913 resultados para TRANSGENIC MOUSE MODEL


Relevância:

100.00% 100.00%

Publicador:

Resumo:

Cerebral amyloid angiopathy (CAA) is an age-associated disease characterized by amyloid deposition in cerebral and meningeal vessel walls. CAA is detected in the majority of the individuals with dementia and also in a large number of non-demented elderly individuals. In addition, CAA is strongly associated with Alzheimer's disease (AD) pathology. Mechanical consequences including intra-cerebral or subarachnoid hemorrhage remains CAA most feared complication, but only a small fraction of CAA results in severe bleeding. On the hand the non-mechanical consequences in cerebrovascular regulation are prevalent and may be even more deleterious. Studies of animal models have provided strong evidence linking the vasoactive A beta 1-40, the main species found in CAA, to disturbances in endothelial-dependent factors, disrupting cerebrovascular regulation Here, we aimed to review experimental findings regarding the non-mechanical consequences of CAA for cerebrovascular regulation and discuss the implications of these results to clinical practice. (C) 2012 Elsevier Inc. All rights reserved.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Anhidrotic Ectodermal Dysplasia (EDA), is the most frequent form among Ectodermal Dysplasias, hereditary genetic disorders causing ectodermal appendages defective development. Indeed, EDA is characterized by defective formation of hair follicles, sweat glands and teeth both in human patients and animals. EDA, the gene mutated in Anhidrotic Ectodermal Dysplasia, encodes Ectodysplasin, a TNF family member that activates NF-kB mediated transcription. This disease can occur with mutations in other EDA-NF-kB pathway members, as EDA receptor, EDAR and its adapter, EDARADD. Moreover, mutations in TRAF6, NEMO, IKB and NF-kBs genes are responsible for Immunodeficiency associated EDA (EDA-ID). Several molecules, as SHH, WNT/DKK, BMP and LTβ, have already been reported to be EDA pathway regulators or effectors although the knowledge of the full spectrum of EDA targets remains incomplete. During the first part of the research project a gene expression analysis was performed in primary keratinocytes from Wild-type and Tabby (EDA model mouse) mice to identify novel EDA target genes. Earlier expression profiling at various developmental time points in Tabby and Wild-type mouse skin reported genes differentially expressed in the two samples and, to increase the resolution to find genes whose expression may be restricted to epidermal cells, the study was extended to primary keratinocyte cultures established from E19 Wild-type and Tabby skin. Using microarrays bearing 44,000 gene probes, we found 385 “preliminary candidate” genes whose expression was significantly affected by Eda defect. By comparing expression profiles to those from Eda-A1 (where Eda-A1 is highly expressed) transgenic skin, we restricted the list to 38 “candidate EDA targets”, 14 of which were already known to be expressed in hair follicles or epidermis. This work confirmed expression changes for 3 selected genes, Tbx1, Bmp7, and Jag1, both in primary keratinocytes and in Wild-type and Tabby whole skin, by Q-PCR and Western blotting analyses. Thus, this study detected novel candidate pathways downstream of EDA. In the second part of the research project, plasmid constructs were produced and analyzed to create a transgenic mouse model for Immunodeficiency associated EDA disease (XL-EDA-ID). In particular, plasmids containing mouse Wild-type and mutated Nemo cDNA under K-17 epidermis-specific promoter control and a Flag tag, were prepared, on the way to confine transgene expression to mice epidermis and to determine EDA phenotype without immunodeficiency for a comparison to Tabby model phenotype. EDA-ID mutations reported in patients and selected for this study are: C417R (C409R in mouse), causing Zinc Finger protein domain destabilization and A288G (A282G in mouse) affecting oligomerization of the protein. Moreover, the ex-novo mutation, ZnF, C-terminal Zinc Finger domain deletion, was tested. Thus, the constructs were analyzed by transient transfection, Western blotting and luciferase assays techniques, detecting Nemo Wild-type and mutant protein products and residue NF-kB activity in presence of mutants, after TNF stimulation. In particular, MEF_Nemo-/- cell line was used to monitor NF-kB activity without endogenous Nemo gene. Results show reduced NF-kB activity in presence of mutated Nemo forms compared to Wild-type: 81% for A282G (A288G in human); 24% for C409R (C417R in human); 15% for ZnF. C409R mutation (C417R in human), reported in 6 EDA-ID human patients, was selected to prepare transgenic model mouse. Mice (white, FVP) born following K17-promoter-Flag-Nemo_C409R plasmid region pronuclear injection, were analyzed for the transgene presence in the genotype and a preliminar examination of their phenotype was performed. In particular, one mouse showed considerable coat defects if compared to Wild-type mice. This preliminar analysis suggests a possible influence of Nemo mutant over-expression in epidermis without immunodeficiency. Still, more microscopic studies to analyze hair subtypes, Guard, Awl and Zigzag (usually alterated inTabby mouse model), Immunohistochemistry experiments to detect epidermis restricted Nemo expression and sweat glands analysis, will follow. This and other transgene positive mice will be crossed with black mice C57BL6 to obtain at least two indipendent agouti lines to analyze. Theses mice will be used in EDA target genes detection through microarrays. Following, plasmid constructs containing other Nemo mutant forms (A282G and ZnF) might be studied by the same experimental approaches to prepare more transgenic model mice to compare to Nemo_C409R and Tabby mouse models.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Neuronal networks exhibit diverse types of plasticity, including the activity-dependent regulation of synaptic functions and refinement of synaptic connections. In addition, continuous generation of new neurons in the “adult” brain (adult neurogenesis) represents a powerful form of structural plasticity establishing new connections and possibly implementing pre-existing neuronal circuits (Kempermann et al, 2000; Ming and Song, 2005). Neurotrophins, a family of neuronal growth factors, are crucially involved in the modulation of activity-dependent neuronal plasticity. The first evidence for the physiological importance of this role evolved from the observations that the local administration of neurotrophins has dramatic effects on the activity-dependent refinement of synaptic connections in the visual cortex (McAllister et al, 1999; Berardi et al, 2000; Thoenen, 1995). Moreover, the local availability of critical amounts of neurotrophins appears to be relevant for the ability of hippocampal neurons to undergo long-term potentiation (LTP) of the synaptic transmission (Lu, 2004; Aicardi et al, 2004). To achieve a comprehensive understanding of the modulatory role of neurotrophins in integrated neuronal systems, informations on the mechanisms about local neurotrophins synthesis and secretion as well as ditribution of their cognate receptors are of crucial importance. In the first part of this doctoral thesis I have used electrophysiological approaches and real-time imaging tecniques to investigate additional features about the regulation of neurotrophins secretion, namely the capability of the neurotrophin brain-derived neurotrophic factor (BDNF) to undergo synaptic recycling. In cortical and hippocampal slices as well as in dissociated cell cultures, neuronal activity rapidly enhances the neuronal expression and secretion of BDNF which is subsequently taken up by neurons themselves but also by perineuronal astrocytes, through the selective activation of BDNF receptors. Moreover, internalized BDNF becomes part of the releasable source of the neurotrophin, which is promptly recruited for activity-dependent recycling. Thus, we described for the first time that neurons and astrocytes contain an endocytic compartment competent for BDNF recycling, suggesting a specialized form of bidirectional communication between neurons and glia. The mechanism of BDNF recycling is reminiscent of that for neurotransmitters and identifies BDNF as a new modulator implicated in neuro- and glio-transmission. In the second part of this doctoral thesis I addressed the role of BDNF signaling in adult hippocampal neurogenesis. I have generated a transgenic mouse model to specifically investigate the influence of BDNF signaling on the generation, differentiation, survival and connectivity of newborn neurons into the adult hippocampal network. I demonstrated that the survival of newborn neurons critically depends on the activation of the BDNF receptor TrkB. The TrkB-dependent decision regarding life or death in these newborn neurons takes place right at the transition point of their morphological and functional maturation Before newborn neurons start to die, they exhibit a drastic reduction in dendritic complexity and spine density compared to wild-type newborn neurons, indicating that this receptor is required for the connectivity of newborn neurons. Both the failure to become integrated and subsequent dying lead to impaired LTP. Finally, mice lacking a functional TrkB in the restricted population of newborn neurons show behavioral deficits, namely increased anxiety-like behavior. These data suggest that the integration and establishment of proper connections by newly generated neurons into the pre-existing network are relevant features for regulating the emotional state of the animal.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Chronic liver inflammation during viral hepatitis is a major health problem worldwide. The role of proinflammatory cytokines, like IL-12, in breaking hepatic immune tolerance, and inducing acute liver inflammation and virus clearance is not clear. Nor is clear its role in uncontrolled severe inflammatory response, leading to fulminant hepatitis and hepatic failure. This work, focused in the study of the role of endogenous produced IL-12 in inducing hepatic inflammatory responses, demonstrates: In vitro, using adenovirus coding for IL-12, that hepatocytes stimulate CD4+ T cells in a tolerogenic manner, and that endogenous IL-12 is able to switch the immune response into Th1; and in vivo, that endogenous IL-12 induces hepatocyte damage and virus elimination in mice infected with adenovirus. In addition, and in order to study in vivo the relevance of IL-12 in acute inflammation, conditional IL-12 transgenic mice expressing IL-12 in the liver after cre-recombinase mediated induction were generated. For this purpose, an IL-12 fusion protein was created, which demonstrated high levels of bioactivity. Induction of IL-12 expression during embryonic development was achieved by crossbreeding with Act-Cre transgenic mice; induction of IL-12 expression in adult mice was achieved by a plasmid coding for the cre-recombinase. This study demonstrates that after induction, IL-12 is expressed in the liver of the transgenic mice. It also demonstrates that hepatic expression of IL-12 induces splenomegaly and liver inflammation, characterized by large infiltrations in portal tracts and veins, associated with hepatic damage, necrosis areas and lethality. Furthermore, constitutive hepatic IL-12 expression does not lead to abortion, but to total lethality, short after delivery. In conclusion, in this study, a transgenic mouse model has been generated, in which the expression of active IL-12 in the liver can be induced at any time; this model will be very helpful for studying hepatic pathologies. This study has also demonstrated that hepatic produced IL-12 is able of breaking liver tolerance inducing inflammation, virus elimination, severe hepatocyte damage, and lethality. These findings suggest IL-12 as a key cytokine in acute liver inflammation and fulminant hepatic failure. 5.1 Future studies Once the importance of IL-12 in inducing hepatic inflammation and virus elimination was demonstrated in this study, understanding the mechanisms of the IL-12 induced liver damage, and more important, how to avoid it will be the main focus in the future. It is very important to achieve hepatic inflammation for a more effective and faster viral elimination, but avoiding the toxicity of IL-12, which leads to massive liver injury and lethality is obviously necessary to allow IL-12 as therapy. For that purpose, future studies will be mainly base on three different points: 1. The determination of different cell populations present in the hepatic infiltration, which of them are responsible for liver injury, and as well their state of activation. 2. The measure of other pro- and anti-inflammatory cytokines and chemokines, which can play a role in IL-12-induced liver inflammation and hepatocyte damage. For these purposes, specific blocking antibodies (anti TNF-alpha, anti IL-12, anti IFN-g) will be used. The study with different transgenic mice: TNF-alpha Receptor knockout, TGF-b, will also help in determining the role of those cytokines during IL-12-induced liver damage and lethality. 3. The establishing of liver pathology models (viral infection, tumours, auto-antigens) in mice. Induction of IL-12 at any time of the pathology development will help in clarifying the role of IL-12 in those models. Finally, the transgenic mice expressing IL-23 in the liver will be generated.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Der Transplantat-gegen-Leukämie (GVL) Effekt als immuntherapeutisches Mittel bei der allogenen hämatopoetischen Stammzell Transplantation (HSZT) ist hauptsächlich durch Spender Lymphozyten vermittelt, welche hämatopoetische Minor-Histokompatibilitäts Antigene bzw. Leukämie-assoziierte Antigene (z. B.: PRAME, p53) erkennen. Der adoptive Transfer von Leukämie-spezifischen T-Zellen kann den GVL-Effekt, ohne ein Auftreten einer Transplantat-gegen-Wirt Erkrankung (GVHD), steigern. Unter Verwendung von HLA-A2 und human CD8 transgenen Mäusen (CD8yCyA2Kb) konnten in dieser Arbeit PRAME spezifische CD8+ zytotoxischen T-Zellen generiert werden. Diese zytotoxischen CD8+ T-Zellen zeigten in Chromfreisetzungsuntersuchungen lytische Aktivität gegen eine Vielzahl von Zelllinien, die PRAME endogen prozessieren sowie gegen das spezifische PRAME-Peptid. Des Weiteren wurden die hier generierten T-Zellen auf ihre zytotoxische Aktivität gegen akute myeloische Leukämie Blasten hin untersucht, und diese Untersuchungen zeigten AML-Reaktivität der PRAME-spezifischen sowie der als Vergleich genutzten p53- und HLA-A2-spezifischen T-Zellen. Das Potenzial der PRAME-spezifischen ZTL die GVL-Immunität in vivo zu erhöhen ohne das Vorkommen einer GVHD wurde in einem Tumor-Protektions-Model unter der Nutzung von NOD/SCIDgcnull Mäusen untersucht. Die PRA100- bzw. p53-ZTL wurden adoptiv in NOD/SCIDgcnull Rezipienten transferiert und gleichzeitig wurden die Tiere mit PRAME-, oder p53-exprimierende Tumorzelllinien inokuliert. Die Reduktion des Tumorwachstums bestätigte die Spezifität der T-Zellen auch in vivo. In weiteren in vivo Experimenten wurden NOD/SCIDgcnull Mäuse mit AML-Blasten rekonstituiert. Durch die Applikation von nur CD34 positiven Zellen aus einer AML-Probe, oder einer CD56 depletierten Probe, konnten Rekonstitutionen in 95 % aller Versuche erfolgreich beendet werden. Wurde eine Rekonstitution mittels PCR- und FACS-Analysen diagnostiziert, so folgten mehrere Applikationen der PRAME- oder p53-spezifischen ZTL. In diesen Untersuchungen konnten wir in einem therapeutischen AML-in vivo-Modell zeigen, dass die in diesen Untersuchungen generierten/verwandten ZTL in der Lage sind AML-Blasten in vivo zu bekämpfen und so die leukämische Last der Tiere im Blut sowie in der Milz auf unter 1 % zu regulieren. Der prozentuale Anteil humaner AML Zellen im Knochenmark konnte deutlich gesenkt werden (< 10 %). Zusammenfassend sind die von uns generierten PRAME-spezifischen T-Zellen in der Lage, in vitro und auch in vivo, endogen prozessiertes Protein auf Zelllinien und AML-Blasten zu erkennen und zu lysieren. Auch die p53-ZTL, welche als eine weitere Antigen-spezifische ZTL-Population in vivo getestet wurden, zeigten GVL-Effekte. Die Kenntnis von Tumor- bzw. Leukämie assoziierten Antigenen und die daraus erwachsene Möglichkeit der Generierung krankheitsspezifischer ZTL bietet die Grundlage für eine spezifische Immuntherapie maligner Erkrankungen.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Im Laufe der Evolution müssen Sauerstoff-metabolisierende Organismen eine Reihe von Anpassungen entwickelt haben, um in der zytotoxischen oxidativen Umgebung der sauerstoff-haltigen Erdatmosphäre überleben zu können. Die im Rahmen dieser Arbeit durchgeführten vergleichenden Analysen mitochondrial kodierter und kern-kodierter Proteome mehrerer hundert Spezies haben ergeben, dass die Evolution eines alternativen genetischen Codes in Mitochondrien eine moderne Adaptation in diesem Sinne war. Viele aerobe Tiere und Pilze dekodieren in Abweichung vom genetischen Standard-Code das Codon AUA als Methionin. In der vorliegenden Arbeit wird gezeigt, dass diese Spezies dadurch eine massive Akkumulation der sehr leicht oxidierbaren Aminosäure Methionin in ihren Atmungskettenkomplexen erreichen, die generell ein bevorzugtes Ziel reaktiver Sauerstoffspezies sind. Der gewonnene Befund lässt sich widerspruchsfrei nur unter Annahme einer antioxidativen Wirkung dieser Aminosäure erklären, wie sie erstmals 1996 von R. Levine anhand von Oxidationsmessungen in Modellproteinen postuliert worden war. In der vorliegenden Arbeit wird diese Hypothese nun direkt mittels neuartiger Modellsubstanzen in lebenden Zellen bestätigt. Die durchgeführten bioinformatischen Analysen und zellbiologischen Experimente belegen, dass kollektive Proteinveränderungen die Triebkraft für die Evolution abweichender genetischer Codes sein können.rnDie Bedeutung von oxidativem Stress wurde darüber hinaus auch im Referenzrahmen einer akuten oxidativen Schädigung im Einzelorganismus untersucht. Da oxidativer Stress in der Pathogenese altersassoziierter neurodegenerativer Erkrankungen wie der Alzheimerschen Krankheit prominent involviert zu sein scheint, wurden die Auswirkungungen von Umwelt-induziertem oxidativem Stress auf den histopathologischen Verlauf in einem transgenen Modell der Alzheimerschen Krankheit in vivo untersucht. Dabei wurden transgene Mäuse des Modells APP23 im Rahmen von Fütterungsversuchen einer lebenslangen Defizienz der Antioxidantien Selen oder Vitamin E ausgesetzt. Während die Selenoproteinexpression durch die selendefiziente Diät gewebespezifisch reduziert wurde, ergaben sich keine Anzeichen eines beschleunigten Auftretens pathologischer Marker wie amyloider Plaques oder Neurodegeneration. Es war vielmehr ein unerwarteter Trend hinsichtlich einer geringeren Plaquebelastung in Vitamin E-defizienten Alzheimermäusen zu erkennen. Auch wenn diese Daten aufgrund einer geringen Versuchstiergruppengröße nur mit Vorsicht interpretiert werden dürfen, so scheint doch ein Mangel an essentiellen antioxidativen Nährstoffen die Progression in einem anerkannten Alzheimermodell nicht negativ zu beeinflussen.rn

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Da Tumorerkrankungen ein enormes Gesundheitsproblem in der westlichen Welt darstellen, wird eine Vielzahl neuer Behandlungsstrategien entwickelt. Neuartige Tumor-Therapeutika werden jedoch üblicherweise zunächst an Tiermodellen evaluiert, bevor sie am Menschen angewandt werden.rnIn der vorliegenden Arbeit wurde ein BAC-transgenes Mausmodell generiert, welches als autochthones Melanommodell zur Anwendung kommen sollte.rnZunächst wurde dafür ein DNA-Konstrukt erzeugt. Dieses enthält die Melanom-Onkogene BrafV600E, Cdk4R24C und Mitf deren Expression durch die Tamoxifen-induzierbare Rekombinase CreERT2 kontrollierbar sein sollte. Die Verwendung des Tyrosinasepromoters sollte die melanozytenspezifische Expression der eingebrachten Gene gewährleisten. Ein weiterer Bestandteil des Konstrukts ist ein Luziferase-Gen, welches die Lokalisierung Onkogen-exprimierender Zellen durch in vivo-Biolumineszenz-Imaging erlaubt, da die Onkogen- und Luziferase-Expression durch 2A-Sequenzen gekoppelt sind.rnVor der Generierung der transgenen Tiere sollten in vitro Analysen die Funktionalität des Konstruktteils, bestehend aus den Onkogenen und der Luziferase, klären. Zu diesem Zweck wurde die Zelllinie C22 mit einem Expressionsvektor transfiziert, welcher den genannten Konstruktteil enthielt. Es konnte ein Anstieg der Braf- und Cdk4-Expression auf Protein Ebene, das Vorhandensein von Luziferase-Aktivität und die Aktivierung des MAP-Kinase-Signalwegs nachgewiesen werden. Die Funktionalität des untersuchten Konstruktteils war damit nahegelegt und die Generierung der transgenen Tiere wurde fortgesetzt.rnDie Pronukeus-Injektion resultierte schließlich in 3 Founder-Tieren, die mittels PCR und Southern Blot identifiziert wurden und die Bezeichnung „B6 tg Tyr iOnkogene“ (TyriOn) erhielten. Durch Verkreuzen der Founder-Tiere mit C57BL/6 Mäusen wurden im weiteren Verlauf 3 Linien erzeugt. Bei in vivo Biolumineszenz-Messungen zeigten Tiere der Linie D einen gewissen Grad an Hintergrund-Luziferase-Aktivität, die jedoch durch Tamoxifen-Injektionen verstärkt werden konnte. In den Folgegenerationen ging diese Tamoxifen-induzierte Verstärkung der Luziferase-Aktivität teilweise verloren. Es wurde die Vermutung angestellt, dass funktionelle und nicht-funktionelle Varianten des Transgens an unterschiedlichen Stellen im Genom von Founder D integriert hatten, und sich in den folgenden Generationen auf die Nachkommen verteilten. Die mangelnde Induzierbarkeit betroffener Tiere konnte nicht auf fehlende Integrität der Sequenz „iOnkogene“ in diesen Tieren oder auf nicht-funktionelle loxP-Stellen im Konstrukt zurückgeführt werden.rnTamoxifen-Injektionen führten in TyriOn-D Tieren im Laufe von 15 Monaten nicht zur Entwicklung von Tumoren. Ebenso wenig konnten in TyriOn-D / Cre del Tieren, welche die eingebrachten Onkogene maximal exprimieren sollten, Tumoren detektiert werden. Um zu analysieren, ob die eingebrachten Onkogene die Bildung von Tumoren begünstigen, wurden TyriOn-D Tiere mit dem Melanom-anfälligen Stamm MT/ret verkreuzt. Hierzu konnte im Rahmen dieser Arbeit noch kein Ergebnis erzielt werden. Allerdings konnte in Melanomen von TyriOn-D / MT/ret Tieren Luziferase-Aktivität bei in vivo Biolumineszenz-Messungen und CreERT2 RNA durch RT-PCR detektiert werden.rnTyriOn-D / MT/ret Tiere werden im weiteren Verlauf dieses Projektes nicht nur der Analyse der Melanomentwicklung dienen. Deren Tumore ermöglichen außerdem weitere Untersuchungen bezüglich der Funktionalität des Konstrukts, die teilweise in TyriOn Tieren keine Resultate ergaben.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Dendritische Zellen sind professionelle Antigenpräsentierende Zellen und übernehmen sowohl in der Aktivierung naiver T-Zellen als auch in der Aufrechterhaltung peripherer Toleranz eine zentrale Funktion. Ruhende Dendritische Zellen im immunologischen Steady State induzieren antigenspezifisch Toleranz in autoreaktiven T-Zellen, welche bei der negativen Selektion im Thymus nicht eliminiert wurden und verhindern somit die Entstehung von Autoimmunität. Mit Hilfe eines transgenen Maus Modells, welches die induzierbare Expression transgen kodierter CD8+ T-Zell-Epitope auf ruhenden Dendritischen Zellen erlaubt, konnten wir zeigen, dass die periphere Toleranz Induktion durch Dendritische Zellen in Abwesenheit von regulatorischen T-Zellen beeinträchtigt ist. Wir konnten verdeutlichen, dass für die Suppression von steady-state Dendritischen Zellen die Erkennung von MHC Klasse II Molekülen auf Dendritischen Zellen durch den T-Zell-Rezeptor regulatorischer T-Zellen zwingend erforderlich ist. In Abwesenheit dieser suppressiven Interaktion hatten Dendritische Zellen einen aktivierten Phänotyp und lösten eine funktionale T-Zell-Antwort aus, anstatt periphere Toleranz zu induzieren. Als Folge dessen entwickelten Mäuse, in denen Dendritische Zellen nicht antigenspezifisch mit suppressiven CD4+ T-Zellen interagieren konnten, spontane Autoimmunität, welche durch CD8+ T-Zellen mediiert wurde. Wir konnten weiterhin zeigen, dass der Verlust peripherer T-Zell Toleranz durch basale Level an Typ I Interferonen mediiert wird sowie durch CD40 Signale, welche von adaptiven Immunzellen geliefert werden.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

The term neurodegeneration defines numerous conditions that modify neuron’s normal functions in the human brain where is possible to observe a progressive and consistent neuronal loss. The mechanisms involved in neurodegenerative chronic and acute diseases evolution are not completely understood yet, however they share common characteristics such as misfolded proteins, oxidative stress, inflammation, excitotoxicity, and neuronal loss. Many studies have shown the frequency to develop neurodegenerative chronic diseases several years after an acute brain injury. In addition, many patients show, after a traumatic brain injury, motor and cognitive manifestations that are close to which are observed in neurodegenerative chronic patients. For this reason it is evident how is fundamental the concept of neuroprotection as a way to modulate the neurodegenerative processes evolution. Neuroinflammation, oxidative stress and the apoptotic process may be functional targets where operate to this end. Taking into account these considerations, the aim of the present study is to identify potential common pathogenetic pathways in neurodegenerative diseases using an integrated approach of preclinical studies. The goal is to delineate therapeutic strategies for the prevention of neuroinflammation, neurodegeneration and dysfunctions associated to Parkinson’s disease (PD) and cerebral ischemia. In the present study we used a murine model of PD treated with an isothiocyanate, 6-MSITC, able to quench ROS formation, restore the antioxidant GSH system, slow down the apoptotic neuronal death and counteract motor dysfunction induced by 6-OHDA. In the second study we utilized a transgenic mouse model knockout for CD36 receptor to investigate the inflammation involvement in a long term study of MCAo, which shows a better outcome after the damage induced. In conclusion, results in this study allow underlying the connection among these pathologies, and the importance of a neuroprotective strategy able to restore neurons activity where current drugs therapies have shown palliative but not healing abilities.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Immunotherapy with T cells genetically modified by retroviral transfer of tumor-associated antigen (TAA)-specific T cell receptors (TCR) is a promising approach in targeting cancer. Therefore, using a universal TAA to target different tumor entities by only one therapeutic approach was the main criteria for our TAA-specific TCR. Here, an optimized (opt) αβ-chain p53(264-272)-specific and an opt single chain (sc) p53(264-272)-specific TCR were designed, to reduce mispairing reactions of endogenous and introduced TCR α and TCR β-chains, which might lead to off-target autoimmune reactions, similar to Graft-versus-host disease (GvHD). rnIn this study we evaluated the safety issues, which rise by the risk of p53TCR gene transfer-associated on/off-target toxicities as well as the anti-tumor response in vivo in a syngeneic HLA-A*0201 transgenic mouse model. We could successfully demonstrate that opt sc p53-specific TCR-redirected T cells prevent TCR mispairing-mediated lethal off-target autoimmunity in contrast to the parental opt αβ-chain p53-specific TCR. Since the sc p53-specific TCR proofed to be safe, all further studies were performed using sc p53-specific TCR redirected T cells only. Infusion of p53-specific TCR-redirected T cells in Human p53 knock-in (Hupki) mice after lymphodepletion-preconditioning regimen with either sublethal body irradiation (5Gy) or chemotherapy (fludarabine and cyclophosphamide) in combination with vaccination (anti-CD40, CpG1668 and p53(257-282) peptide) did not result in a depletion of hematopoietic cells. Moreover, adoptive transfer of high numbers of p53-specific TCR-redirected T cells in combination with Interleukin 2 (IL-2) also did not lead to toxic on-target reactions. The absence of host tissue damage was confirmed by histology and flow cytometry analysis. Furthermore, p53-specific TCR-redirected T cells were able to lyse p53+A2.1+ tumor cells in vitro. However, in vivo studies revealed the potent suppressive effect of the tumor microenvironment (TME) mediated by tumor-infiltrating myeloid-derived suppressor cells (MDSC). Accordingly, we could improve an insufficient anti-tumor response in vivo after injection of the sc p53-specific TCR-redirected T cells by additional depletion of immunosuppressive cells of the myeloid lineage.rnTogether, these data suggest that the optimized sc p53(264-272)-specific TCR may represent a safe and efficient approach for TCR-based gene therapy. However, combinations of immunotherapeutic strategies are needed to enhance the efficacy of adoptive cell therapy (ACT)-mediated anti-tumor responses.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

The Nef protein of HIV-1 is important for AIDS pathogenesis, but it is not targeted by current antiviral strategies. Here, we describe a single-domain antibody (sdAb) that binds to HIV-1 Nef with a high affinity (K(d) = 2 × 10(-9)M) and inhibited critical biologic activities of Nef both in vitro and in vivo. First, it interfered with the CD4 down-regulation activity of a broad panel of nef alleles through inhibition of the Nef effects on CD4 internalization from the cell surface. Second, it was able to interfere with the association of Nef with the cellular p21-activated kinase 2 as well as with the resulting inhibitory effect of Nef on actin remodeling. Third, it counteracted the Nef-dependent enhancement of virion infectivity and inhibited the positive effect of Nef on virus replication in peripheral blood mononuclear cells. Fourth, anti-Nef sdAb rescued Nef-mediated thymic CD4(+) T-cell maturation defects and peripheral CD4(+) T-cell activation in the CD4C/HIV-1(Nef) transgenic mouse model. Because all these Nef functions have been implicated in Nef effects on pathogenesis, this anti-Nef sdAb may represent an efficient tool to elucidate the molecular functions of Nef in the virus life cycle and could now help to develop new strategies for the control of AIDS.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

PURPOSE: Although metabolic changes make diagnosis of insulinoma relatively easy, surgical removal is hampered by difficulties in locating it, and there is no efficient treatment for malignant insulinoma. We have previously shown that the high density of glucagon-like peptide-1 receptors (GLP-1R) in human insulinoma cells provides an attractive target for molecular imaging and internal radiotherapy. In this study, we investigated the therapeutic potential of [Lys(40)(Ahx-DTPA-(111)In)NH(2)]-Exendin-4, an (111)In-labeled agonist of GLP-1, in a transgenic mouse model of human insulinoma. EXPERIMENTAL DESIGN: [Lys(40)(Ahx-DTPA-(111)In)NH(2)]-Exendin-4 was assessed in the Rip1Tag2 mouse model of pancreatic beta-cell carcinogenesis, which exhibits a GLP-1R expression comparable with human insulinoma. Mice were injected with 1.1, 5.6, or 28 MBq of the radiopeptide and sacrificed 7 days after injection. Tumor uptake and response, the mechanism of action of the radiopeptide, and therapy toxicity were investigated. RESULTS: Tumor uptake was >200% injected activity per gram, with a dose deposition of 3 Gy/MBq at 40 pmol [Lys(40)(Ahx-DTPA-(111)In)NH(2)]-Exendin-4. Other GLP-1R-positive organs showed > or =30 times lower dose deposition. A single injection of [Lys(40)(Ahx-DTPA-(111)In)NH(2)]-Exendin-4 resulted in a reduction of the tumor volume by up to 94% in a dose-dependent manner without significant acute organ toxicity. The therapeutic effect was due to increased tumor cell apoptosis and necrosis and decreased proliferation. CONCLUSIONS: The results suggest that [Lys(40)(Ahx-DTPA-(111)In)NH(2)]-Exendin-4 is a promising radiopeptide capable of selectively targeting insulinoma. Furthermore, Auger-emitting radiopharmaceuticals such as (111)In are able to produce a marked therapeutic effect if a high tumor uptake is achieved.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

In many human carcinomas, expression of the lymphangiogenic factor vascular endothelial growth factor-D (VEGF-D) correlates with up-regulated lymphangiogenesis and regional lymph node metastasis. Here, we have used the Rip1Tag2 transgenic mouse model of pancreatic beta-cell carcinogenesis to investigate the functional role of VEGF-D in the induction of lymphangiogenesis and tumor progression. Expression of VEGF-D in beta cells of single-transgenic Rip1VEGF-D mice resulted in the formation of peri-insular lymphatic lacunae, often containing leukocyte accumulations and blood hemorrhages. When these mice were crossed to Rip1Tag2 mice, VEGF-D-expressing tumors also exhibited peritumoral lymphangiogenesis with lymphocyte accumulations and hemorrhages, and they frequently developed lymph node and lung metastases. Notably, tumor outgrowth and blood microvessel density were significantly reduced in VEGF-D-expressing tumors. Our results demonstrate that VEGF-D induces lymphangiogenesis, promotes metastasis to lymph nodes and lungs, and yet represses hemangiogenesis and tumor outgrowth. Because a comparable transgenic expression of vascular endothelial growth factor-C (VEGF-C) in Rip1Tag2 has been shown previously to provoke lymphangiogenesis and lymph node metastasis in the absence of any distant metastasis, leukocyte infiltration, or angiogenesis-suppressing effects, these results reveal further functional differences between VEGF-D and VEGF-C.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Members of the vascular endothelial growth factor (VEGF) family are critical players in angiogenesis and lymphangiogenesis. Although VEGF-A has been shown to exert fundamental functions in physiologic and pathologic angiogenesis, the exact role of the VEGF family member placental growth factor (PlGF) in tumor angiogenesis has remained controversial. To gain insight into PlGF function during tumor angiogenesis, we have generated transgenic mouse lines expressing human PlGF-1 in the beta cells of the pancreatic islets of Langerhans (Rip1PlGF-1). In single-transgenic Rip1PlGF-1 mice, intra-insular blood vessels are found highly dilated, whereas islet physiology is unaffected. Upon crossing of these mice with the Rip1Tag2 transgenic mouse model of pancreatic beta cell carcinogenesis, tumors of double-transgenic Rip1Tag2;Rip1PlGF-1 mice display reduced growth due to attenuated tumor angiogenesis. The coexpression of transgenic PlGF-1 and endogenous VEGF-A in the beta tumor cells of double-transgenic animals causes the formation of low-angiogenic hPlGF-1/mVEGF-A heterodimers at the expense of highly angiogenic mVEGF-A homodimers resulting in diminished tumor angiogenesis and reduced tumor infiltration by neutrophils, known to contribute to the angiogenic switch in Rip1Tag2 mice. The results indicate that the ratio between the expression levels of two members of the VEGF family of angiogenic factors, PlGF-1 and VEGF-A, determines the overall angiogenic activity and, thus, the extent of tumor angiogenesis and tumor growth.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Introduction: The Virtual Molecular Biology Lab is an innovative, computer-based educational program designed to teach advanced high school biology students how to create a transgenic mouse model in a simulated laboratory setting. It was created in an effort to combat the current decrease in adolescent enthusiasm for and academic achievement in science and science careers, especially in Hispanic students. Because studies have found that hands-on learning, particularly computer-based instruction, is effective in enhancing science achievement, the Virtual Lab is a potential tool for increasing the number of Hispanic students that choose to enter science fields. [See PDF for complete abstract]