130 resultados para INDUCED ENHANCEMENT
Resumo:
Multisensory interactions have been documented within low-level, even primary, cortices and at early post-stimulus latencies. These effects are in turn linked to behavioral and perceptual modulations. In humans, visual cortex excitability, as measured by transcranial magnetic stimulation (TMS) induced phosphenes, can be reliably enhanced by the co-presentation of sounds. This enhancement occurs at pre-perceptual stages and is selective for different types of complex sounds. However, the source(s) of auditory inputs effectuating these excitability changes in primary visual cortex remain disputed. The present study sought to determine if direct connections between low-level auditory cortices and primary visual cortex are mediating these kinds of effects by varying the pitch and bandwidth of the sounds co-presented with single-pulse TMS over the occipital pole. Our results from 10 healthy young adults indicate that both the central frequency and bandwidth of a sound independently affect the excitability of visual cortex during processing stages as early as 30 msec post-sound onset. Such findings are consistent with direct connections mediating early-latency, low-level multisensory interactions within visual cortices.
Resumo:
Lactate release by astrocytes is postulated to be of importance for neuroenergetics but its regulation is poorly understood. Basigin, a chaperone protein for specific monocarboxylate transporters (MCTs), represents a putatively important regulatory element for lactate fluxes. Indeed, basigin knockdown by RNA interference in primary cultures of astrocytes partially reduced both proton-driven lactate influx and efflux. But more strikingly, enhancement of lactate efflux induced by glutamate was prevented while the effect of sodium azide was significantly reduced by treatment of cultured astrocytes with anti-basigin small interfering RNA. Enhancement of glucose utilization was unaffected under the same conditions. Basal lactate uptake and release were significantly reduced by MCT1 knockdown, even more so than with basigin knockdown, whereas glutamate-driven or sodium azide-induced enhancement of lactate release was not inhibited by either MCT1, 2, or 4 small interfering RNAs. In conclusion, MCT1 plays a pivotal role in the control of basal proton-driven lactate flux in astrocytes while basigin is only partly involved, most likely via its interaction with MCT1. In contrast, basigin appears to critically regulate the enhancement of lactate release caused by glutamate (or sodium azide) but via an effect on another unidentified transporter at least present in astrocytes in vitro.
Resumo:
Summary of the thesis Glucose has been considered the major, if not the exclusive, energy substrate for the brain. But under certain conditions other substrates, namely monocarboxylates (lactate, pyruvate, and ketone bodies), can contribute significantly to satisfy brain energy demands. These monocarboxylates need to be transported across the blood brain barrier as well as out of astrocytes into the extracellular space and taken up into neurons. It has been shown that monocarboxylates are transported by a family of proton-linked transporters called monocarboxylate transporters (MCTs). In the central nervous system, MCT2 is the predominant neuronal form and little is known about the regulation of its expression. The neurotransmitter noradrenaline (NA) was shown previously to enhance the expression of MCT2 in cultured cortical neurons via a translational mechanism. Here, we demonstrate that two other substances, namely, insulin and IGF-1 enhance MCT2 protein expression in cultured mouse cortical neurons in a time- and concentrationdependent manner without affecting MCT2 mRNA levels. This result confirmed that MCT2 protein expression is translationally regulated and extend the observation to different types of neuroactive substances. Then we sought to determine by which signaling pathway(s) NA, insulin and IGF-1 can induce MCT2 protein expression. First, we observed by Western blot that all three substances cause activation of the MAP kinase ERK as well as the kinase Akt via their phosphorylation. Moreover, the mTOR/S6K pathway which is known to play an important role in translation initiation regulation was also strongly stimulated by all three substances. Second, we sought to determine the implication of these signaling pathways on the NA-, insulin- and IGF-1-induced enhancement of MCT2 protein expression and used specific inhibitors of these signaling pathways. We observed that the Pia kinase and mTOR inhibitors LY294002 and rapamycin respectively, strongly prevent the enhancement. of MCT2 expression caused by either NA, insulin ar IGF-1. In contrast, the MEK inhibitor PD98059 and the p38 MAP kinase inhibitor SB202190 had only a slight effect on the enhancement of MCT2 expression in all three cases. These results suggest that NA, insulin and IGF-1 regulate MCT2 protein expression by a common mechanism most likely involving the Akt/PKB pathway and translational activation via mTOR. In conclusion, considering the roles of NA, insulin and IGF-1 in synaptic plasticity, the tight translational regulation of MCT2 expression by these substances may represent a common mechanism through which supply of potentiated synapses with nonglucose energy substrates can be adapted to the level of activity. Résumé du travail de thèse Le glucose représente le substrat énergétique majeur pour le cerveau. Cependant, dans certaines conditions physiologiques ou pathologiques, le cerveau a la capacité d'utiliser des substrats énergétiques appartenant à la classe des monocarboxylates (lactate, pyruvate et corps cétoniques) afin de satisfaire ses besoins énergétiques. Ces monocarboxylates doivent être transportés à travers la barrière hématoencéphalique mais aussi hors des astrocytes vers l'espace extracellulaire puis re-captés par les neurones. Leur transport est assuré par une famille de transporteurs spécifiques, protons-dépendants, appelés transporteurs aux monocarboxylates (MCTs). Dans le système nerveux central, les neurones expriment principalement l'isoforme MCT2 mais peu d'informations sont disponibles concernant la régulation de son expression. Il a été montré que le neurotransmetteur noradrénaline (NA) augmente l'expression de MCT2 dans les cultures de neurones corticaux de souris par le biais d'un mécanisme de régulation traductionnel. La présente étude nous a permis de démontrer que deux autres substances, l'insuline et 17GF-1, induisent une augmentation de la protéine MCT2 dans ces mêmes cultures selon un décours temporel et une gamme de concentrations particulière. Etonnamment, aucun changement n'a été observé concernant les niveaux d'ARNm de MCT2. Ce résultat .confirme que la protéine MCT2 est régulée de manière traductionnelle et révèle que différentes substances neuro-actives peuvent réguler l'expression de MCT2. Compte tenu de ces observations, nous avons voulu déterminer par quelle(s) voie(s) de signalisation la NA, l'insuline et l'IGF-1 exercent leur effet sur l'expression de MCT2. Dans un premier temps, nous avons pu observer par Western blot que ces trois substances activent la MAP kinase ERK ainsi que la kinase Akt via leur phasphorylation. De plus, la voie mTOR/S6K, connue pour son implication dans la régulation de l'initiation de la traduction est aussi fortement activée par ces trois substances. Dans un second temps, nous avons voulu déterminer I implication de chacune de ces voies de signalisation dans l'augmentation de l'expression de la protéine MCT2 observée après stimulation à la NA, à l'insuline et à l'IGF-1. Pour ce faire, nous avons utilisé des inhibiteurs spécifiques de chacune de ces voies. (Vous avons observé que les inhibiteurs des voies PI3 kinase et mTOR (LY294002 et rapamycin respectivement), prévenaient fortement l'augmentation de l'expression de MCT2 induite par la NA, l'insuline ou (IGF-1. A l'inverse, les inhibitions de la MAP kinase .kinase MEK ainsi que de la MAP kinase p38 (par l'utilisation des inhibiteurs spécifiques PD98059 et SB202190 respectivement) n'ont eu qu'un léger effet dans ces mêmes conditions. Ces résultats suggèrent que la NA, 'l'insuline et I~GF-1 régulent l'expression de la protéine MCT2 par un mécanisme commun impliquant probablement la voie Akt/PKB et l'activation de la traduction via mTOR. En conclusion, considérant l'implication de la NA, de l'insuline et de I`IGF-1 dans la plasticité synaptique, le contrôle traductionnel étroit exercé par ces substances sur l'expression de MCT2 pourrait être un moyen d'alimenter en substrats énergétiques autres que le glucose les synapses activées et également d'adapter l'approvisionnement en substrats énergétiques au niveau d'activité. Résumé « grand public » Le cerveau est un organe qui réalise des tâches complexes nécessitant un apport important en énergie. La principale source d'énergie du cerveau est le glucose. Bien que le cerveau ne représente que 2% de la masse corporelle, il consomme à lui seul plus de 25% du glucose et 20% de l'oxygène provenant de la circulation sanguine. La nécessité d'un tel apport en énergie réside dans la nature -même du fonctionnement des milliards de neurones qui utilisent des signaux électriques et chimiques pour communiquer entre eux. Hormis l'utilisation massive du glucose comme source d'énergie, le cerveau est capable de consommer d'autres substrats énergétiques dans certaines conditions physiologiques ou pathologiques. Les monocarboxylates (lactate, pyruvate et corps cétoniques) font partie de ces autres sources d'énergie. Contrairement au glucose, les monocarboxylates ne diffusent pas facilement de la circulation sanguine vers les neurones. Afin de pouvoir être consommés par les neurones, ils doivent être transportés par un système adapté. Ce sont des transporteurs appelés transporteurs aux monocarboxylates ou MCT qui permettent le passage de ces substrats énergétiques du sang vers les neurones. Le but de ce travail de thèse a été de comprendre comment est régulée l'expression de MCT2, l'un de ces transporteurs exprimé spécifiquement à la surface des neurones. Cette étude nous a permis de mettre en évidence que le neurotransmetteur noradrénaline ainsi que les hormones insuline et IGF-1 (insulinlike growth factor-1) sont capables d'induire une augmentation d'expression de MCT2 à la surface des neurones en culture. Nous avons ensuite voulu déterminer par quels mécanismes de signalisation ces substances agissent sur l'expression de MCT2. Nous avons pu observer que la surexpression de la protéine MCT2 est due à une augmentation d'activité traductionnelle (la traduction étant une des étapes qui permet la synthèse des protéines) induite par le biais d'une voie de signalisation particulière. En conclusion, lorsque la noradrénaline, l'insuline ou 17GF-1 agissent sur les neurones, la traduction de la protéine MCT2 est activée et on observe une augmentation de l'expression de MCT2. Ce mécanisme pourrait permettre d'augmenter l'apport énergétique au niveau des neurones en augmentant le nombre de transporteurs pour les substrats énergétiques que sont les monocarboxylates. D'un point de vue physiologique, cette régulation d'expression pourrait jouer un rôle primordial dans des situations d'apprentissage et de mémorisation. Sur le plan pathologique, cela pourrait permettre de prévenir les dommages causes aux neurones dans certains cas d'atteintes cérébrales.
Resumo:
BACKGROUND AND PURPOSE: Second mitochondria-derived activator of caspase (SMAC)-mimetics are a new class of targeted drugs that specifically induce apoptotic cancer cell death and block pro-survival signaling by antagonizing selected members of the inhibitor of apoptosis protein (IAP) family. MATERIAL AND METHODS: The present study was designed to investigate the radiosensitizing effect and optimal sequence of administration of the novel SMAC-mimetic Debio 1143 in vitro and in vivo. Apoptosis, alteration of DNA damage repair (DDR), and tumor necrosis factor-alpha (TNF-α) signaling were examined. RESULTS: In vitro, Debio 1143 displayed anti-proliferative activity and enhanced intrinsic radiation sensitivity in 5/6 head and neck squamous cell carcinoma (HNSCC) cell lines in a synergistic manner. In vivo, Debio 1143 dose-dependently radio-sensitized FaDu and SQ20B xenografts, resulting in complete tumor regression in 8/10 FaDu-xenografted mice at the high dose level. At the molecular level, Debio 1143 combined with radiotherapy (RT) induced enhancement of caspase-3 activity, increase in Annexin V-positive cells and karyopyknosis, and increase in TNF-α mRNA levels. Finally, in a neutralization experiment using a TNF-α-blocking antibody and a caspase inhibitor, it was shown that the radiosensitizing effect of Debio 1143 is mediated by caspases and TNF-α. CONCLUSIONS: These results demonstrate that the novel SMAC-mimetic Debio 1143 is a radiosensitizing agent that is worthy of further investigation in clinical trials in combination with radiotherapy.
Resumo:
The multiplicity of cell death mechanisms induced by neonatal hypoxia-ischemia makes neuroprotective treatment against neonatal asphyxia more difficult to achieve. Whereas the roles of apoptosis and necrosis in such conditions have been studied intensively, the implication of autophagic cell death has only recently been considered. Here, we used the most clinically relevant rodent model of perinatal asphyxia to investigate the involvement of autophagy in hypoxic-ischemic brain injury. Seven-day-old rats underwent permanent ligation of the right common carotid artery, followed by 2 hours of hypoxia. This condition not only increased autophagosomal abundance (increase in microtubule-associated protein 1 light chain 3-11 level and punctuate labeling) but also lysosomal activities (cathepsin D, acid phosphatase, and beta-N-acetylhexosaminidase) in cortical and hippocampal CA3-damaged neurons at 6 and 24 hours, demonstrating an increase in the autophagic flux. In the cortex, this enhanced autophagy may be related to apoptosis since some neurons presenting a high level of autophagy also expressed apoptotic features, including cleaved caspase-3. On the other hand, enhanced autophagy in CA3 was associated with a more purely autophagic cell death phenotype. In striking contrast to CA3 neurons, those in CA1 presented only a minimal increase in autophagy but strong apoptotic characteristics. These results suggest a role of enhanced autophagy in delayed neuronal death after severe hypoxia-ischemia that is differentially linked to apoptosis according to the cerebral region.
Resumo:
OBJECTIVES: The pre-treatment of tumour neovessels by low-level photodynamic therapy (PDT) improves the distribution of concomitantly administered systemic chemotherapy. The mechanism by which PDT permeabilizes the tumour vessel wall is only partially known. We have recently shown that leukocyte-endothelial cell interaction is essential for photodynamic drug delivery to normal tissue. The present study investigates whether PDT enhances drug delivery in malignant mesothelioma and whether it involves comparable mechanisms of actions. METHODS: Human mesothelioma xenografts (H-meso-1) were grown in the dorsal skinfold chambers of 28 nude mice. By intravital microscopy, the rolling and recruitment of leukocytes were assessed in tumour vessels following PDT (Visudyne(®) 400 μg/kg, fluence rate 200 mW/cm(2) and fluence 60 J/cm(2)) using intravital microscopy. Likewise, the distribution of fluorescently labelled macromolecular dextran (FITC-dextran, MW 2000 kDa) was determined after PDT. Study groups included no PDT, PDT, PDT plus a functionally blocking anti-pan-selectin antibody cocktail and PDT plus isotype control antibody. RESULTS: PDT significantly enhanced the extravascular accumulation of FITC-dextran in mesothelioma xenografts, but not in normal tissue. PDT significantly increased leukocyte-endothelial cell interaction in tumour. While PDT-induced leukocyte recruitment was significantly blunted by the anti-pan-selectin antibodies in the tumour xenograft, this manipulation did not affect the PDT-induced extravasation of FITC-dextran. CONCLUSIONS: Low-level PDT pre-treatment selectively enhances the uptake of systemically circulating macromolecular drugs in malignant mesothelioma, but not in normal tissue. Leukocyte-endothelial cell interaction is not required for PDT-induced drug delivery to malignant mesothelioma.
Resumo:
Serum-free aggregating cell cultures of fetal rat telencephalon grown in the presence of 3 ng/ml (5 X 10(-10) M) epidermal growth factor (EGF) until day 12 showed 2- to 3-fold increased activities in the two glial enzymes, glutamine synthetase (GLU-S) and 2',3'-cyclic nucleotide 3'-phosphohydrolase (CNPase). This effect was concentration-dependent, with maximal stimulation in cultures treated daily with 3 ng/ml EGF. Addition of EGF during the first 10 culture days was sufficient to produce a maximal stimulation of both GLU-S and CNPase on day 19, whereas treatments starting on day 12 were ineffective. The stimulation of GLU-S preceded that of CNPase. The EGF-induced increase in GLU-S activity was not directly dependent on the presence of insulin, triiodothyronine, or hydrocortisone in the medium, whereas insulin was required for the stimulation of CNPase. A single dose of 5 ng/ml EGF on day 2 caused a slight but significant decrease in DNA synthesis after day 6. The present results indicate that in serum-free aggregating cell cultures of fetal rat telencephalon EGF partially inhibits DNA synthesis, and stimulates an early step in glial differentiation.
Resumo:
The monocarboxylate transporter 1 (MCT1 or SLC16A1) is a carrier of short-chain fatty acids, ketone bodies, and lactate in several tissues. Genetically modified C57BL/6J mice were produced by targeted disruption of the mct1 gene in order to understand the role of this transporter in energy homeostasis. Null mutation was embryonically lethal, but MCT1 (+/-) mice developed normally. However, when fed high fat diet (HFD), MCT1 (+/-) mice displayed resistance to development of diet-induced obesity (24.8% lower body weight after 16 weeks of HFD), as well as less insulin resistance and no hepatic steatosis as compared to littermate MCT1 (+/+) mice used as controls. Body composition analysis revealed that reduced weight gain in MCT1 (+/-) mice was due to decreased fat accumulation (50.0% less after 9 months of HFD) notably in liver and white adipose tissue. This phenotype was associated with reduced food intake under HFD (12.3% less over 10 weeks) and decreased intestinal energy absorption (9.6% higher stool energy content). Indirect calorimetry measurements showed ∼ 15% increase in O2 consumption and CO2 production during the resting phase, without any changes in physical activity. Determination of plasma concentrations for various metabolites and hormones did not reveal significant changes in lactate and ketone bodies levels between the two genotypes, but both insulin and leptin levels, which were elevated in MCT1 (+/+) mice when fed HFD, were reduced in MCT1 (+/-) mice under HFD. Interestingly, the enhancement in expression of several genes involved in lipid metabolism in the liver of MCT1 (+/+) mice under high fat diet was prevented in the liver of MCT1 (+/-) mice under the same diet, thus likely contributing to the observed phenotype. These findings uncover the critical role of MCT1 in the regulation of energy balance when animals are exposed to an obesogenic diet.
Resumo:
Aim: 5-fluoro-2'-deoxyuridine (FdUrd) depletes the endogenous 5'-deoxythymidine triphosphate (dTTP) pool. We hypothesized whether uptake of exogenous dThd analogues could be favoured through a feedback enhanced salvage pathway and studied the FdUrd effect on cellular uptake of 3'-deoxy-3'-18F-fluorothymidine (18F-FLT) and 5-125I-iodo-2'-deoxyuridine (125I-IdUrd) in different cancer cell lines in parallel. Methods: Cell uptake of 18F-FLT and 125I-IdUrd was studied in 2 human breast, 2 colon cancer and 2 glioblastoma lines. Cells were incubated with/without 1 µmol/l FdUrd for 1 h and, after washing, with 1.2 MBq 18F-FLT or 125I-IdUrd for 0.3 to 2 h. Cell bound 18F-FLT and 125I-IdUrd was counted and expressed in % incubated activity (%IA). Kinetics of 18F-FLT cell uptake and release were studied with/without FdUrd modulation. 2'-3H-methyl-fluorothymidine (2'-3H-FLT) uptake with/without FdUrd pretreatment was tested on U87 spheroids and monolayer cells. Results: Basal uptake at 2 h of 18F-FLT and 125I-IdUrd was in the range of 0.8-1.0 and 0.4-0.6 Bq/cell, respectively. FdUrd pretreatment enhanced 18F-FLT and 125I-IdUrd uptake 1.2-2.1 and 1.7-4.4 fold, respectively, while co-incubation with excess thymidine abrogated all 18F-FLT uptake. FdUrd enhanced 18F-FLT cellular inflow in 2 breast cancer lines by factors of 1.8 and 1.6, respectively, while outflow persisted at a slightly lower rate. 2'-3H-FLT basal uptake was very low while uptake increase after FdUrd was similar in U87 monolayer cells and spheroids. Conclusions: Basal uptake of 18F-FLT was frequently higher than that of 125I-IdUrd but FdUrd induced uptake enhancement was stronger for 125I-IdUrd in five of six cell lines. 18F-FLT outflow from cells might be an explanation for the observed difference with 125I-IdUrd.
Resumo:
A variety of chemokines and inflammatory molecules are concomitantly produced at target sites of leukocyte trafficking and homing, accounting for the complex cellular responses occurring in homeostasis and inflammation. The chemokine CXCL12 plays an essential and unique role in homeostatic regulation of leukocyte traffic and tissue regeneration. The chromatin protein HMGB1 is released by dying and distressed cells, and acts as a Damage Associated Molecular Pattern or alarmin, promoting cell migration towards the site of tissue damage. We show here that HMGB1 synergises with CXCL12 by forming a heterocomplex that we characterized by NMR chemical shift mapping. The heterocomplex enhances CXCR4-induced responses on cells of the immune system, acting exclusively through the CXCL12 receptor CXCR4, and not through the HMGB1 receptors RAGE, TLR2 and TLR4. FRET analysis show that CXCL12 and CXCL12+HMGB1 promote a different conformational change in the homodimer CXCR4. The enhancement induced by HMGB1 on CXCL12-induced migration is selective, since little changes in migration of neutrophils and PreB 300.19-CCR2+ or -CCR7+ are observed towards CXCL8 and CCR2 or CCR7 agonists. HMGB1 also promotes CXCL 12 release, which is ultimately responsible for the chemoattractant activities of HMGB1. This study highlights the role of HMGB1 in promoting CXCL12-dependent cell migration, and suggests a cooperative role of these two molecules in tissue repair as well as in pathological conditions, such as rheumatoid arthritis.
Resumo:
BACKGROUND: Cardiac toxicity is a side-effect of anti-cancer treatment including radiotherapy and this translational study was initiated to characterize radiation-induced cardiac side effects in a population of breast cancer patients and in experimental models in order to identify novel therapeutic target. METHODS: The size of the heart was evaluated in CO-HO-RT patients by measuring the Cardiac-Contact-Distance before and after radiotherapy (48months of follow-up). In parallel, fibrogenic signals were studied in a severe case of human radiation-induced pericarditis. Lastly, radiation-induced cardiac damage was studied in mice and in rat neonatal cardiac cardiomyocytes. RESULTS: In patients, time dependent enhancement of the CCD was measured suggesting occurrence of cardiac hypertrophy. In the case of human radiation-induced pericarditis, we measured the activation of fibrogenic (CTGF, RhoA) and remodeling (MMP2) signals. In irradiated mice, we documented decreased contractile function, enlargement of the ventricular cavity and long-term modification of the time constant of decay of Ca(2+) transients. Both hypertrophy and amyloid deposition were correlated with the induction of Epac-1; whereas radiation-induced fibrosis correlated with Rho/CTGF activation. Transactivation studies support Epac contribution in hypertrophy stimulation and showed that radiotherapy and Epac displayed specific and synergistic signals. CONCLUSION: Epac-1 has been identified as a novel regulator of radiation-induced hypertrophy and amyloidosis but not fibrosis in the heart.
Resumo:
In order to test whether an improvement of maximal sprinting speed after creatine (Cr) supplementation was due to the increase of stride frequency (SF), stride length (SL) or both, 7 subjects ran 4 consecutive sprints after 1 week of placebo or Cr supplementation. SF and SL were assessed by a triaxial accelerometer. Compared to the placebo, Cr induced an increase of running speed (+1.4% p < 0.05) and SF (+1.5%, p < 0.01), but not of SL. The drop in performance following repeated sprints was partially prevented by Cr. In conclusion, exogenous Cr enhanced sprinting performance by increasing SF. This result may be related to the recent findings of shortening in muscular relaxation time after Cr supplementation.
Resumo:
PURPOSE: In this study, we investigated the mechanisms by which temozolomide enhances radiation response in glioblastoma cells. EXPERIMENTAL DESIGN: Using a panel of four primary human glioblastoma cell lines with heterogeneous O(6)-methylguanine-DNA methyltransferase (MGMT) protein expression, normal human astrocytes, and U87 xenografts, we investigated (a) the relationship of MGMT status with efficacy of temozolomide-based chemoradiation using a panel of in vitro and in vivo assays; (b) underlying mechanisms by which temozolomide enhances radiation effect in glioblastoma cells; and (c) strategies to overcome resistance to radiation + temozolomide. RESULTS: Temozolomide enhances radiation response most effectively in glioblastomas without detectable MGMT expression. On concurrent radiation + temozolomide administration in MGMT-negative glioblastomas, there seems to be decreased double-strand DNA (dsDNA) repair capacity and enhanced dsDNA damage compared either with radiation alone or with sequentially administered temozolomide. Our data suggest that O(6)-benzylguanine can enhance the antitumor effects of concurrent radiation + temozolomide in MGMT-positive cells by enhancing apoptosis and the degree of dsDNA damage. O(6)-Benzylguanine was most effective when administered concurrently with radiation + temozolomide and had less of an effect when administered with temozolomide in the absence of radiation or when administered sequentially with radiation. Our in vivo data using U87 xenografts confirmed our in vitro findings. CONCLUSIONS: The present study shows that temozolomide enhances radiation response most effectively in MGMT-negative glioblastomas by increasing the degree of radiation-induced double-strand DNA damage. In MGMT-positive glioblastomas, depletion of MGMT by the addition of O(6)-benzylguanine significantly enhances the antitumor effect of concurrent radiation + temozolomide. These are among the first data showing mechanisms of synergy between radiation and temozolomide and the effect of MGMT.
Resumo:
Breathing-induced bulk motion of the myocardium during data acquisition may cause severe image artifacts in coronary magnetic resonance angiography (MRA). Current motion compensation strategies include breath-holding or free-breathing MR navigator gating and tracking techniques. Navigator-based techniques have been further refined by the applications of sophisticated 2D k-space reordering techniques. A further improvement in image quality and a reduction of relative scanning duration may be expected from a 3D k-space reordering scheme. Therefore, a 3D k-space reordered acquisition scheme including a 3D navigator gated and corrected segmented k-space gradient echo imaging sequence for coronary MRA was implemented. This new zonal motion-adapted acquisition and reordering technique (ZMART) was developed on the basis of a numerical simulation of the Bloch equations. The technique was implemented on a commercial 1.5T MR system, and first phantom and in vivo experiments were performed. Consistent with the results of the theoretical findings, the results obtained in the phantom studies demonstrate a significant reduction of motion artifacts when compared to conventional (non-k-space reordered) gating techniques. Preliminary in vivo findings also compare favorably with the phantom experiments and theoretical considerations. Magn Reson Med 45:645-652, 2001.
Resumo:
Interferon-gamma (IFN-gamma) modulates the expression of Class II major histocompatibility antigens (MHC), thus providing a potential regulatory mechanism for local immune reactivity in the context of MHC-restricted antigen presentation. Within the central nervous system (CNS), the expression of MHC Class II antigens has been demonstrated on human reactive astrocytes and glioma cells. In order to investigate the modulation of HLA-DR on normal astrocytes, two cell lines were grown from a 20-week-old fetal brain. In situ none of the fetal brain cells expressed HLA-DR as determined by immunohistology on frozen tissue sections. The two cell lines, FB I and FB II, expressed GFAP indicating their astrocytic origin. FB I was HLA-DR negative at the first tissue culture passages, but could be induced to express HLA-DR when treated with 500 U/ml IFN-gamma. FB II was spontaneously HLA-DR positive in the early passages, lost the expression of this antigen after 11 passages and could also be induced to express HLA-DR by IFN-gamma. The induction of HLA-DR expression was demonstrated both by a binding RIA and by immunoprecipitation using a monoclonal antibody (MAB) directed against a monomorphic determinant of HLA-DR. The HLA-DR alloantigens were determined on FB II cells after IFN-gamma treatment, by immunofluorescence and by cytotoxicity assays, and were shown to be DR4, DR6, Drw52, DRw53 and DQwl. These results show that human fetal astrocytes can be induced to express HLA-DR by IFN-gamma in vitro and support the concept that astrocytes may function as antigen-presenting cells.