• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 155
  • 73
  • 16
  • 12
  • 10
  • 7
  • 7
  • 6
  • 4
  • 2
  • 1
  • 1
  • 1
  • Tagged with
  • 338
  • 99
  • 99
  • 90
  • 68
  • 58
  • 52
  • 52
  • 45
  • 44
  • 34
  • 33
  • 31
  • 31
  • 29
  • 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.
161

Interaction of a G protein-coupled receptor (Ste2p) of <i>Saccharomyces cerevisiae</i> with its ligand and its G-protein alpha subunit

Huang, Li-Yin 01 December 2011 (has links)
The G protein-coupled receptor (GPCR) family is composed of hundreds of members and is expressed in eukaryotes. Each GPCR has seven transmembrane domains and is in charge of sensing changes from the environment, transducing signals, and activating a series of biological responses. The signal transduction pathway of the receptor starts from sensing outside signal and then activates G proteins. This signaling requires a tight control for activation without which impaired cellular function leads to pathology. We have used the pheromone alpha-factor receptor (Ste2p) of the yeast Saccharomyces cerevisiae as a model system to understand ligand binding, receptor activation, and G protein interaction. One method we have used to study ligand binding is to incorporate the photo-reactive crosslinker p-benzoyl-L-phenylalanine (Bpa) into Ste2p to capture alpha-factor. This powerful tool requires the incorporation of Bpa, an unnatural amino acid, into Ste2p by a special genetic manipulation designed in the lab of Peter Schulz (Scripps Institute) and adapted by our lab for Ste2p. Another method to study ligand binding that we have adapted for use in our system is to incorporate a chemical crosslinker [3,4-dihydroxylphenylacetyl (DHPA)] into alpha-factor for periodate-mediated crosslinking to Ste2p. The interacting domain between alpha-factor and transmembrane domain 2 to 3 of Ste2p was identified after DOPAC crosslinking, cyanogen bromide digestion and MALDI-TOF mass spectrometry. After ligand binding, signal transduction is mediated by the interaction of activated Ste2p with its G protein (Gpa1p). We studied this interaction by replacing natural residues in the intracellular loop 3 of Ste2p and C-terminal end of Gpa1p with cysteine and then determining disulfide crosslinking between Ste2p and Gpa1p. Some residues were found to be in close proximity and displayed different interacting patterns due to conformational changes of the receptor upon ligand binding. The information we gathered here allows us to understand more about the physical interactions of alpha-factor, Ste2p, and Gpa1p and provides us insights about the initiation and activation of the signal transduction pathway of a peptide ligand receptor.
162

The role of beta-arrestin in regulating the muscarinic acetylcholine type II receptor

Jones, Kymry Thereasa 06 July 2007 (has links)
The muscarinic acetylcholine type 2 receptor (M2 mAChR), a member of the GPCR superfamily, is found throughout the parasympathetic nervous system where it controls pulmonary, urinary, and cardiac function, and neurotransmission. The molecular mechanisms that regulate M2 mAChR availability at the cell surface are an important component in controlling these physiological events. Since beta-arrestin proteins are known to regulate the activity of other GPCRs, we sought to identify their role in regulating M2 mAChR activity, a topic that remains contentious in the field. To achieve this goal we utilized mouse embryonic fibroblasts (MEFs) derived from beta-arrestin knockout mice lacking one or both isoforms (MEF KO1, KO2, or KO1/2 cells) in addition to exogenous expression of beta-arrestin mutants. This study demonstrates that agonist-induced internalization of M2 mAChR is beta-arrestin- and clathrin-dependent, and that the receptor stably co-localizes with beta-arrestin in early endosomal vesicles suggesting it behaves as a class B receptor. Next, we sought to identify beta-arrestin s function in regulating the post-endocytic trafficking (down-regulation) of the M2 mAChR. MEF KO1/2 cells were unable to down-regulate M2 mAChRs whereas MEF KO1 or KO2 cells retained the ability to do so. In MEFwt cells, both M2 mAChR and beta-arrestin exhibited basal ubiquitination that increased following agonist stimulation. Receptor degradation appeared to be regulated by the ubiquitination status of beta-arrestin 2, since expression of a chimeric â-arrestin 2 form fused to ubiquitin increased both constitutive and agonist-promoted down-regulation, whereas expression of a beta-arrestin 2 mutant lacking putative ubiquitination sites, beta-arrestin 2K18R, K107R, K108R, K207R, K296R, significantly blocked degradation while internalization and stable association remained intact. Upon further analysis, the beta-arrestin 2K18R, K107R, K108R, K207R, K296R mutant blocked delivery of M2 mAChR to the late endosome/lysosome, presumably where degradation occurs. Inhibition of proteasome-dependent recycling of ubiquitin blocked receptor down-regulation without affecting internalization or the ubiquitination state of the M2 mAChR while ubiquitination of beta-arrestin 2 diminished significantly. These results support a role for ubiquitinated beta-arrestin in mediating M2 mAChR sorting and degradation in the lysosome. Collectively, these studies give us new insight on the function of beta-arrestin in regulating the activity of the M2 mAChR.
163

G Protein-Coupled Receptors; Discovery of New Human Members and Analyses of the Entire Repertoires in Human, Mouse and Rat

Gloriam, David E. January 2006 (has links)
G protein-coupled receptors (GPCRs) are signal mediators that have a prominent role in the regulation of physiological processes and they make up the targets for 30-45% of all drugs. Papers I and II describe the discovery of new human GPCRs belonging to the Rhodopsin family, a family which contains many common drug targets. The new receptors have only weak relationships to previously known GPCRs. However, they have been evolutionary conserved in several species and most of them display distinct expression patterns. In paper III we identified new human GPCRs belonging to the Adhesion family, which is characterised by very long N-termini containing conserved domains. The different compositions of conserved domains as well as the expression patterns suggest that the Adhesions can have several different functions. In paper IV we revealed remarkable species variations in the repertoires of Trace Amine-Associated Receptors (TAARs), which are relatives of the biogenic amine receptors. The human, mouse and rat TAAR genes are located in only one locus and are therefore most likely the result of gene tandem duplications. 47 of the 57 zebrafish TAARs were mapped to nine different loci on six chromosomes containing from 1 to 27 genes each. This study suggests that the TAARs arose through several different mechanisms involving tetraploidisation, block duplications, and local duplication events. Papers V and VI are overall analyses of the repertoires of GPCRs in humans, mice and rats; which contain approximately 800, 1800 and 1900 members, respectively. The repertoires were compared to distinguish between species-specific and common (orthologous) members, something which is important for example when predicting drug effects from experiments in rodents. The Glutamate, Adhesion, Frizzled and Secretin families show no or very little variation between human and rodents, whereas the repertoires of olfactory, vomeronasal and Taste2 receptors display large differences between all three species.
164

Molecular mechanism of MC1R association with skin cancer risk phenotypes

Ms Kimberley Beaumont Unknown Date (has links)
The melanocortin-1 receptor (MC1R) is a G-protein coupled receptor (GPCR) expressed on the surface of the melanocyte. MC1R activation after UV exposure results in the production of the dark eumelanin pigment and the tanning process in humans, providing protection from UV induced DNA damage. MC1R activation has also recently been linked to DNA repair. The MC1R gene is highly polymorphic in Caucasian populations with a number of MC1R variant alleles associated with red hair, fair skin, poor tanning and increased risk of melanoma and non-melanoma skin cancer. These MC1R variant receptors were thought to be loss of function, however the type of defect and the extent of the loss of function for individual variants was relatively unknown before the commencement of this PhD project. Many GPCR mutant proteins are intracellularly retained, resulting in a loss of signalling ability. To determine if this was the case for MC1R variant receptors, the localisation of the wild type and variant MC1R protein was investigated using immunofluorescence and radio-ligand binding on transfected melanocytic cells as well as primary melanocyte strains. For the first time, several MC1R variants including V60L, R151C, I155T, R160W and R163Q, were shown to have reduced cell surface expression compared to wild type MC1R. cAMP assays were used to determine the signalling ability of activated wild type and variant MC1R, importantly, variant receptors with reduced cell surface expression showed corresponding impairment in cAMP signalling. In contrast, the R142H and D294H variants, which have normal cell surface expression but significantly impaired cAMP signalling, are thought to have a defect in G-protein coupling. Some MC1R variants were found to have dominant negative activity on the wild type receptor in co-expression studies, this result may explain the MC1R heterozygote effect on human pigmentation phenotypes. This dominant negative effect resulted in either reduced wild type cell surface expression or reduced G-protein coupling and may be mediated by receptor dimerisation. In order to validate the in vitro studies, comparison of variant receptor characteristics with skin and hair colour data of individuals both homozygous and heterozygous for MC1R variant alleles was performed. This revealed parallels between variant MC1R cell surface expression, functional ability, dominant negative activity and the strength of the effects of variant alleles on human pigmentation. From the in vitro functional studies, it was clear that most variant receptors retained some signaling ability, although the relative abilities varied. An important unanswered question in the literature was whether the phenotype of carriers of the high penetrance MC1R variant alleles was actually representative of complete loss of function for MC1R. Due to the rarity of MC1R null alleles they had only previously been found in the heterozygous state, however we described the phenotype of one individual compound heterozygous for two frameshift mutations resulting in an individual unable to produce any functional MC1R protein. Phenotypic analysis indicated that red hair and fair skin is found in the absence of MC1R. Finally, preliminary studies using low temperature, chemical or pharmacological chaperones indicated that the cell surface expression of some MC1R variants could be rescued in cell transfection experiments. This resulted in a restoration of signaling ability after stimulation with agonist. These studies into the localization and function of MC1R variants have contributed to a greater understanding of the molecular mechanism underlying the association of MC1R with skin cancer risk phenotypes, and may lead to future drug based therapies that are able to rescue the function of MC1R variants that are intracellularly retained.
165

Regulating Protease Activated Receptor 2

Yung Suen Unknown Date (has links)
Protease-Activated Receptors (PARs) belong to an unusual family of G Protein Coupled Receptors (GPCRs). Each of the four known members is activated by its own N-terminus exposed by proteolytic cleavage and there is no other endogenous agonist known to date. PAR2 is the second member of the family and it has been implicated in wide range of pathophysiological conditions, particularly in various inflammatory diseases and cancers. In contrast, very little is known about the PAR2 receptor itself despite having been discovered more than 10 years ago. The purpose of this project was to improve our understanding of PAR2 regulation by discovering new agonists and antagonists and using them to probe the structural and functional properties of the receptor. Chapter 1 provides a brief literature overview of the initial discovery of PAR2, what is known about the mechanism of receptor activation, information on the structures and properties of current agonists and an antagonist for PAR2, and the putative physiological roles of human PAR2. As well, it summarizes the aims of this thesis. Chapter 2 investigates the regulation of gene expression by two different agonists of PAR2, a synthetic hexapeptide, 2f-LIGRLO-NH2, and the endogenous activator, trypsin, the idea being that genes up- or down- regulated by both agonists may more accurately profile PAR2-selective events. The effects of PAR2 activation on gene transcription in the human kidney HEK293 cell line were studied using a DNA microarray consisting of 19,000 human genes in an attempt to broadly cover the human genome and associated cell pathways with PAR2 activation. About 2,500 genes were regulated similarly by both agonists and, for genes expressed more than 5-fold, the mRNA results were further analyzed by quantitative RT-PCR techniques. PAR2 activation was shown to be associated with cellular metabolism, cell cycle, mitogen-activated protein kinase pathways, histone deacetylase and sirtuin enzymes, inflammatory cytokines and anti-complement function. Chapter 3 described a range of molecular events surrounding the activation of the receptor. PAR2 mRNA expression was quantitated by qRT-PCR and cross-checked with an intracellular Ca2+ assay. In this way whole cell PAR2 could be correlated with cell surface expression of PAR2. Three cell lines expressing high levels of PAR2 were chosen for subsequent experiments, these being colorectal carcinoma HT29, lung carcinoma A549 and human embryonic kidney HEK293 cells. Receptor activation, internalization, desensitization and resensitization assays were carried out on these cell lines to define some key functions relevant for investigating inhibitors in subsequent chapters. Chapter 4 reports a PAR2 mutagenesis study designed to identify the location of the binding site on PAR2 for a specific peptide agonist. A homology model of PAR2 based on bovine rhodopsin was used for docking of an agonist ligand, and the docking results were then investigated via two successive rounds of PAR2 mutagenesis in which the effect of each mutation (20 in all) was separately investigated by changes in agonist potency in the intracellular calcium release assay. Five PAR2 mutants showed more than a 5-fold reduction in agonist potency, while three others showed up to a 7-fold reduction. Mutations found to be important for agonist activity were mapped back to the model. Because there was extensive clustering of these key mutated amino acids, it is likely that this study has pinpointed the precise binding site of the agonist peptide in PAR2. Interestingly, this site is within the transmembrane region of the receptor. Chapter 5 reports the design, discovery and development of novel PAR2 agonists and antagonists and their regulatory effects in a diverse array of cell types. Structure-activity relationships were used to examine influences on the first, sixth and seventh positions of a PAR2 agonist peptide. At least five compounds were found herein to be equiopotent with the most potent PAR2 agonist reported. Knowledge obtained from this study was then used to create the first non-peptidic agonists for PAR2. The most potent nonpeptidic agonist (retaining one natural amino acid) was at least equipotent with the best peptide agonists. Conversion to nonpeptidic antagonists proved to be successful and this chapter reports the most potent known nonpeptide antagonist, which was selective for PAR2 and active at low micromolar concentrations. It inhibited intracellular Ca2+ release induced by different PAR2 agonists (trypsin, 2f-LIGRLO-NH2, nonpeptide agonists) in multiple cell lines (HT29, Panc-1, A549, MKN1, MKN45, MDA-MB-231, HUVEC) that have been physiologically associated with PAR2. It also inhibited release of inflammatory cytokines IL-8 and IL-6 and shows antiproliferative activity against primary human cells. The antagonist is competitive, reversible and surmountable (pA2 6.11). This thesis summarizes a large body of work that provides valuable molecular insights to PAR2 regulation, and lays the groundwork for rational design and development of novel nonpeptidic agonists and antagonists of PAR2 as potentially valuable pharmacological probes in vivo and as useful leads to development of therapeutics for inflammatory diseases and cancers.
166

Characterizing the extracellular domains of the relaxin and INSL3 receptors, LGR7 and LGR8

Scott, Daniel James Unknown Date (has links) (PDF)
Relaxin and insulin-like peptide-3 (INSL3) are closely related reproductive hormones that are derived from a common ancestor. Relaxin was initially named for its ability to relax the pubic symphysis in pregnant guinea pigs at parturition. Since then relaxin has been found to be involved in many physiological processes based on its ability to stimulate the breakdown and remodeling of collagen fibers. INSL3 is also known as Leydig insulin-like hormone and the relaxin-like factor, and is produced by the Leydig cells in the testis, the thecal and luteal cells of the ovary, the thyroid, placenta and mammary gland. INSL3 induces transabdominal testicular descent by stimulating the development of the gubernacular ligament, which subsequently swells and contracts to pull the fetal testes towards the inguinal wall. In adults however, evidence suggests that INSL3 is involved in reproductive function, acting to promote the survival of male and female germ cells.
167

Variants A189V et N680S du récepteur humain de l'hormone folliculo-stimulante (FSH) : caractérisation fonctionnelle et implications cliniques / Variants A189V and N680S of the human follicle-stimulating hormone (FSH) receptor : functional characterization and clinical involvement

Tranchant, Thibaud 09 December 2011 (has links)
La FSH est une hormone qui joue un rôle central dans la fonction de reproduction. De ce fait, elle est utilisée en assistance médicale à la procréation (AMP) afin de recruter un pool de follicules et de l’amener jusqu'à l'ovulation. La FSH agit sur un récepteur spécifique (RFSH) qui active des voies de signalisation par l'intermédiaire des protéines G et des β-arrestines. L'étude in vitro d’un mutant et de variants du RFSH décrits chez l’homme nous a permis de mettre en évidence différents mécanismes conduisant à des biais de signalisation de ce récepteur. Ces altérations génétiques, en modifiant l'équilibre qui existe entre les différentes voies de signalisation activées par le RFSH, conduisent à des manifestations cliniques. En parallèle, nous avons mené une étude clinique sur le polymorphisme N680S du RFSH, qui nous a permis de confirmer et de prolonger les résultats de la littérature tout en corrélant les résultats obtenus in vitro à la signalisation des récepteurs N680 et S680. L’ensemble de nos résultats ouvre des perspectives pour le développement de nouvelles stratégies en AMP. / FSH is a hormone which is centrally involved in reproduction. For this reason, FSH is extensively used in in vitro fertilization (IVF) to recruit and lead a pool of follicle to ovulation. FSH acts on its cognate receptor (FSHR) which activates signaling pathways through the canonical G-protein pathways as well as through β-arrestin-dependent transduction mechanisms. In vitro studies of a mutant and of variants of the FSHR identified in patients allowed us to highlight different mechanisms leading to bias in the signaling pathways triggered by this receptor. These genetic alterations, by modifying the equilibrium that exists between the different signaling pathways activated by the FSHR, lead to clinical consequences. In parallel, we have carried out a clinical study centered on the N680S polymorphism of the FSHR. Our results confirm and extend previous studies from the literature while correlating the results we obtained in vitro with the functional consequences of the N680S polymorphism of the FSHR. Together, our results open new avenues for developing new strategies in IVF.
168

Etude de la régulation du répertoire exhaustif des récepteurs couplés aux protéines G murins dans un modèle de différenciation neuronale et adipocytaire. / The comprehensive murin G protein-coupled receptors repertoire studying in neuronal and adipocyte differentiation models

Maurel, Benjamin 13 December 2010 (has links)
Les récepteurs couplés aux protéines G (RCPG) constituent la plus grande famille de récepteurs membranaires. Ils contrôlent de nombreux processus physiologiques et leurs implications dans de nombreuses pathologies fait de cette famille une cible de près de 30% des médicaments. Cependant nombre d'entre eux sont orphelins et/ou possèdent des fonctions inconnues, constituant ainsi un réservoir important de cibles pharmacologiques.Afin de déterminer quelles peuvent être les applications thérapeutiques des RCPG, il faut être capable d'identifier les tissus dans lesquels ils s'expriment et ont des effets biologiques. Pour cela, nous avons développé une approche de PCR quantitative à haut débit permettant l'étude de l'ensemble des gènes codants les RCPG murin.Nous avons tout d'abord recherché tous les endoRCPG de souris et pour chacune des séquences identifiées un couple d'amorces a été synthétisé.Nous avons ensuite utilisé cette banque d'amorces dans deux modèles de différenciation cellulaire. La culture primaire de neurones granulaires du cervelet et une lignée de préadipocytes.Nous nous sommes appuyés sur l'hypothèse suivante : si l'expression d'un RCPG varie au cours d'un phénomène particulier, c'est qu'il est possiblement impliqué dans la régulation de celui-ci.Nous avons donc dans un premier temps identifié les endoRCPG dont l'expression variait au cours de la différenciation cellulaire. Dans un deuxième temps nous avons testé par des approches pharmacologiques et de biologie cellulaire l'implication de ces récepteurs dans la régulation des divers aspects de la différenciation cellulaire des modèles étudiés. / G protein-coupled receptors (GPCRs) constitute the largest family of membrane receptors and control numerous physiological processes. Accordingly, ~45% of drugs used to treat human pathologies target a GPCR. However, a number of GPCRs are orphans and/or involved in unknown functions, which makes GPCRs an attractive reservoir of pharmacological targets.To identify novel therapeutic applications of GPCR ligands, it is necessary to identify organs in which GPCRs are expressed and have biological effects. To that end, we developed a high-throughput, real-time PCR approach to quantify each and every murine GPCR transcripts. We first established a census of all endo-GPCRs, i.e. those GPCRs having an endogenous ligand, encoded in the murine genome. We then designed and validated a primer pair for each identified sequence.We used this primer collection in 2 models of cell differentiation, namely primary cultures of cerebellar granule neuroblasts and the preadipocyte 3T3-L1 cell line. Our basic premise in this approach is that changes in the expression levels of a GPCR in a given biological phenomenon is an indication that this GPCR might be functionally relevant to the biological process under study.We first focused on GPCRs whose expression changed during cellular differentiation, which enabled the identification of candidate GPCRs in this phenomenon. Using pharmacological and genomic tools, we tested the implication of those candidates in various aspects of cell differentiation. In particular, we performed a detailed study of the role of the F2r thrombin receptor and Gprc5a orphan receptor in preadipocyte proliferation.
169

Adenosine signaling in Drosophila / Adenosine signaling in Drosophila

KUČEROVÁ, Lucie January 2013 (has links)
This thesis characterizes adenosine signalization in Drosophila and describes response to adenosine in various cell types. Extracellular adenosine mediates most of its physiological effects through its receptors but recent data also indicate that adenosine transport also has important physiological functions. It was shown in this thesis that adenosine stimulates only cAMP second messenger system in Drosophila cells endogenously expressing AdoR. The pharmacological profile of the DmAdoR was established using the cAMP functional assay. The utility of the agonist 2-chloroadenosine and antagonist SCH58261 were examined in flies in vivo and compared with phenotypes of DmAdoR mutants. The responses of Drosophila cells to adenosine mediated by adenosine transport were also examined. Different cell types exhibited striking differences in adenosine uptake and adenosine recycling that were closely connected with the regulation of carbohydrate and lipid metabolism. This thesis provides an important foundation for the study of interactions between adenosine receptor and adenosine transport.
170

The biology of ELTD1/ADGRL4 : a novel regulator of tumour angiogenesis

Favara, David M. January 2017 (has links)
<strong>Background:</strong> Our laboratory identified ELTD1, an orphan GPCR belonging to the adhesion GPCR family (aGPCR), as a novel regulator of angiogenesis and a potential anti-cancer therapeutic target. ELTD1 is normally expressed in both endothelial cells and vascular smooth muscle cells and expression is significantly increased in the tumour vasculature. The aim of this project was to analyse ELTD1's function in endothelial cells and its role in breast cancer. <strong>Method:</strong> 62 sequenced vertebrate genomes were interrogated for ELTD1 conservation and domain alterations. A phylogenetic timetree was assembled to establish time estimates for ELTD1's evolution. After ELTD1 silencing, mRNA array profiling was performed on primary human umbilical vein endothelial cells (HUVECs) and validated with qPCR and confocal microscopy. ELTD1's signalling was investigated by applying the aGPCR ‘Stinger/tethered-agonist Hypothesis'. For this, truncated forms of ELTD1 and peptides analogous to the proposed tethered agonist region were designed. FRET-based 2<sup>nd</sup> messenger (Cisbio IP-1;cAMP) and luciferase-reporter assays (NFAT; NFÎoB; SRE; SRF-RE; CREB) were performed to establish canonical GPCR activation. To further investigate ELTD1's role in endothelial cells, ELTD1 was stably overexpressed in HUVECS. Functional angiogenesis assays and mRNA array profiling were then performed. To investigate ELTD1 in breast cancer, a panel of cell lines representative of all molecular subtypes were screened using qPCR. Furthermore, an exploratory pilot study was performed on matched primary and regional nodal secondary breast cancers (n=43) which were stained for ELTD1 expression. Staining intensity was then scored and compared with relapse free survival and overall survival. <strong>Results:</strong> ELTD1 arose 435 million years ago (mya) in bony fish and is present in all subsequent vertebrates. ELTD1 has 3 evolutionary variants of which 2 are most common: one variant with 3 EGFs and a variant with 2 EGFs. Additionally, ELTD1 may be ancestral to members of aGPCR family 2. HUVEC mRNA expression profiling after ELTD1 silencing showed upregulation of the mitochondrial citrate transporter SLC25A1, and ACLY which converts cytoplasmic citrate to Acetyl CoA, feeding fatty acid and cholesterol synthesis, and acetylation. A review of lipid droplet (fatty acid and cholesterol) accumulation by confocal microscopy and flow cytometry (FACS) revealed no changes with ELTD1 silencing. Silencing was also shown to affect the Notch pathway (downregulating the Notch ligand JAG1 and target gene HES2; upregulating the Notch ligand DLL4) and inducing KIT, a mediator of haematopoietic (HSC) and endothelial stem cell (ESC) maintenance. Signalling experiments revealed that unlike other aGPCRs, ELTD1 does not couple to any canonical GPCR pathways (Gαi, Gαs, Gαq, Gα12/13). ELTD1 overexpression in HUVECS revealed that ELTD1 induces an endothelial tip cell phenotype by promoting sprouting and capillary formation, inhibiting lumen anastomoses in mature vessels and lowering proliferation rate. There was no effect on wound healing or adhesion to angiogenesis associated matrix components. Gene expression changes following ELTD1 overexpression included upregulation of angiogenesis associated ANTRX1 as well as JAG1 and downregulation of migration associated CCL15 as well as KIT and DLL4. In breast cancer, none of the representative breast cancer cell lines screened expressed ELTD1. ELTD1 breast cancer immunohistochemistry revealed higher levels of vascular ELTD1 staining intensity within the tumour stroma contrasted to normal stroma and expression within tumour epithelial cells. Additionally, ELTD1 expression in tumour vessels was differentially expressed between the primary breast cancer microenvironment and that of the matched regional node. Due to the small size of the pilot study population, survival comparisons between the various subgroups did not yield significant results. <strong>Conclusion:</strong> ELTD1 is a novel regulator of endothelial metabolism through its suppression of ACLY and the related citrate transporter SLC25A1. ELTD1 also represses KIT, which is known to mediate haematopoietic and endothelial progenitors stem cell maintenance, a possible mechanism through which endothelial cells maintain terminal endothelial differentiation. ELTD1 does not signal like other adhesion GPCRS with CTF and FL forms of ELTD1 not signalling canonically. Additionally, ELTD1 regulates various functions of endothelial cell behaviour and function, inducing an endothelial tip cell phenotype and is highly evolutionarily conserved. Lastly, ELTD1 is differentially expressed in tumour vessels between primary breast cancer and regional nodal metastases and is also expressed in a small subset of breast cancer cells in vivo despite no cancer cell lines expressing ELTD1. The pilot study investigating ELTD1 in the primary breast cancer and regional involved nodes will be followed up with a larger study including the investigation of ELTD1 in distant metastases.

Page generated in 0.0414 seconds