• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 46
  • 12
  • 7
  • 3
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 104
  • 71
  • 64
  • 26
  • 13
  • 13
  • 13
  • 10
  • 10
  • 9
  • 9
  • 9
  • 8
  • 8
  • 7
  • 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.
11

Design and Synthesis of Orally Bioavailable Sphingosine Kinase 2 Selective Inhibitors

Sibley, Christopher David 16 July 2020 (has links)
In humans, mammals, and perhaps all vertebrates, sphingolipids exist as a family of cellular signaling molecules and have been shown to be involved in a wide range of biological processes ranging from proliferation to apoptosis. As such, sphingolipid signaling has garnered the attention of numerous researchers as an attractive candidate for pharmacological manipulation. The synthetic pathway of one prominent sphingolipid, sphingosine 1-phosphate (S1P), has been implicated in a variety of disease states such as cancer, sickle cell disease, multiple sclerosis, and renal fibrosis. Formation of S1P is facilitated from the ATP dependent phosphorylation of sphingosine (Sph) through its generative enzyme's sphingosine kinase 1 and 2 (SphK1 and SphK2). Inhibition of SphK1 and SphK2 results in the manipulation of S1P levels, which has been shown to be therapeutic in various animal models of disease. While there are multiple examples of potent SphK1-selective and dual SphK1/2 inhibitors, SphK2-selective inhibitors are scarce. Herein, we describe the design, synthesis and biological testing of SphK2-selective inhibitors. We first describe the discovery that introducing a trifluoromethyl group onto the internal aryl ring of our inhibitor scaffold led to superior selectivity and potency towards SphK2. We demonstrate that the trifluoromethyl moiety is interacting with a previously unknown side cavity in the substrate binding site of SphK2 that is unique and could be exploited in the design of SphK2-selective inhibitors. The synthesis of 21 derivatives with various substituents spanning off the internal aryl ring was completed, therefore characterizing the preferred size and chemical nature of moieties positioned in that portion of the binding site. This work led to the development of the most potent SphK2-selective inhibitor known at the time. We then describe the transformation of our SphK2-selective inhibitors into an orally bioavailable drug. We explain how the guanidine functionality on our inhibitor scaffold hinders our compounds from being orally bioavailable. Consequently, a library of 24 derivatives with various modifications to the guanidine functionality was synthesized and evaluated for improved orally bioavailability. Highlighted in this work is the development of the most potent SphK2-selective inhibitor currently known 3.14 (SLS1081832), which displays a hSphK2 Ki of 82 nM and 122-fold selectivity for SphK2. Chemical modification and in vivo assessment of 3.14 (SLS1081832) prodrugs was explored. / Doctor of Philosophy / In humans, sphingosine 1-phosphate (S1P) is a signaling molecule that is generated through an ATP dependent reaction of sphingosine (Sph) via sphingosine kinase 1 and 2 (SphK1 and SphK2). Furthermore, S1P has been shown to be implicated in various diseases such as cancer, sickle cell disease, multiple sclerosis, and renal fibrosis. Inhibition of SphK1 and SphK2 has been shown to be therapeutic towards the symptoms of these diseases. Therefore, in order to alleviate these disorders, the concentrations of S1P must be controlled through pharmacological inhibition of SphK1 and SphK2. There are multiple reported examples of potent SphK1-selective and dual SphK1/2 inhibitors; however, SphK2-selective inhibitors are scarce. This work describes the synthesis and biological assessment of 21 compounds for their effectiveness in selectively targeting and inhibiting SphK2. The work led to the discovery of a previously unrecognized side cavity in the binding pocket of SphK2 that enhances inhibitor potency and selectivity towards SphK2. Furthermore, studies characterizing the preferred size and chemical nature of moieties positioned in that portion of the binding site led to the development of the most potent SphK2- selective inhibitor known at the time. Building on this work, we next focused on the transformation of our SphK2-selective inhibitors into a drug that could be administered orally. We describe the synthesis of 24 compounds with various modifications to one portion of our scaffold and their effect on improved orally bioavailability. This work led to the development of the most potent SphK2-selective inhibitor currently known 3.14 (SLS1081832).
12

Structure Activity Relationship Studies on Isoform Selective Sphingosine Kinase Inhibitors

Congdon, Molly D. 23 August 2016 (has links)
A variety of diseases including Alzheimer's disease, asthma, cancer, fibrosis, multiple sclerosis, and sickle cell disease have been associated with elevated levels of sphingosine-1-phosphate (S1P). S1P, a pleiotropic lipid mediator involved in a broad range of cellular processes, is synthesized solely by the phosphorylation of sphingosine (Sph) and is catalyzed by the two isoforms of sphingosine kinase (SphK1 and SphK2). Therefore, SphKs are a potential therapeutic target; however, the physiological role of SphK2 is still emerging. In order to determine the role of SphK2 in vivo, more potent and selective small molecule inhibitors of SphK2, as well as dual inhibitors are necessary. Herein, explorations and advancements on the second generation SphK2 selective inhibitor SLR080811 are disclosed. Investigations into the lipophilic tail region of the hSphK2 inhibitor SLR080811 are detailed. This investigation highlights the dependency of SphK2 selectivity and potency on overall compound length. More importantly, this study identified the internal aryl ring of SLR080811 as a key pharmacophore of the scaffold. To further probe the significance of the aromatic region, the phenyl ring was replaced by a 2,6-naphthyl ether skeleton. Investigations into the tail region of this scaffold are described in detail. Key discoveries from this structure-activity relationship study include SLC5111312 (hSphK2 Ki = 0.90 μM, dual hSphK inhibitor), SLC5091592 (hSphK2 Ki = 1.02 μM, > 20-fold hSphK2 selective) and SLC5121591 (hSphK2 Ki = 0.61 μM, >16-fold hSphK2 selective). Molecular modeling studies with hSphK2 indicate that the extended aromatic group is able to participate in π-π stacking interactions with Phe548. In silico docking studies indicate that a guanidine hydrogen bond to Asp211 is key for SphK2 selectivity, and incorporation of a 3'-hydroxyl group on the pyrrolidine ring increases hydrogen bonding to Asp308, thereby increasing SphK1 potency and reducing selectivity. Additionally, biological studies employing SLC5111312 have helped to further elucidate the role of SphK2, suggesting that SphK2 has a catalytic role in the regulation of blood S1P levels. The shape of the hSphK2 binding pocket was probed by introducing an indole moiety in place of the naphthyl ring and varying its substitution pattern. One key discovery from this study is SLC5101465 (hSphK2 Ki = 0.09 μM, > 111 fold SphK2 selective), which has a 1,5-indole substitution pattern with an N-nonyl "tail". Molecular docking simulations highlight the importance of rotatable bonds and a relatively linear orientation between the "head group" and "tail group" to maintain essential hydrogen bond interactions to Asp residues with the guanidine moiety while minimizing steric interactions in the middle of the binding pocket. Expanding upon the 1,5-indole scaffold of SLC5101465, a series of aryl tail derivatives are examined. This study confirms the necessity of electron withdrawing groups located at the end of the inhibitor scaffold to optimize binding in the tail region of the SphK2 binding pocket. / Ph. D.
13

Rôle du métabolisme de la sphingosine 1-phosphate dans la résistance thérapeutique des cellules de mélanome aux inhibiteurs de BRAF / Role of sphingosine 1-phosphate metabolism in the therapeutic resistance of melanoma cells to braf inhibitors

Garandeau, David 22 June 2016 (has links)
Le traitement du mélanome métastatique a été révolutionné par le développement de thérapies ciblées, qui ont montré un bénéfice significatif sur la survie globale. En particulier, l'inhibition de la sérine-thréonine kinase BRAF, mutée dans 60% des mélanomes, par le Vémurafénib (PLX4032), a montré un gain de survie de 6 à 8 mois comparée à la chimiothérapie de référence, la Dacarbazine. Cependant, une très faible proportion de patients répond sur le long terme. En effet, la majorité des patients développent un échappement thérapeutique dans un délai médian de 6 mois. Des mécanismes cellulaires ont été mis en évidence dans l'apparition de cette résistance acquise, notamment l'implication de MITF, un facteur de transcription majeur des mélanocytes, ainsi que des modifications de l'expression de plusieurs membres de la famille de Bcl-2. Cependant, une meilleure compréhension des mécanismes de résistance aux thérapies anti-BRAF semble essentielle, tout comme l'utilisation de nouvelles approches thérapeutiques combinées afin d'optimiser l'efficacité des traitements et la durée du bénéfice clinique. Notre groupe a récemment identifié des altérations du métabolisme du céramide et de l'un de ses dérivés, la Sphingosine 1-phosphate (S1P), dans les cellules de mélanome humain comparé à des mélanocytes sains. En effet, nous avons montré que la S1P lyase (SPL), qui dégrade irréversiblement la S1P est sous exprimée. Au contraire, l'expression de la sphingosine kinase 1 (SK1), qui produit la S1P, est augmentée dans les cellules de mélanome, conséquence directe de la mutation BRAF. Ces perturbations ont pour effet d'augmenter les niveaux de S1P. Ce lysophospholipide favorise la survie cellulaire ainsi que la résistance vis-à-vis d'agents thérapeutiques dans diverses cellules tumorales. L'objectif de cette thèse a été d'évaluer si le métabolisme de la S1P peut moduler la résistance acquise des cellules de mélanome humain aux inhibiteurs de BRAF. Nous avons montré que la surexpression de la SPL ou l'inhibition pharmacologique de la SK1 (SKI-I) sensibilise les mélanomes métastatiques à l'apoptose induite par la thérapie ciblée. Ce phénomène est associé à une diminution de MITF et de l'une de ses cibles directes, la protéine anti-apoptotique Bcl-2. La diminution d'expression protéique de MITF peut être réversée par un traitement de S1P exogène. De plus, nous avons montré pour la première fois une augmentation de l'expression des récepteurs 1 et 3 à la S1P (S1PR1 et S1PR3), dans les cellules de mélanome présentant une résistance acquise au PLX4032. Ces modifications sont associées à l'expression accrue de MITF. La surexpression de la SPL, le traitement par le SKI-I ou par des inhibiteurs ciblant les S1PR1 et S1PR3, surmonte la résistance acquise de ces cellules au PLX4032 via la diminution d'expression des S1PRs, de MITF, et de Bcl-2. Par conséquent, en contrôlant l'expression de protéines clés de la survie et de la résistance, le métabolisme de la S1P représente une nouvelle approche thérapeutique pour améliorer l'efficacité des thérapies ciblées. / The treatment of metastatic melanoma has changed considerably in recent years with the development of targeted therapies, which have shown a significant benefit in overall survival. In particular, the inhibition of the frequently mutated serine-threonine kinase BRAF, by Vemurafenib (PLX4032) showed that survival rates increase by 6 to 8 months compared to standard chemotherapy, Dacarbazine. However, a very small proportion of patients will respond to the long term, and the majority of patients relapses in a median of 6 months. Cellular mechanisms have been identified in the appearance of this acquired resistance, including the involvement of MITF, a major transcription factor of melanocytes, as well as changes in the expression of several members of Bcl-2 family. However, a better understanding of these mechanisms seems essential, as is the use of new therapeutic strategies to optimize treatment efficacy and duration of clinical benefit. Our group recently showed some alterations of ceramide metabolism and its derivative sphingosine 1-phosphate (S1P) in human melanoma cells compared to healthy melanocytes. For instance, S1P lyase (SPL), which degrades S1P, is under-expressed. Conversely, sphingosine kinase 1 (SK1), which produces S1P, is over-expressed in tumor cells, as a direct result of BRAF mutation. These alterations increases the levels of S1P. This lysophospholipid promotes cell survival and the resistance to therapeutic agents in a variety of tumor cells. This PhD project aimed at defining whether S1P metabolism could modulate the resistance of human melanoma cells to PLX4032. Here, we show that SPL overexpression or pharmacological inhibition of SK1 by SKI-I sensitizes metastatic melanoma cells to PLX4032-induced apoptosis. This phenomenon is associated with a decreased expression of the master regulator of melanocyte differentiation MITF as well as its direct cellular target Bcl-2. The decrease in MITF protein can be reversed by treating cells with exogenous S1P. Interestingly, we also report for the first time an increased expression of SK1 as well as the S1P receptors, S1PR1 and S1PR3, in melanoma cells with acquired resistance to PLX4032 as compared to sensitive counterparts. These modifications are associated with high expression of MITF. Overexpression of SPL, treatment with SKI-I or antagonists of S1PR1 ans S1PR3, strongly overcomes acquired resistance to PLX4032 through a decrease in the expression of S1PR, MITF as well as Bcl-2. Thus, by controlling the expression of key proteins in melanoma cell survival and resistance, S1P metabolism could represent a new therapeutic approach to enhance the effectiveness of targeted therapies.
14

Sphingolipid metabolic enzymes modulate anticancer drug resistance

Min, Junxia. January 2006 (has links)
Thesis (Ph. D.)--University of Missouri-Columbia, 2006. / The entire dissertation/thesis text is included in the research.pdf file; the official abstract appears in the short.pdf file (which also appears in the research.pdf); a non-technical general description, or public abstract, appears in the public.pdf file. Title from title screen of research.pdf file viewed on (March 5, 2007) Vita. Includes bibliographical references.
15

The role of sphingosine kinase 2 in cell growth and apoptosis /

Sankala, Heidi Milka, January 2007 (has links)
Thesis (Ph. D.)--Virginia Commonwealth University, 2007. / Prepared for: Dept. of Biochemistry. Bibliography: leaves 99-126. Available online via the Internet.
16

Développement d'un capteur à base de polymère à empreintes moléculaires pour la quantification de la sphingosine 1-phosphate libre et circulante comme biomarqueur du mélanome cutané / Development of a moleculary imprinted polymer based sensor for the quantification of the free circulating sphingosine 1-phosphate as a biomarker in cutaneous melanoma

Sahun, Maxime 17 October 2017 (has links)
Le mélanome est le plus agressif et le plus sévère des cancers cutanés du fait de son fort potentiel métastatique. Pourtant à ce jour, aucun biomarqueur pour la détection précoce du mélanome n'est unanimement reconnu. Notre groupe a récemment démontré que le métabolisme du céramide est fortement altéré dès les premiers stades de la maladie, conduisant à l'augmentation de la production d'un dérivé du céramide, la sphingosine 1-phosphate (S1P). La S1P est sécrétée par les cellules du mélanome et a été identifiée comme une molécule majeure du remodelage du microenvironnement tumoral, qui favorise la progression du cancer. De façon physiologique, la S1P circulante se trouve majoritairement sous forme liée aux protéines de haute densité (HDLs), aux protéines de basse et très basse densité (LDLs et VLDLs) ainsi qu'à l'albumine. Les cellules de mélanome pourraient produire de la S1P non liée qui pourrait rendre compte des effets produits par ce lysosphopholipide sur les cellules du microenvironnement tumoral suite à sa fixation sur les récepteurs S1PR présents à la surface des cellules stromales. Ainsi, cette forme libre de S1P pourrait représenter un nouveau biomarqueur pour la détection précoce du mélanome. Cependant, il n'existe à l'heure actuelle aucun moyen permettant de la quantifier. Le but de ce travail interdisciplinaire a été de développer un nouveau capteur basé sur un polymère à empreintes moléculaires (MIP) dans le but de quantifier la S1P libre dans le sang de patients atteints de mélanome. Cette étude a été réalisée entre l'équipe " Ingénierie pour les sciences du vivant (ELiA) " du Laboratoire d'Analyse et d'Architecture des Systèmes (LAAS), et l'équipe " Sphingolipides, métabolisme, mort cellulaire et progression tumorale " du Centre de Recherche en Cancérologie de Toulouse (CRCT), en étroite collaboration avec l'équipe " Biomimétisme et structures bioinspirées " de l'Université Technologique de Compiègne (UTC). Dans un premier temps, nous avons synthétisé un nouveau MIP dirigé contre la S1P par une méthode de thermopolymérisation en masse. Nous avons caractérisé puis optimisé ce MIP en effectuant des mesures de spectrométrie de masse couplée à la chromatographie en phase liquide et des mesures de spectroscopie de fluorescence. Le MIP a été comparé à un NIP (Non Imprinted Polymer) et exposé à des analogues de la S1P afin d'évaluer sa sélectivité. Dans un second temps, en vue de l'utilisation d'un MIP en tant que couche sensible d'un futur capteur et pour anticiper son immobilisation et sa structuration sur le transducteur, nous avons mis au point un nouveau MIP photopolymérisable en 2D. Ce MIP a d'abord été structuré en motifs par photolithographie sur des surfaces de silicium puis validé par des mesures de microscopie de fluorescence. Le MIP a également été structuré sous la forme de couches minces sur les surfaces actives de capteurs de Microbalance à Cristal de Quartz (QCM) dans le but de le valider par cette méthode sans marquage. Enfin, nous avons exploré l'utilisation d'une fibre optique recouverte d'une couche de MIP photopolymérisé dans le but de détecter, par spectroscopie infrarouge, la liaison de la S1P avec le MIP à la surface de la fibre. / Melanoma is the most aggressive and severe form of cutaneous cancer due to its high metastatic potential. However, to date, no marker for the early detection of melanoma has been unanimously accepted. Our group has demonstrated that ceramide metabolism is strongly altered in melanoma, leading to the overproduction of sphingosine 1-phosphate (S1P), one of its derivatives. S1P is secreted by melanoma cells and has been identified as a critical molecule of tumor microenvironment remodeling that supports cancer progression. Physiologically, circulating S1P is predominantly linked to high density lipoproteins (HDLs), low and very low density lipoproteins (LDLs and VLDLs), as well as albumin. Melanoma cells produce unbound S1P that could be responsible for the effects induced by this lysophospholipid on the tumor microenvironment, as a result of its binding to S1PR receptors present on the surface of stromal cells. Thus, secreted tumor S1P could represent a new biomarker for the early detection of melanoma. However, there are currently no means to quantify it. The goal of this interdisciplinary work was to develop a new sensor based on a Molecularly Imprinted Polymer (MIP) in order to quantify unbound S1P present in the blood of melanoma patients. This study has been conducted between the "Engineering for Life science Applications (EliA)" group at the Laboratory for Analysis and Architecture of Systems (LAAS) and the "Sphingolipids, metabolism, cell death and tumor progression" group at the Cancer Research Center of Toulouse (CRCT), in strong collaboration with the team "Biomimetism and Bioinspired Structures" of the University of Technology of Compiègne (UTC). First, we synthesized a new MIP against S1P employing a bulk thermopolymerization approach. The resulting MIP was characterized and optimized by performing both mass spectrometry and fluorescence spectroscopy measurements. It was compared to a Non Imprinted Polymer (NIP) and exposed to S1P analogues to assess its selectivity. Second, in order to use the MIP as the sensitive layer of a future sensor and prepare its immobilization and structuration onto a transducer, we synthesized a new surface photopolymerizable MIP. This MIP was first structured by photolithography onto silicon substrates and validated by fluorescence microscopy measurements. The MIP was also structured as a thin layer onto Quartz Crystal Microbalance (QCM) chips in order to validate its binding capacities using this label-free method. Finally, the use of a MIP-coated optical fiber as an infrared sensor was explored, with the aim of detecting S1P in blood using Attenuated Total Reflectance (ATR) spectroscopy.
17

Trans Addition of B-X Reagents Across Polarized Triple Bonds and Development of Sphingosine-1-Phosphate Transport Inhibitors

Fritzemeier, Russell Glenn 31 March 2020 (has links)
Organoboron compounds are ubiquitous in organic chemistry. Fundamental transformations utilizing organoboron compounds are a necessary addition to any organic chemist's synthetic toolbox. In addition to their extensive use as synthetic intermediates, organoboron compounds are being increasingly studied for their material and medicinal properties. Excitingly, significant advances have been made over the years towards the synthesis of a wide variety of organoboron substrates. In the case of vinylboronic acids, synthesis primarily occurs through cis addition of boron reagents across triple bonds. However, methods affording trans addition products are scarce. Furthermore, many current methods rely on the use of expensive and toxic transition-metal catalysts. Herein, we describe the development of trans addition of boron reagents across polarized triple bonds to afford novel vinylboronic acids. Emphasis is placed on the transition metal-free nature of the reactions as well as the regio- and stereoselectivity observed in the products. In addition, the synthetic utility of the resulting trans addition products is demonstrated in the synthesis of biologically relevant molecules. We first describe the Brønsted base-mediated trans silaboration of propiolamides in which two functional groups with orthogonal reactivity are simultaneously installed. We then go on to describe an organocatalyzed trans hydroboration of propiolate esters as well as a complementary Brønsted base-mediated trans hydroboration reaction of propiolamides. To conclude this portion, we demonstrate how the products from the previous methods can be used to synthesize difluoroborylacrylamides which possess unique and versatile reactivity. Herein we disclose the first small-molecule inhibitors of the sphingosine-1-phosphate (S1P) transporter spinster homolog 2 (SPNS2). While little is known in regard to the structure and function of SPNS2, previous studies have demonstrated the vital role SPNS2 plays in S1P mediated processes and have identified SPNS2 as a potential clinical target. For example, SPNS2 is critical to S1P-mediated lymphocyte egress from primary lymphoid tissues. Thus, small molecule inhibition of SPNS2 represents a novel therapeutic strategy for the treatment of autoimmune disorders such as multiple sclerosis. In this study, we report the discovery of small molecule inhibitors that display low micromolar activity using a novel yeast-based SPNS2 assay. Inhibitor structure-activity-relationship studies led to the discovery of the imidazole-based amine inhibitor 7.54. Furthermore, administration of 7.54 to mice recapitulates the lymphopenic phenotype observed in previous SPNS2 knockout studies. / Doctor of Philosophy / Boron-containing compounds are important in organic chemistry and are involved in the synthesis of a variety of materials and medicines used in everyday life. As such, the ability to efficiently and sustainably prepare boron-containing compounds has far reaching consequences. Access to an important class of boron-containing compounds known as vinylboronic acids has previously been established; however, product selectivity is often limited to what is referred to as cis addition products. Furthermore, access to the corresponding trans addition products is often limited to processes involving expensive transition metal catalysts that produce environmentally toxic waste. Herein, novel transition metal-free trans addition processes are described for preparing vinylboronic acids. In addition, the application of the resulting products is demonstrated through the synthesis of biologically relevant compounds. Sphingosine-1-phosphate (S1P) is an important signaling lipid that is involved in a variety of physiological processes. Improper balance in the amount of S1P in the body is associated with a variety of disease states such as autoimmunity and cancer. Two drugs that inhibit S1P-mediated processes have been approved by the FDA, fingolimod (Gilenya®) and siponimod (Mayzent®). However, there are drawbacks to targeting the S1P receptor directly, including dose-limiting side effects that are associated with these drugs. Consequently, recent efforts have focused on developing new ways to control the effects of S1P. Herein, we describe the discovery and development of the first reported inhibitors of the S1P transporter, spinster homolog 2 (SPNS2). A library of compounds was synthesized and tested for SPNS2 inhibition. The resulting structure-activity-relationship studies led to the discovery of the imidazole-based propanamine derivative 7.54. Furthermore, we demonstrate the potential of SPNS2 inhibition to control the effects of S1P in mice. These studies provide a foundation for future SPNS2-based drug discovery that will hopefully lead to the development of improved therapies for the treatment of autoimmune disease and cancer.
18

Activation du facteur de transcription Hypoxia-Inducible Factor-1 par la sphingosine-1-phosphate chez les cellules vasculaires

Michaud Dumont, Maude 16 April 2018 (has links)
Hypoxia-inducible factor-1 (HIF-1) est un facteur de transcription hétérodimérique ubiquitaire responsable de l’activation de nombreux gènes essentiels à l’adaptation des cellules suite à une diminution de la disponibilité en oxygène. En raison de l’induction du facteur de croissance de l’endothélium vasculaire (VEGF), une puissante molécule pro-angiogénique, HIF-1 joue un rôle particulièrement important au niveau des cellules vasculaires et dans la formation de nouveaux vaisseaux sanguins. Dernièrement, plusieurs études ont clairement démontré que la sphingosine-1-phosphate (S1P) est également un facteur pro-angiogénique majeur. Relâché dans le sérum principalement par les plaquettes activées, ce phospholipide bioactif vital lie et stimule des récepteurs spécifiques des cellules endothéliales (ECs) et musculaires lisses vasculaires (VSMCs), engendrant ainsi une variété de réponses cellulaires cruciales et essentielles dans la régulation du système vasculaire dont la prolifération, la migration et la survie. D’autres études ont clairement démontré que des stimuli non-hypoxiques peuvent aussi mener à l’activation de HIF-1 en conditions normales d’oxygénation. Puisque HIF-1 et S1P jouent un rôle central au niveau de l’angiogenèse et de la biologie des cellules vasculaires et qu’ils sont tout deux impliqués dans la pathogénèse de maladies comme l’athérosclérose et le cancer, cette thèse visait à déterminer le rôle potentiel de la S1P dans l’induction et l’activation de HIF-1 au niveau vasculaire et à identifier les mécanismes moléculaires conduisant à cette activation. Brièvement, nous montrons que le traitement des ECs et des VSMCs avec la S1P induit fortement l’expression de la protéine HIF-1αla sous-unité active de HIF-1. Le complexe nucléaire ainsi formé est actif transcriptionnellement et se lie spécifiquement à la séquence promotrice de ses gènes cibles. Nous démontrons également que la stabilisation protéique, indépendante de pVHL (protéine von Hippel-Lindau), est le mécanisme principal à l’origine de cette induction et ce, suite à l’activation spécifique du récepteur S1P2. Finalement, l’expression de gènes dépendants de HIF-1, apportée par la S1P, est fortement diminuée suite à l’utilisation d’ARN interférants ciblant la protéine HIF-1α. Nous croyons que les résultats de ces travaux, qui identifient S1P comme étant un nouvel et puissant activateur de HIF-1, auront un impact certain sur différents aspects de la biologie vasculaire. / Hypoxia-inducible factor-1 (HIF-1) is a ubiquitous heterodimeric transcription factor responsible for the activation of many genes essential for adaptation to low oxygen conditions. Among these genes, HIF-1 strongly induces vascular endothelial growth factor (VEGF), a potent angiogenic molecule. Therefore, HIF-1 plays a crucial role in vascular cell biology and the formation of new blood vessels. Recent studies have clearly shown that sphingosine-1-phosphate (S1P) is also a key player in the angiogenic process. Released into circulation mainly upon platelet activation, this bioactive phospholipid binds to and activates specific receptors located on vascular endothelial (ECs) and smooth muscle cells (VSMCs). This leads to the stimulation of a wide range of essential vascular cell responses like proliferation, migration and survival, which are crucial in the regulation of the vascular system. Other studies have shown that non-hypoxic stimuli can also activate HIF-1 in oxygenated conditions. Since S1P and HIF-1 are both important regulators of vascular cell biology and especially angiogenesis and that they are also both implicated in the pathogenesis of different diseases like atherosclerosis and cancer, the goal of the present thesis was to determine whether S1P can modulate the vascular induction and activation of HIF-1 and to identify the molecular mechanisms underlying this activation. Briefly, we show that treatment of ECs and VSMCs leads to a strong induction of HIF-1α protein levels through the specific activation of the S1P type-2 receptor in a time and dose-dependant manner. We also demonstrate that the S1P-dependant HIF-1 nuclear complex formation, achieved through pVHL-independent (protein von Hippel-Lindau) stabilization of HIF-1α, is transcriptionally active and specifically binds to hypoxia-responsive elements. Moreover, S1P activates the expression of genes known to be closely regulated by HIF-1 and this induction could be blocked by the use of RNA interference oligonucleotides targeting HIF-1α protein. Thus, this work identifies S1P as a novel and potent non-hypoxic activator of HIF-1. We believe that understanding the role played by HIF-1 in S1P gene regulation will have a strong impact on different aspects of vascular biology.
19

Regulation of vascular development and homeostasis by platelet-derived Sphingosine 1-Phosphate / Régulation de l’homéostasie et du développement vasculaire par la Sphingosine 1-Phosphate

Gazit, Salomé 05 November 2015 (has links)
Résumé confidentiel / Confidential abstract
20

Regulation of vascular development and homeostasis by platelet-derived Sphingosine 1-Phosphate / Régulation de l’homéostasie et du développement vasculaire par la Sphingosine 1-Phosphate

Gazit, Salomé 05 November 2015 (has links)
Résumé confidentiel / Confidential abstract

Page generated in 0.0745 seconds