• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 92
  • 18
  • 17
  • 7
  • 6
  • 4
  • 1
  • 1
  • Tagged with
  • 158
  • 158
  • 42
  • 36
  • 34
  • 32
  • 29
  • 26
  • 23
  • 21
  • 21
  • 19
  • 15
  • 15
  • 14
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
71

Procoagulant Extracellular Vesicles Alter Trophoblast Differentiation inMice by a Thrombo-InflammatoryMechanism

Markmeyer, Paulina, Lochmann, Franziska, Singh, Kunal Kumar, Gupta, Anubhuti, Younis, Ruaa, Shahzad, Khurrum, Biemann, Ronald, Huebner, Hanna, Ruebner, Matthias, Isermann, Berend, Kohli, Shrey 26 February 2024 (has links)
Procoagulant extracellular vesicles (EV) and platelet activation have been associated with gestational vascular complications. EV-induced platelet-mediated placental inflammasome activation has been shown to cause preeclampsia-like symptoms in mice. However, the effect of EV-mediated placental thrombo-inflammation on trophoblast differentiation remains unknown. Here, we identify that the EV-induced thrombo-inflammatory pathway modulates trophoblast morphology and differentiation. EVs and platelets reduce syncytiotrophoblast differentiation while increasing giant trophoblast and spongiotrophoblast including the glycogen-rich cells. These effects are plateletdependent and mediated by the NLRP3 inflammasome. In humans, inflammasome activation was negatively correlated with trophoblast differentiation marker GCM1 and positively correlated with blood pressure. These data identify a crucial role of EV-induced placental thrombo-inflammation on altering trophoblast differentiation and suggest platelet activation or inflammasome activation as a therapeutic target in order to achieve successful placentation.
72

The therapeutic/anti-carcinogenic effect of cord blood stem cells-derived exosomes in malignant melanoma

Naeem, Parisa January 2022 (has links)
Malignant melanoma is an invasive type of skin cancer with high mortality rates, if not detected promptly. The mortality trends are generally linked to multiple dysplastic nevi, positive family history, genetic susceptibility and phenotypic features including fair skin, freckles, numerous atypical nevi, light coloured hair and eyes, inability to tan and prolonged exposure to ultraviolet radiation B (UVB). To date, the major anti-cancer therapeutics for melanoma include surgery, chemotherapy, radiotherapy, and immunotherapy. Recently, extracellular vesicles, especially exosomes, have been highlighted for their therapeutic benefits in numerous chronic diseases such as cancer. Exosomes display multifunctional properties, including inhibition of cancer cell proliferation and initiation of apoptosis. Hence, this study aimed to evaluate the genotoxicity and cytotoxicity of cord blood stem cell-derived (CBSC) exosomes on 6 samples of peripheral blood lymphocytes taken from healthy individuals and melanoma patients and on 3 samples of melanoma (CHL-1) cells. The limited number of samples was due to the time limitations and restrictions that were in place due to the COVID-19 pandemic. In this in vitro study, the optimal concentration of CBSC-derived exosomes (0, 100, 200, 300, 400 μg/ml protein at 24, 48 and 72h treatments) was confirmed by the CCK-8 assay. CBSC exosomes (300 μg/ml) were used to treat lymphocytes and CHL-1 cells in the Comet assay and evaluated using the real-time polymerase chain reaction (qPCR) and Western blotting (WB). The data of the CCK-8 and Comet assays illustrated that exosomes exerted genotoxic effects on CHL-1 cells (CCK-8 assay, ****p < 0.0001), (Comet assay, *p <0.05, **p < 0.01). However, the data portraying a reduction in the viability of lymphocytes needs further investigation as the number of samples was limited, therefore, further clarification is required. Importantly, no significant adverse effect was observed in healthy lymphocytes when treated with the same exosomes (p = ns). When further challenged with UVA+B radiation, the exosomes did not induce any genoprotective effect on ROS-induced CHL-1 cells, compared to the positive control (p = ns). Our data insinuates that the damage might be caused by inducing apoptosis. The anti-tumourigenic potential of exosomes was observed by activating the p53-mediated apoptotic pathway in CHL-1 cells, up-regulating p53, p21 and caspase 3 and down-regulating BCL-2 at mRNA (**p < 0.01, ***p <0.001, ****p <0.0001) and protein levels (*p < 0.05, **p <0.01). The potency of CBSC exosomes in inhibiting cancer progression in CHL-1 cells whilst causing no harm to the healthy lymphocytes makes it an ideal potential candidate for anti-cancer therapy. More samples are required to evaluate the therapeutic effect of exosomes on lymphocytes from cancer patients to fully understand their mechanism of action.
73

Development of macrophage-targeted therapy using peptide/protein-loaded extracellular vesicles / ペプチド及びタンパク質搭載細胞外小胞を利用したマクロファージを標的とする疾患治療法の開発

Takenaka, Misako 23 March 2023 (has links)
京都大学 / 新制・課程博士 / 博士(薬科学) / 甲第24549号 / 薬科博第166号 / 新制||薬科||18(附属図書館) / 京都大学大学院薬学研究科薬科学専攻 / (主査)教授 髙倉 喜信, 教授 山下 富義, 教授 小野 正博 / 学位規則第4条第1項該当 / Doctor of Pharmaceutical Sciences / Kyoto University / DFAM
74

Development of immunotherapy using antigen-loaded multifunctional small extracellular vesicles / 抗原搭載多機能性細胞外小胞を利用した免疫療法の開発に関する研究

Liu, Wen 23 March 2022 (has links)
京都大学 / 新制・課程博士 / 博士(薬科学) / 甲第23837号 / 薬科博第152号 / 新制||薬科||17(附属図書館) / 京都大学大学院薬学研究科薬科学専攻 / (主査)教授 髙倉 喜信, 教授 山下 富義, 教授 小野 正博 / 学位規則第4条第1項該当 / Doctor of Pharmaceutical Sciences / Kyoto University / DFAM
75

Exploiting extracellular vesicles for ultrasensitive detection of cancer biomarkers from liquid biopsies

Notarangelo, Michela 23 October 2019 (has links)
Extracellular vesicles (EVs) are small membrane-surrounded structures containing transmembrane proteins and enclosing cytosolic proteins and nucleic acids. They are released in the extracellular space by both normal and neoplastic cells and play an important role in cell-cell communication in numerous physiological processes and pathological conditions, through the transfer of their functional cargo to recipient cells. EVs are highly abundant in biological fluids, and even more represented in cancer patients’ biofluids, therefore many studies suggested that they can be instrumental in liquid biopsies as prognostic markers or for early detection of tumors. Moreover, being secreted by potentially all the cells, they can serve in oncology to represent the tumor heterogeneity, which is underestimated by the current diagnostic tools. Given their small size, EVs are difficult to isolate in a high-throughput way and, therefore, one of the main obstacles to their clinical application, is that the existing isolation methods are impractical. During these years, I worked at the development and optimization of a novel technique that allows purification of heterogeneous EVs from biological fluids in an efficient, fast and reproducible way. This technique, named Nickel-Based Isolation (NBI), is a biochemical assay that allows obtaining polydisperse EVs in a physiological pH solution, therefore, preserving their morphology, heterogeneity, and stability. We tested and optimized this assay in protein-enriched systems and comparing it to the techniques currently used to characterize and measure EVs, such as flow cytometry and Tunable Resistive Pulse Sensing. We challenged the reproducibility of this method by isolating EVs from different biological fluids. Interestingly, the EVs purified with NBI result more intact and stable compared to the ones obtained with other methods, and can be studied in a clinical setting and used as an innovative tool for detection of molecules associated with diseases. We demonstrated the specificity of the procedure by using individual isolated vesicles in biochemical and molecular assay, optimized to characterize the biological content of EVs. We were able to detect picomolar concentration of PSMA on 105 EVs isolated from plasma of prostate cancer patients and BRAF-V600E transcript in just 103 EVs from the plasma of colon cancer patients, reaching unprecedented matching with tissue biopsy results. We also investigated the transcriptome of EVs isolated from glioblastoma cancer stem cells, in order to exploit the potential of EVs as diagnostic markers.
76

Las vesículas extracelulares derivadas de células mesenquimales estromales de pulpa dental como producto terapéutico frente a la respuesta inmune que se desencadena tras el infarto agudo de miocardio

Amaro Prellezo, Elena 02 August 2024 (has links)
[ES] El infarto agudo de miocardio (IAM) es una de las principales causas de morbilidad y mortalidad en los países desarrollados. A lo largo de los últimos años se ha visto que la respuesta inflamatoria que ocurre tras desencadenarse el IAM es muy importante en el desarrollo clínico de esta patología. Si se produce una respuesta inflamatoria exacerbada aumenta el riesgo de remodelado cardiaco adverso y fallo cardiaco, pero el hecho de que no se desencadene la respuesta inflamatoria también tiene consecuencias negativas. Debido a la importancia de la respuesta inflamatoria en el IAM, recientemente se han intentado desarrollar terapias dirigidas frente componentes celulares o moleculares que participan en esta respuesta. Dentro de estas terapias, la terapia celular con células mesenquimales estromales (MSC) se ha postulado como un buen candidato. Las MSC se caracterizan fundamentalmente por su capacidad inmunomoduladora, lo que ha conducido a su empleo como agentes terapéuticos en diferentes enfermedades que cursan con procesos inflamatorios. Sin embargo, a lo largo de los últimos años, numerosos estudios han mostrado que el efecto terapéutico de las MSC está mediado fundamentalmente por las vesículas extracelulares (EVs) que liberan. Estas EVs recapitulan los efectos terapéuticos de las células de origen, por lo que también presentan efectos inmunomoduladores. El empleo de las EVs de MSC como agentes terapéuticos presenta ventajas respecto al uso de las MSC como, por ejemplo, una mayor bioseguridad. No obstante, el uso clínico de las EVs todavía tiene que hacer frente a retos como la obtención de grandes cantidades de EVs que constituyan un producto clínico estable y homogéneo. En este trabajo se ha querido evaluar, por un lado, si las EVs obtenidas de diferentes biopsias de la misma fuente tisular de MSC pueden constituir un producto biológico homogéneo que presente las mismas características y funcionalidad. Por otro lado, se ha evaluado si estas EVs se pueden emplear como agente terapéutico frente a la respuesta inflamatoria que se desencadena tras el IAM. Para ello se ha estudiado el efecto inmunomodulador de las EVs sobre células del sistema inmune, fundamentalmente macrófagos, in vitro y en un modelo in vivo de IAM en ratas. Los resultados mostraron que las EVs favorecen la diferenciación de los macrófagos M1 proinflamatorios hacia un fenotipo similar al M2, aumentando la expresión de marcadores M2 y reduciendo la secreción de citocinas proinflamatorias. Además, las EVs promovieron la activación de neutrófilos in vitro y la reducción de su estrés oxidativo. La administración de EVs en ratas sometidas a IAM amortiguó la caída de la función cardiaca y limitó la extensión de la zona infartada a los 7 y 21 días postinfarto. Las EVs también redujeron el número de macrófagos y neutrófilos proinflamatorios dentro de la zona infartada, favoreciendo la resolución de la inflamación. En conclusión, las EVs empleadas en este trabajo han demostrado ser un producto biológico estable con independencia de la biopsia de la que proceden, y han demostrado ser capaces de ejercer respuestas pro-resolutivas eficaces en un modelo de isquemia miocárdica, lo que las convierte en potenciales agentes terapéuticos para tratar la inflamación en el IAM. / [CA] L'infart agut de miocardi (IAM) és una de les principals causes de morbiditat i mortalitat als països desenvolupats. Al llarg dels darrers anys s'ha vist que la resposta inflamatòria que passa després de desencadenar-se l'IAM és molt important en el desenvolupament clínic d'aquesta patologia. Si es produeix una resposta inflamatòria exacerbada augmenta el risc de remodelat cardíac advers i fallada cardíaca, però el fet que no es desencadeni la resposta inflamatòria també té conseqüències negatives. A causa de la importància de la resposta inflamatòria a l'IAM, recentment s'han intentat desenvolupar teràpies dirigides davant de components cel·lulars o moleculars que participen en aquesta resposta. Dins aquestes teràpies, la teràpia cel·lular amb cèl·lules mesenquimals estromals (MSC) s'ha postulat com un bon candidat. Les MSC es caracteritzen fonamentalment per la seva capacitat immunomoduladora, cosa que ha conduït a la seva ocupació com a agents terapèutics en diferents malalties que cursen amb processos inflamatoris. Tot i això, al llarg dels últims anys, nombrosos estudis han mostrat que l'efecte terapèutic de les MSC està intervingut fonamentalment per les vesícules extracel·lulars (EVs) que alliberen. Aquestes EVs recapitulen els efectes terapèutics de les cèl·lules dorigen, per la qual cosa també presenten efectes immunomoduladors. L'ús de les EVs de MSC com a agents terapèutics presenta avantatges respecte a l'ús de les MSC com, per exemple, una bioseguretat més gran. Tot i això, l'ús clínic de les EVs encara ha de fer front a reptes com l'obtenció de grans quantitats d'EVs que constitueixin un producte clínic estable i homogeni. En aquest treball s'ha volgut avaluar, d'una banda, si les EV obtingudes de diferents biòpsies de la mateixa font tissular de MSC poden constituir un producte biològic homogeni que presenti les mateixes característiques i funcionalitat. D'altra banda, s'ha avaluat si aquestes EVs es poden fer servir com a agent terapèutic davant de la resposta inflamatòria que es desencadena després de l'IAM. Per això s'ha estudiat l'efecte immunomodulador de les EV sobre cèl·lules del sistema immune, fonamentalment macròfags, in vitro i en un model in vivo d'IAM en rates. Els resultats van mostrar que les EVs afavoreixen la diferenciació dels macròfags M1 proinflamatoris cap a un fenotip similar al M2, augmentant l'expressió de marcadors M2 i reduint la secreció de citocines proinflamatòries. A més, les VE van promoure l'activació de neutròfils in vitro i la reducció del seu estrès oxidatiu. L'administració d'EVs en rates sotmeses a IAM va esmorteir la caiguda de la funció cardíaca i va limitar l'extensió de la zona infartada als 7 i 21 dies postinfart. Les EVs també van reduir el nombre de macròfags i neutròfils proinflamatoris dins de la zona infartada, afavorint la resolució de la inflamació. En conclusió, les EVs emprades en aquest treball han demostrat ser un producte biològic estable amb independència de la biòpsia de què procedeixen, i han demostrat ser capaços d'exercir respostes pro-resolutives eficaces en un model d'isquèmia miocàrdica, cosa que les converteix en agents terapèutics potencials per tractar la inflamació a l'IAM. / [EN] Acute myocardial infarction (AMI) is one of the main causes of morbidity and mortality in developed countries. Over the last few years, it has been shown that the inflammatory response that occurs after AMI is triggered is very important in the clinical development of this pathology. If an exacerbated inflammatory response occurs, the risk of adverse cardiac remodeling and heart failure increases, but failure to trigger the inflammatory response also has negative consequences. Because of the importance of the inflammatory response in AMI, recent attempts have been made to develop therapies that target cellular or molecular components involved in this response. Within these therapies, cell therapy with mesenchymal stromal cells (MSC) has been postulated as a good candidate. MSC are mainly characterized by their immunomodulatory capacity, which has led to their use as therapeutic agents in different diseases involving inflammatory processes. However, in recent years, numerous studies have shown that the therapeutic effect of MSCs is mainly mediated by the extracellular vesicles (EVs) they release. These EVs recapitulate the therapeutic effects of the cells of origin and therefore also have immunomodulatory effects. The use of MSC-EVs as therapeutic agents has advantages over the use of MSC, such as increased biosafety. However, the clinical use of EVs still faces challenges such as obtaining large quantities of EVs that constitute a stable and homogeneous clinical product. The aim of this study was to evaluate, on the one hand, whether EVs obtained from different biopsies of the same MSC tissue source can constitute a homogeneous biological product with the same characteristics and functionality. On the other hand, we have evaluated whether these EVs can be used as a therapeutic agent against the inflammatory response triggered after AMI. To this end, the immunomodulatory effect of EVs on immune system cells, mainly macrophages, was studied in vitro and in an in vivo model of AMI in rats. The results showed that EVs favored the differentiation of proinflammatory M1 macrophages towards an M2-like phenotype, increasing the expression of M2 markers and reducing the secretion of proinflammatory cytokines. In addition, EVs promoted the activation of neutrophils in vitro and the reduction of their oxidative stress. The administration of EVs in rats subjected to AMI blunted the decline in cardiac function and limited the extent of the infarct zone at 7- and 21-days post-infarction. EVs also reduced the number of proinflammatory macrophages and neutrophils within the infarct zone, favoring the resolution of inflammation. In conclusion, the EVs used in this work have been shown to be a stable biological product regardless of the biopsy from which they are derived and have been shown to be able to exert effective pro-resolving responses in a model of myocardial ischemia, making them potential therapeutic agents to treat inflammation in AMI. / Amaro Prellezo, E. (2024). Las vesículas extracelulares derivadas de células mesenquimales estromales de pulpa dental como producto terapéutico frente a la respuesta inmune que se desencadena tras el infarto agudo de miocardio [Tesis doctoral]. Universitat Politècnica de València. https://doi.org/10.4995/Thesis/10251/202973
77

Caracterização de vesículas extracelulares liberadas por células de melanoma murino tratadas com quimioterápicos: possível papel modulador na sobrevivência das celulas tumorais? / Characterization of extracellular vesicles released by murine melanoma cells treated with chemotherapeutic agents: a possible modulating role in cell survival?

Ikoma, Mariana Mari 05 September 2017 (has links)
O Melanoma é um tipo de neoplasia que se origina de melanócitos normalmente presentes na epiderme. Uma das características do melanoma é a capacidade de adquirir resistência a terapias. As células de melanoma podem aumentar a liberação de vesículas extracelulares (VEs) em resposta ao tratamento com quimioterápicos. A cisplatina (CDDP) e a temozolomida (TMZ) são drogas utilizadas para o tratamento de tumores. Ambas as drogas formam adutos no DNA, mas as vias de sinalização que deflagram a morte celular são distintas. O objetivo desse estudo é investigar a morte celular da linhagem B16-F10 na presença de VEs oriundas de células B16-F10 tratadas com cisplatina CDDP ou TMZ. Inicialmente as VEs oriundas de células de melanoma murino, B16-F10, tratadas com CDDP ou TMZ e seus controles, foram isoladas por ultracentrifugações sucessivas. Para os experimentos in vitro, as células foram tratadas com as drogas em combinação com as respectivas VEs. As amostras foram realizados avaliações de ciclo celular e de morte e ensaio clonogênico. Para os experimentos in vivo, as células B16-F10 foram pré-tratadas com VEs, e posteriormente, as células foram inoculadas via subcutânea em camundongos C57BL/6 e os tumores foram mensurados diariamente. Em nosso estudo concluimos que a metodologia do isolamento de VEs é eficiente. Além disso, observamos que o tratamento com CDDP ou TMZ aumenta a liberação de VEs por células tumorais. Apesar do resultado contraditorio, as VEs liberadas por células tumorais tratadas com quimioterápicos aumentam a capacidade de sobrevivência das células de melanoma in vitro. VEs oriundas de células de melanoma não participam inicialmente da sensibilização à morte de células tumorais causada pelas mesmas drogas, mas a longo prazo, as VEs oriundas de células tratadas com a TMZ podem conferir uma resposta celular de sobrevivência às células tumorais in vitro. In vivo, o resultado é inconclusivo, uma vez que para confirmar se as VEs fazem parte da adaptação tumoral conferindo fenômenos de sobrevivência celular in vivo, é necessário avaliar em outros modelos celulares e animais / Melanoma is a neoplasm derived from melanocytes normally present in the skin specifically in the epidermis. One of the malignancies of melanoma is the ability to acquire chemoresistance. Cisplatin (CDDP) and temozolomide (TMZ) are drugs used for the treatment of tumors. Both drugs can form alkylating adducts in DNA, however, the pathways that trigger cell death are distinct. Tumor cells, including melanoma, may increase the release of extracellular vesicles (EVs) in response to chemotherapeutic treatment. The aim of this study is to investigate the cell death phenomenon in B16-F10 cell line in presence of EVs derived from chemotherapeutic-treated B16-F10 cells. For in vitro experiments, the cells were treated with CDDP or TMZ in combination with EVs from chemotherapictreated samples. For in vivo experiments, B16-F10 cells were exposed to EVs and inoculated subcutaneously in C57BL/6 mice. The growth was measured daily. In this work, we established and characterized VEs released by melanoma cells treated with chemotherapics and we established chemotherapics treatments to isolate EVs for next EVs isolation. Our results showed that CDDP or TMZ treatment increase the release of EVs by tumor cells. The EVs released by melanoma cells after CDDP or TMZ treatment seem to increase the survival capacity of melanoma cells. Thus, we concluded that EVs derived from melanoma cells do not participate in the cell death sensitization induced by CDDP or TMZ. However, EVs derived from TMZ treated cells may offer a survival effect to tumor cells in vitro a long term. In vivo, The result is inconclusive since to confirm how VEs are part of the tumor adaptation conferring cellular survival phenomena in vivo, it is necessary to evaluate in other cellular and animal models
78

Vesículas extracelulares liberadas pelas células cancerosas modulam a proliferação, morte e migração celular no melanoma humano? / Extracellular vesicles released by cancer cells modulate the cell proliferation, death and migration in human melanoma?

Cardim, Sílvia Guedes Braga 06 October 2017 (has links)
As células que compõem o tumor podem interagir entre si, através da liberação e incorporação de vesículas extracelulares, muitas vezes contribuindo para a progressão tumoral. Dessa maneira, o presente trabalho teve como objetivo observar se as vesículas extracelulares , como as microvesículas e os exossomas liberados pelas células cancerosas em condições de estresse celular, após quimioterapia e indução de hipóxia conferem alguma vantagem adaptativa às células tumorais. Nossos resultados mostram que vesículas liberadas por células de melanoma humano em hipóxia ou normóxia apresentam tamanho médio característico de exossomos e microvesículas e não modulam os processos de proliferação, morte e migração celular. As vesículas liberadas pelas células após tratamento com o quimioterápico temozolamida também apresentam tamanho característico de exossomos e microvesículas; em adição, o tratamento com a temozolamida induziu um aumento na secreção dessas vesículas pelas células de melanoma. A incubação das células tumorais com vesículas oriundas da terapêutica com a temozolamida aumentou a proliferação celular, conferindo vantagem proliferativa às células de melanoma humano / Tumor cells can interact with each other by releasing and incorporating extracellular vesicles, contributing to tumor progression. Therefore, the aim of this study was to evaluate if extracellular vesicles, such as microvesicles and exossomes, released by cancer cells under cell stress conditions like chemotherapy and hypoxia, induce an adaptive advantage to tumor cells. Our results show that vesicles shed by human melanoma cells under hypoxia, or normoxia exhibit the characteristic size of exossomes and microvesicles and do not modulate cell proliferation, death or migration. The vesicles released by melanoma cells after temozolomide treatment also showed the average size of exossomes and microvesicles; moreover, temozolomide treatment induced an increase in extracellular vesicles shedding by tumor cells. Incubation of tumor cells with vesicles released under temozolamide therapeutics caused an increase in cell proliferation, providing a proliferative advantage to human melanoma cells
79

Optimisation de la thérapie photodynamique par la nanovectorisation du photosensibilisateur mTHPC à l’aide de vésicules extracellulaires / Optimization of photodynamic therapy by the nanovectorization of mTHPC photosensitizer using extracellular vesicles

Millard, Marie 14 December 2018 (has links)
La thérapie photodynamique (PDT) est un traitement alternatif à la chirurgie en oncologie utilisant un photosensibilisateur (PS), la lumière visible et l’oxygène moléculaire. La méta-tétra(hydroxyphényl)chlorine (mTHPC) est l’un des PS de deuxième génération les plus utilisés en clinique en raison de son absorption dans le rouge lointain et d’un rendement quantique en 1O2 élevé. De par sa nature hydrophobe, la mTHPC est partiellement agrégée dans la circulation sanguine diminuant sa biodistribution. Dans le but d’améliorer la sélectivité tumorale de la mTHPC, différentes stratégies de vectorisation ont été développées. La formulation liposomale de mTHPC non PEGylée (Foslip®) améliore la biodistribution ainsi que les propriétés pharmacocinétiques de la mTHPC. Cependant, une rapide destruction des liposomes en circulation ainsi qu’une rapide libération de la mTHPC sont des inconvénients majeurs. Une alternative possible est l’utilisation de vésicules extracellulaires (VE). Dérivées des cellules, les VE possèdent une stabilité naturelle dans la circulation sanguine et une capacité à transporter et délivrer leur contenu de manière spécifique aux cellules cancéreuses. Cette vectorisation est intéressante en PDT en raison d’une importante capacité d’encapsulation des porphyrines. Le but de cette étude était d’évaluer l’intérêt des VE en tant que nanovecteur de la mTHPC dans divers modèles précliniques comparé au Foslip®. Contrairement au Foslip®, l’intégrité membranaire des VE est conservée en présence de 20% de plasma. In vitro, les mTHPC-VE ont montré une internalisation cellulaire par un mécanisme actif d’endocytose. Dans un modèle cellulaire en 3D de sphéroïdes multicellulaires, les mTHPC-VE ont permis d’accroitre l’accumulation cellulaire, la diffusion au sein de ce modèle ainsi que l’efficacité PDT. In vivo, les mTHPC-VE apparaissent plus efficace au niveau PDT avec un retard de croissance tumorale significativement augmenté. En conclusion, l’intégration de la mTHPC au sein des VE améliore l’efficacité PDT dans les différents modèles d’étude. Le suivi des mTHPC-VE à l’aide d’un traceur radioactif chez la souris ainsi que l’étude du ciblage de la vascularisation tumorale seront étudiés dans la suite du travail / Photodynamic therapy (PDT) is an alternative treatment to surgery in oncology using photosensitizer (PS), light and oxygen. Meta-tetra(hydroxylphenyl)chlorin (mTHPC) is one of the most used PS in clinics due to its high absorption in the deep red and high 1O2 quantum yield. In order to improve the mTHPC tumor selectivity different attempts of nanovectorisation were conducted. Non-PEGylated liposomal mTHPC (Foslip®) increase biodistribution and pharmacokinetic properties. However, the rapid liposome destruction during circulation and rapid mTHPC release are obvious shortcomings. Alternatively, mTHPC vectorization could be realized by extracellular vesicles (EVs). Derived from the cell, EVs possess a natural stability in bloodstream and ability to transport and deliver cargo molecules into cancer cells. This formulation is interesting for PDT due to the ability to encapsulate porphyrins. The aim of the present study was to determine the interest of EVs as mTHPC nanocarriers in various preclinical models compared to Foslip®. In contrast to Foslip®, membrane integrity of mTHPC-EVs was conserved in 20% of plasma. In vitro, mTHPC-EVs showed cellular internalization by an active endocytosis mechanism. In a 3D model of spheroids, mTHPC-EVs have improved cellular uptake, better diffusion inside spheroid and increased PDT efficacy. In vivo, mTHPC-EVs appeared to be more potent in terms of PDT efficacy, with a tumor growth delay significantly higher. In conclusion, integration of mTHPC in EVs improves PDT efficacy in various preclinical models. The tracking of mTHPC-EVs using a radioactive tracer in xenografted rodents as well as the study of vascularization targeting will be studied in the next step of this work
80

Évaluation des vésicules extracellulaires dérivées de cellules cardiaques humaines comme une alternative à la greffe des cellules : applications dans un modèle d'insuffisance cardiaque chronique / Evaluation of extracellular vesicles secreted by human induced pluripotent stem cell-derived beating cardiomyocytes as an alternative to cell transplantation : applications to myocardial repair in a model of chronic heart failure

Kervadec, Anaïs 07 September 2017 (has links)
L’insuffisance cardiaque (IC) est un problème majeur de santé publique. La pénurie des greffons cardiaques et la résistance de nombreux patients aux traitements conventionnels ont poussé les chercheurs à développer de nouvelles thérapeutiques dont la thérapie cellulaire. Bien que l’idée initiale de la thérapie cellulaire ait été de repeupler la partie nécrosée du cœur par l’administration de cellules viables et fonctionnelles, leur disparition rapide alors que les bénéfices perdurent dans le temps a conduit à l’hypothèse que les cellules agiraient via un mécanisme paracrine. Les vésicules extracellulaires (VE), incluant les exosomes et les microparticules, seraient principalement impliquées dans ce processus. Elles agiraient ainsi comme de véritables navettes transportant des biomolécules actives permettant d’activer des voies de réparation endogènes dans le tissu traité. Ce projet de thèse a pu mettre en évidence : 1) La non-infériorité des VE issues de progéniteurs cardiovasculaires (Pg) dérivés de cellules souches embryonnaires humaines par rapport à leurs cellules d’origine dans un modèle murin d’IC chronique (ICC). Ces VE activeraient des voies de signalisation endogènes impliquées dans la stimulation de la prolifération cellulaire, la survie cellulaire, la réparation de l’ADN et la diminution de la fibrose. Leur contenu moléculaire spécifique, et notamment les microARN, pourrait être impliqué dans ces phénomènes. 2) L’importance du choix du type cellulaire dans la production de VE efficaces sur le plan thérapeutique puisque ni les VE dérivées de cardiomyocytes matures ni celles de cellules souches mésenchymateuses n’ont eu d’effets bénéfiques sur la fonction cardiaque de souris en ICC. 3) L’implication des VE dans l’effet paracrine des cellules, confirmée par l’amélioration de la fonction cardiaque chez des souris présentant une ICC traitées avec des VE issues de Pg dérivés d’iPS. Des tests fonctionnels in vitro ont montré que les VE auraient un rôle pro-angiogénique, pro-prolifératif et amélioreraient la survie des cellules. Une thérapie a-cellulaire aurait une réelle pertinence clinique en réglant une partie des problèmes techniques, immunologiques et sécuritaires associés aux greffes de cellules. Si cette hypothèse est confirmée, il pourrait en résulter une simplification des problèmes réglementaires, une diminution des coûts de production et de ce fait une plus grande diffusion clinique de la méthode. / Heart failure (HF) is a major public health concern. The lack of donor hearts and the resistance of numerous patients to conventional treatments has led scientists to develop new therapies such as cell therapy. The initial goal of cell therapy was to repopulate the infarcted heart by directly injecting viable and functional cells. However, the rapid disappearance of the transplanted cells contrasts with their long term, ongoing functional benefits, suggesting that cells may act through a paracrine mechanism. Extracellular vesicles (EV), including exosomes and microparticles, may be key to this process, acting as shuttles to transport bioactive macromolecules that stimulate endogenous repair pathways in the host tissue. This PhD project demonstrates: 1) The non-inferiority of EV secreted by cardiovascular progenitors (Pg) derived from human embryonic stem cells as compared to their parent cells in a mouse model of chronic HF (CHF). These EV could act by the activation of endogenous signaling pathways implicated in cell proliferation, survival, DNA repair and decreased fibrosis. Their specific content, such as miRNA, could be involved in these benefits. 2) The importance of the cell type in the production of therapeutically effective EV, since EV derived from mature cardiomyocytes and mesenchymal stem cells did not improve cardiac function in mice with CHF. 3) The importance of EV in paracrine effects of cells, confirmed by the improvement of cardiac function in mice with CHF treated with EV secreted by Pg derived from iPS cells. In vitro data shows that EV might have pro-angiogenic, pro-proliferative and pro-survival effects. An acellular therapy should be clinically relevant by reducing technical, immunological and safety problems associated with cell transplantation. If this hypothesis is confirmed, regulatory concerns would be simplified and production costs reduced, facilitating large-scale production.

Page generated in 0.4514 seconds