der Knochenregeneration verbindet.
Resumo:
Glioblastoma multiforme (GBM) is the most common and most aggressive astrocytic tumor of the central nervous system (CNS) in adults. The standard treatment consisting of surgery, followed by a combinatorial radio- and chemotherapy, is only palliative and prolongs patient median survival to 12 to 15 months. The tumor subpopulation of stem cell-like glioma-initiating cells (GICs) shows resistance against radiation as well as chemotherapy, and has been suggested to be responsible for relapses of more aggressive tumors after therapy. The efficacy of immunotherapies, which exploit the immune system to specifically recognize and eliminate malignant cells, is limited due to strong immunosuppressive activities of the GICs and the generation of a specialized protective microenvironment. The molecular mechanisms underlying the therapy resistance of GICs are largely unknown. rnThe first aim of this study was to identify immune evasion mechanisms in GICs triggered by radiation. A model was used in which patient-derived GICs were treated in vitro with fractionated ionizing radiation (2.5 Gy in 7 consecutive passages) to select for a more radio-resistant phenotype. In the model cell line 1080, this selection process resulted in increased proliferative but diminished migratory capacities in comparison to untreated control GICs. Furthermore, radio-selected GICs downregulated various proteins involved in antigen processing and presentation, resulting in decreased expression of MHC class I molecules on the cellular surface and diminished recognition potential by cytotoxic CD8+ T cells. Thus, sub-lethal fractionated radiation can promote immune evasion and hamper the success of adjuvant immunotherapy. Among several immune-associated proteins, interferon-induced transmembrane protein 3 (IFITM3) was found to be upregulated in radio-selected GICs. While high expression of IFITM3 was associated with a worse overall survival of GBM patients (TCGA database) and increased proliferation and migration of differentiated glioma cell lines, a strong contribution of IFITM3 to proliferation in vitro as well as tumor growth and invasiveness in a xenograft model could not be observed. rnMultiple sclerosis (MS) is the most common autoimmune disease of the CNS in young adults of the Western World, which leads to progressive disability in genetically susceptible individuals, possibly triggered by environmental factors. It is assumed that self-reactive, myelin-specific T helper cell 1 (Th1) and Th17 cells, which have escaped the control mechanisms of the immune system, are critical in the pathogenesis of the human disease and its animal model experimental autoimmune encephalomyelitis (EAE). It was observed that in vitro differentiated interleukin 17 (IL-17) producing Th17 cells co-expressed the Th1-phenotypic cytokine Interferon-gamma (IFN-γ) in combination with the two respective lineage-associated transcription factors RORγt and T-bet after re-isolation from the CNS of diseased mice. Pathogenic molecular mechanisms that render a CD4+ T cell encephalitogenic have scarcely been investigated up to date. rnIn the second part of the thesis, whole transcriptional changes occurring in in vitro differentiated Th17 cells in the course of EAE were analyzed. Evaluation of signaling networks revealed an overrepresentation of genes involved in communication between the innate and adaptive immune system and metabolic alterations including cholesterol biosynthesis. The transcription factors Cebpa, Fos, Klf4, Nfatc1 and Spi1, associated with thymocyte development and naïve T cells were upregulated in encephalitogenic CNS-isolated CD4+ T cells, proposing a contribution to T cell plasticity. Correlation of the murine T-cell gene expression dataset to putative MS risk genes, which were selected based on their proximity (± 500 kb; ensembl database, release 75) to the MS risk single nucleotide polymorphisms (SNPs) proposed by the most recent multiple sclerosis GWAS in 2011, revealed that 67.3% of the MS risk genes were differentially expressed in EAE. Expression patterns of Bach2, Il2ra, Irf8, Mertk, Odf3b, Plek, Rgs1, Slc30a7, and Thada were confirmed in independent experiments, suggesting a contribution to T cell pathogenicity. Functional analysis of Nfatc1 revealed that Nfatc1-deficient CD4+ T cells were restrained in their ability to induce clinical signs of EAE. Nfatc1-deficiency allowed proper T cell activation, but diminished their potential to fully differentiate into Th17 cells and to express high amounts of lineage cytokines. As the inducible Nfatc1/αA transcript is distinct from the other family members, it could represent an interesting target for therapeutic intervention in MS.rn
Resumo:
Resistance of cancer cells towards chemotherapy is the major cause of therapy failure. Hence, the evaluation of cellular defense mechanisms is essential in the establishment of new chemotherapeutics. In this study, classical intrinsic and acquired as well as new resistance mechanisms relevant in the cellular response to the novel vacuolar H+-ATPase inhibitor archazolid B were investigated. Archazolid B, originally produced by the myxobacterium Archangium gephyra, displayed cytotoxicity in the low nanomolar range on a panel of cancer cell lines. The drug showed enhanced cytotoxic activity against nearly all cancerous cells compared to their non-cancerous pendants. With regards to ABC transporters, archazolid B was identified as a moderate substrate of ABCB1 (P-glycoprotein) and a weak substrate of ABCG2 (BCRP), whereas hypersensitivity was observed in ABCB5-expressing cells. The cytotoxic effect of archazolid B was shown to be independent of the cellular p53 status. However, cells expressing constitutively active EGFR displayed significantly increased resistance. Acquired drug resistance was studied by establishing an archazolid B-resistant MCF-7 cell line. Experiments showed that this secondary resistance was not conferred by aberrant expression or DNA mutations of the gene encoding vacuolar H+-ATPase subunit c, the direct target of archazolid B. Instead, a slight increase of ABCB1 and a significant overexpression of EGFR as well as reduced proliferation may contribute to acquired archazolid B resistance. For identification of new resistance strategies upon archazolid B treatment, omics data from bladder cancer and glioblastoma cells were analyzed, revealing drastic disturbances in cholesterol homeostasis, affecting cholesterol biosynthesis, uptake and transport. As shown by filipin staining, archazolid B led to accumulation of free cholesterol in lysosomes, which triggered sterol responses, mediated by SREBP-2 and LXR, including up-regulation of HMGCR, the key enzyme of cholesterol biosynthesis. Furthermore, inhibition of LDL uptake as well as impaired LDLR surface expression were observed, indicating newly synthesized cholesterol to be the main source of cholesterol in archazolid B-treated cells. This was proven by the fact that under archazolid B treatment, total free cholesterol levels as well as cell survival were significantly reduced by inhibiting HMGCR with fluvastatin. The combination of archazolid B with statins may therefore be an attractive strategy to circumvent cholesterol-mediated cell survival and in turn potentiate the promising anticancer effects of archazolid B.
Resumo:
PURPOSE The aim of this study was to evaluate the antibiotic treatment of postoperative endophthalmitis with combined systemic meropenem and linezolid. METHODS A retrospective analysis of endophthalmitis treated with systemic meropenem and linezolid compared to conventional systemic antibiotics by evaluation of outcome and adverse effects was carried out. RESULTS 26 patients with unilateral postoperative endophthalmitis with a systemic combination regimen of meropenem (2 g TID, mean duration of treatment 5.5 days) and linezolid (600 mg BID, mean duration of treatment 8.9 days) (group 1, mean follow-up time 140 days) were included in this study and compared to 45 postoperative endophthalmitis patients treated with conventional systemic antibiotics (group 2; mean follow-up time 320 days). In group 1, 69.2 % of eyes additionally received intravitreal amikacin and vancomycin (vs. 24.4 % in group 2; p < 0.001), in 92.3 % pars plana vitrectomy was performed (vs. 68.9 % in group 2, p = 0.047). Mean best corrected visual acuity improved from a baseline of 1.76 logMar for group 1 and 1.83 logMar for group 2 to 0.91 logMar (p = 0.0001) and 0.90 logMar (p < 0.0001), respectively, at the end of the follow-up, revealing no significant differences between the two groups at any time point (p > 0.05). Ocular complications were observed in 34.6 % of eyes in group 1 (vs. 37.8 % in group 2; p > 0.05). Adverse drug effects occurred significantly more frequently in group 1 (26.9 % vs. 4.4 % p = 0.02). CONCLUSION In spite of the reported better penetration through the blood-ocular barrier and the broader antibacterial spectrum of meropenem and linezolid, no benefit in outcome was found in the present study. In contrast, adverse effects and costs of the combination regimen were significantly higher.
Resumo:
Lung recruitment maneuvers (RMs), used to reopen atelectatic lung units and to improve oxygenation during mechanical ventilation, may result in hemodynamic impairment. We hypothesize that pulmonary arterial hypertension aggravates the consequences of RMs in the splanchnic circulation. Twelve anesthetized pigs underwent laparotomy and prolonged postoperative ventilation. Systemic, regional, and organ blood flows were monitored. After 6 h (= baseline), a recruitment maneuver was performed with sustained inflation of the lungs. Thereafter, the pigs were randomly assigned to group C (control, n = 6) or group E with endotoxin-induced pulmonary arterial hypertension (n = 6). Endotoxemia resulted in a normotensive and hyperdynamic state and a deterioration of the oxygenation index by 33%. The RM was then repeated in both groups. Pulmonary artery pressure increased during lipopolysaccharide infusion from 17 ± 2 mmHg (mean ± SD) to 31 ± 10 mmHg and remained unchanged in controls (P < 0.05). During endotoxemia, RM decreased aortic pulse pressure from 37 ± 14 mmHg to 27 ± 13 mmHg (mean ± SD, P = 0.024). The blood flows of the renal artery, hepatic artery, celiac trunk, superior mesenteric artery, and portal vein decreased to 71% ± 21%, 69% ± 20%, 76% ± 16%, 79% ± 18%, and 81% ± 12%, respectively, of baseline flows before RM (P < 0.05 all). Organ perfusion of kidney cortex, kidney medulla, liver, and jejunal mucosa in group E decreased to 65% ± 19%, 77% ± 13%, 66% ± 26%, and 71% ± 12%, respectively, of baseline flows (P < 0.05 all). The corresponding recovery to at least 90% of baseline regional blood flow and organ perfusion lasted 1 to 5 min. Importantly, the decreases in regional blood flows and organ perfusion and the time to recovery of these flows did not differ from the controls. In conclusion, lipopolysaccharide-induced pulmonary arterial hypertension does not aggravate the RM-induced significant but short-lasting decreases in systemic, regional, and organ blood flows.
Resumo:
The incidence and prevalence of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) have increased in the past 20 years. GEP-NETs are heterogeneous tumors, in terms of clinical and biological features, that originate from the pancreas or the intestinal tract. Some GEP-NETs grow very slowly, some grow rapidly and do not cause symptoms, and others cause hormone hypersecretion and associated symptoms. Most GEP-NETs overexpress receptors for somatostatins. Somatostatins inhibit the release of many hormones and other secretory proteins; their effects are mediated by G protein-coupled receptors that are expressed in a tissue-specific manner. Most GEP-NETs overexpress the somatostatin receptor SSTR2; somatostatin analogues are the best therapeutic option for functional neuroendocrine tumors because they reduce hormone-related symptoms and also have antitumor effects. Long-acting formulations of somatostatin analogues stabilize tumor growth over long periods. The development of radioactive analogues for imaging and peptide receptor radiotherapy has improved the management of GEP-NETs. Peptide receptor radiotherapy has significant antitumor effects, increasing overall survival times of patients with tumors that express a high density of SSTRs, particularly SSTR2 and SSTR5. The multi-receptor somatostatin analogue SOM230 (pasireotide) and chimeric molecules that bind SSTR2 and the dopamine receptor D2 are also being developed to treat patients with GEP-NETs. Combinations of radioactive labeled and unlabeled somatostatin analogues and therapeutics that inhibit other signaling pathways, such as mammalian target of rapamycin (mTOR) and vascular endothelial growth factor, might be the most effective therapeutics for GEP-NETs.
Resumo:
Isolated GH deficiency type II (IGHD II) is the autosomal dominant form of GHD. In the majority of the cases, this disorder is due to specific GH-1 gene mutations that lead to mRNA missplicing and subsequent loss of exon 3 sequences. When misspliced RNA is translated, it produces a toxic 17.5-kDa GH (Delta3GH) isoform that reduces the accumulation and secretion of wild-type-GH. At present, patients suffering from this type of disease are treated with daily injections of recombinant human GH in order to maintain normal growth. However, this type of replacement therapy does not prevent toxic effects of the Delta3GH mutant on the pituitary gland, which can eventually lead to other hormonal deficiencies. We developed a strategy involving Delta3GH isoform knockdown mediated by expression of a microRNA-30-adapted short hairpin RNA (shRNA) specifically targeting the Delta3GH mRNA of human (shRNAmir-Delta3). Rat pituitary tumor GC cells expressing Delta3GH upon doxycycline induction were transduced with shRNAmir-Delta3 lentiviral vectors, which significantly reduced Delta3GH protein levels and improved human wild-type-GH secretion in comparison with a shRNAmir targeting a scrambled sequence. No toxicity due to shRNAmir expression could be observed in cell proliferation assays. Confocal microscopy strongly suggested that shRNAmir-Delta3 enabled the recovery of GH granule storage and secretory capacity. These viral vectors have shown their ability to stably integrate, express shRNAmir, and rescue IGHD II phenotype in rat pituitary tumor GC cells, a methodology that opens new perspectives for the development of gene therapy to treat IGHD patients.
Resumo:
We investigated here the effects of S2T1-6OTD, a novel telomestatin derivative that is synthesized to target G-quadruplex-forming DNA sequences, on a representative panel of human medulloblastoma (MB) and atypical teratoid/rhabdoid (AT/RT) childhood brain cancer cell lines. S2T1-6OTD proved to be a potent c-Myc inhibitor through its high-affinity physical interaction with the G-quadruplex structure in the c-Myc promoter. Treatment with S2T1-6OTD reduced the mRNA and protein expressions of c-Myc and hTERT, which is transcriptionally regulated by c-Myc, and decreased the activities of both genes. In remarkable contrast to control cells, short-term (72-hour) treatment with S2T1-6OTD resulted in a dose- and time-dependent antiproliferative effect in all MB and AT/RT brain tumor cell lines tested (IC(50), 0.25-0.39 micromol/L). Under conditions where inhibition of both proliferation and c-Myc activity was observed, S2T1-6OTD treatment decreased the protein expression of the cell cycle activator cyclin-dependent kinase 2 and induced cell cycle arrest. Long-term treatment (5 weeks) with nontoxic concentrations of S2T1-6OTD resulted in a time-dependent (mainly c-Myc-dependent) telomere shortening. This was accompanied by cell growth arrest starting on day 28 followed by cell senescence and induction of apoptosis on day 35 in all of the five cell lines investigated. On in vivo animal testing, S2T1-6OTD may well represent a novel therapeutic strategy for childhood brain tumors.
Resumo:
Use of norepinephrine to increase blood pressure in septic animals has been associated with increased efficiency of hepatic mitochondrial respiration. The aim of this study was to evaluate whether the same effect could be reproduced in isolated hepatic mitochondria after prolonged in vivo exposure to faecal peritonitis. Eighteen pigs were randomized to 27 h of faecal peritonitis and to a control condition (n = 9 each group). At the end, hepatic mitochondria were isolated and incubated for one hour with either norepinephrine or placebo, with and without pretreatment with the specific receptor antagonists prazosin and yohimbine. Mitochondrial state 3 and state 4 respiration were measured for respiratory chain complexes I and II, and state 3 for complex IV using high-resolution respirometry, and respiratory control ratios were calculated. Additionally, skeletal muscle mitochondrial respiration was evaluated after incubation with norepinephrine and dobutamine with and without the respective antagonists (atenolol, propranolol and phentolamine for dobutamine). Faecal peritonitis was characterized by decreasing blood pressure and stroke volume, and maintained systemic oxygen consumption. Neither faecal peritonitis nor any of the drugs or drug combinations had measurable effects on hepatic or skeletal muscle mitochondrial respiration. Norepinephrine did not improve the efficiency of complex I- and complex II-dependent isolated hepatic mitochondrial respiration [respiratory control ratio (RCR) complex I: 5.6 ± 5.3 (placebo) vs. 5.4 ± 4.6 (norepinephrine) in controls and 2.7 ± 2.1 (placebo) vs. 2.9 ± 1.5 (norepinephrine) in septic animals; RCR complex II: 3.5 ± 2.0 (placebo) vs. 3.5 ± 1.8 (norepinephrine) in controls; 2.3 ± 1.6 (placebo) vs. 2.2 ± 1.1 (norepinephrine) in septic animals]. Prolonged faecal peritonitis did not affect either hepatic or skeletal muscle mitochondrial respiration. Subsequent incubation of isolated mitochondria with norepinephrine and dobutamine did not significantly influence their respiration.
Resumo:
Sorafenib targets the Raf/mitogen-activated protein kinase, VEGF, and platelet-derived growth factor pathways and prolongs survival patients in advanced hepatocellular carcinoma (HCC). Everolimus inhibits the mammalian target of rapamycin, a kinase overactive in HCC. To investigate whether the antitumor effects of these agents are additive, we compared a combined and sequential treatment regimen of everolimus and sorafenib with monotherapy. After hepatic implantation of Morris Hepatoma (MH) cells, rats were randomly allocated to everolimus (5 mg/kg, 2×/week), sorafenib (7.5 mg/kg/d), combined everolimus and sorafenib, sequential sorafenib (2 weeks) then everolimus (3 weeks), or control groups. MRI quantified tumor volumes. Erk1/2, 4E-BP1, and their phosphorylated forms were quantified by immunoblotting. Angiogenesis was assessed in vitro by aortic ring and tube formation assays, and in vivo with Vegf-a mRNA and vascular casts. After 35 days, tumor volumes were reduced by 60%, 85%, and 55%, relative to controls, in everolimus, the combination, and sequential groups, respectively (P < 0.01). Survival was longest in the combination group (P < 0.001). Phosphorylation of 4E-BP1 and Erk1/2 decreased after everolimus and sorafenib, respectively. Angiogenesis decreased after all treatments (P < 0.05), although sorafenib increased Vegf-a mRNA in liver tumors. Vessel sprouting was abundant in control tumors, lower after sorafenib, and absent after the combination. Intussusceptive angiogenic transluminal pillars failed to coalesce after the combination. Combined treatment with everolimus and sorafenib exerts a stronger antitumoral effect on MH tumors than monotherapy. Everolimus retains antitumoral properties when administered sequentially after sorafenib. This supports the clinical use of everolimus in HCC, both in combination with sorafenib or after sorafenib.