• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 265
  • 85
  • 49
  • 43
  • 14
  • 7
  • 6
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 3
  • Tagged with
  • 631
  • 631
  • 267
  • 193
  • 138
  • 66
  • 64
  • 59
  • 52
  • 48
  • 47
  • 46
  • 44
  • 43
  • 43
  • 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.
541

Apelin Regulation of K-Cl Cotransport in Vascular Smooth Muscle Cells.

Sharma, Neelima 11 June 2014 (has links)
No description available.
542

ASPECTS OF AIRWAY STRETCH-ACTIVATED CONTRACTIONS ASSESSED IN PERFUSED INTACT BOVINE BRONCHIAL SEGMENTS

Hernandez, Jeremy M. January 2011 (has links)
<p>Asthma is a disease characterized by transient airway smooth muscle contraction leading to episodes of reversible airway narrowing. It affects over 300 million people worldwide and is implicated in over 250 000 deaths annually. The primary clinical features of asthma include airway inflammation, hyperresponsiveness, and remodeling. Generally, asthmatic patients experience exacerbations between periods of diminished symptoms. Interestingly, in addition to these above mentioned hallmarks, asthmatics have also been shown to react differently to ventilatory mechanical strain. This is most evident when assessing the effect of a deep inspiration (DI), clinically measured as a breath taken from functional residual capacity to total lung capacity, in healthy individuals <em>versus</em> asthmatics. These deep inspiratory efforts have been shown to produce a bronchodilatory response in healthy individuals, whereas in asthmatics, DIs are less effective in producing bronchodilation, can cause more rapid airway re-narrowing, and even bronchoconstriction in moderate to severe asthmatics. The mechanism by which a DI is able to cause bronchoconstriction remains ambiguous. Previous theories suggest that this phenomenon is intrinsic to airway smooth muscle (ASM) itself. However, the airway inflammation present in asthmatic airways may also add to the increased ASM contractility following stretch, by the release of mediators that can prime the contractile apparatus to react excessively in the presence of stretch.</p> <p>Thus, collectively, the studies contained in this thesis are linked to the general theme of greater characterization of the signalling mechanisms that regulate airway stretch-activated contractions using a pharmacological approach in intact bovine bronchial segments, with the hope of providing novel insights into the mechanisms that regulate the DI-induced bronchoconstriction seen in asthmatics.</p> / Doctor of Philosophy (Medical Science)
543

INVOLVEMENT OF SRC TYROSINE KINASE AND CALCIUM-HANDLING IN AIRWAY SMOOTH MUSCLE EXCITATION-CONTRACTION COUPLING

Humber, Brent T. 04 1900 (has links)
<p><strong>Introduction</strong></p> <p>Asthma is a chronic respiratory disease that is becoming more prevalent. Airway hyperresponsivness, a key feature of asthma, involves increased narrowing of the airways in response to bronchoconstricting agents. Airway smooth muscle (ASM) functioning is largely responsible for hyperresponsiveness yet the mechanisms behind excitation-contraction coupling are not fully understood. Src tyrosine kinase contributes to contraction in other smooth muscle types. Furthermore, STIM1, Orai1, IPLA<sub>2</sub>b and RyRs play a role in ASM excitation-contraction coupling.</p> <p><strong>Aim</strong></p> <p>We sought to determine whether Src activity is involved in serotonin (5-HT)- and acetylcholine (ACh)-induced ASM contraction. We also examined whether the gene expression of molecules involved in sarcoplasmic reticulum emptying and refilling is altered during airway hyperresponsiveness.</p> <p><strong>Methods</strong></p> <p>Bovine tracheal ASM strips were pre-treated with the non-specific tyrosine kinase inhibitor genistein (10<sup>-4 </sup>M), src kinase family inhibitors PP1 (10<sup>-5 </sup>M) and PP2 (10<sup>-5 </sup>M) or vehicle and challenged with either 5-HT or ACh to determine the involvment of Src in contraction. Western blotting was used to examine Src activity following 5-HT or ACh treatment. Female BALB/c mice were exposed to an intranasal injection of [1.7mg/ml] HDM extract or saline. Real time, reverse-transcriptase polymerase chain reaction was used to examine gene expression.</p> <p><strong> </strong></p> <p><strong>Results</strong></p> <p>Genistein, PP1 and PP2 significantly reduced 5-HT-induced ASM contractions and Src activity was significantly increased in response to 5-HT. ACh-induced contractions were significantly reduced by genistein, but not PP1 and PP2. However, Src activity was significantly increased by ACh. RyR3 mRNA expression was significantly increased, Orai1 was significantly decreased, and STIM1, IPLA<sub>2</sub>b, RyR1 and RyR2 were unchanged by the house dust mite treatment.</p> <p><strong>Conclusion</strong></p> <p>These data suggets 5-HT-induced ASM contraction involves Src activity. However, ACh-induced ASM contractions might not require Src. The changes in RyR3 and Orai1 expression might alter Ca<sup>2+</sup>-handling in such a way as to potentiate airway hyperresponsiveness but further investigation is required.</p> / Master of Science (MSc)
544

The Regulation of Vascular Wall Cells by a TLR Ligand and Gp130 Cytokines

Schnittker, David L.K. 10 1900 (has links)
<p>Atherosclerosis is a disease affecting the blood vessels that is inflammatory in nature, and plays an important role in cardiovascular disease (CVD), one of the leading causes of morbidity and mortality worldwide. Oncostatin M (OSM), a member of the IL-6/gp130 cytokine family, has been implicated in atherosclerosis both in mouse models and in humans. OSM synergizes with other stimuli in various systems to regulate cells. Infectious pathogens as well as danger associated host molecules stimulate members of the innate immune system, including Toll-like Receptors (TLRs), to respond in a pro-inflammatory manner to cause cell activation and cytokine release. Experiments were performed to determine whether OSM and LPS (a TLR-4 ligand) synergize in regulation of vascular wall cells <em>in vitro</em>.</p> <p>Upon stimulation of Aortic Adventitial Fibroblasts from mice (MAAFs) and humans (HAoAFs) as well as Human Aortic Smooth Muscle Cells (HAoSMCs) with LPS in combination with OSM, it was determined that there was a synergistic increase in IL-6 and VEGF levels in the cell supernatants as measured by ELISA compared to either treatment alone. MAAFs were also able to synergistically express KC upon stimulation with LPS and OSM, while in HAoAFs and HAoSMCs, LPS induced IL-8 levels were supressed by OSM. These effects were unique to OSM among gp130 cytokine members, as treatment of these cells with LPS in combination with LIF, IL-6, IL-31, or IL-11 had no marked effects compared to LPS alone. Furthermore, MCP-1 steady state mRNA levels were elevated 6 hours post stimulation with LPS and OSM compared to either treatment alone in HAoAFs and HAoSMCs.</p> <p>While OSM did not appear to modulate TLR-4 expression, OSM treatment resulted in an increased phosphorylation signal in STAT-1,-3, and -5, as well as Akt in MAAFs and HAoAFs. In addition, combined LPS and OSM stimulation resulted in an increased phosphorylation signal of the MAPK p38 compared to either treatment alone. Furthermore, a neutralizing antibody to the OSMr-β was able to inhibit HAoAF IL-6 responses to PBMC conditioned medium. Together, these findings indicate that OSM and LPS can synergize <em>in vitro </em>to induce the expression of inflammatory factors in vascular wall cells, emphasizing the potential role of OSM, TLR-4 ligands, and adventitial fibroblasts in vascular inflammation.</p> / Master of Science (MSc)
545

THERAPEUTIC MECHANISMS OF INTERLEUKIN-19 FOR VASCULAR PROLIFERATIVE DISEASES

Cuneo, Anthony January 2012 (has links)
Cardiovascular disease is the leading cause of mortality in the western world. The pro-inflammatory and pro-proliferative etiology of vascular proliferative diseases is well characterized, while much less is known about the mechanisms of anti-inflammatory and anti-proliferative processes. Interleukin-19 (IL-19) is a newly described member of the IL-10 family of anti-inflammatory interleukins, and our group was the first to discover IL-19 expression in activated, synthetic, but not quiescent, contractile human vascular smooth muscle cells (hVSMC). We also found that IL-19 is anti-inflammatory and anti-proliferative for hVSMC. IL-19 is able to reduce the abundance of COX-2, IL-1&beta;, IL-8, and Cyclin D1 transcripts which contain AU-rich elements (ARE) in their 3'-untranslated regions (3'-UTR). IL-19 is able to reduce the abundance of HuR, a stabilizing RNA-binding protein, which we feel provides a mechanism for these effects. The overall goal of this study is to elucidate IL-19's anti-inflammatory and anti-proliferative mechanism(s) in hVSMC in the context of vascular proliferative diseases. This goal has directed our overall hypothesis: IL-19's anti-proliferative and anti-inflammatory effects in hVSMC are mediated, at least in part, by modulation of HuR abundance and translocation, resulting in decreased stability of mRNA transcripts. HuR functions through a translocation mechanism, and IL-19 is able to reduce HuR cytoplasmic abundance. IL-19 also reduces HuR phosphorylation, which is a pre-requisite for HuR translocation, possibly through a PKC&alpha;-dependent mechanism. The stability of ARE-containing transcripts is reduced with IL-19 treatment, and reducing HuR expression by siRNA has the same inhibitory effect. VSMC are important mediators in the initiation of atherosclerosis. Oxidized low-density lipoprotein (ox-LDL) is able to induce IL-19 expression in these cells. VSMC are known to express scavenger receptors that take up ox-LDL. IL-19 is able to reduce the uptake of ox-LDL and the abundance of ox-LDL induced LOX-1 and CX-CL16 scavenger receptors. Interestingly, these scavenger receptors also have ARE in their 3'-UTR. IL-19 is able to reduce ox-LDL induced HuR cytoplasmic abundance. HuR knockdown by siRNA reduces the uptake of ox-LDL by hVSMC. These data suggest that IL-19 reduced scavenger receptor abundance may be due to decreased total and cytoplasmic HuR abundance. IL-19 reduces the abundance of ox-LDL induced COX-2 expression. Taken together, these results demonstrate that IL-19 down-regulates vital steps in vascular proliferative disease processes through an HuR-dependent mechanism. / Molecular and Cellular Physiology
546

Keratose Hydrogels Promote Vascular Smooth Muscle Differentiation from c-kit+ Human Cardiac Stem Cells: Underlying Mechanism and Therapeutic Potential

Ledford, Benjamin 23 March 2018 (has links)
Cardiovascular disease is the leading cause of death in the United States, and coronary artery disease (CAD) kills over 370,000 people annually. There are available therapies that offer a temporary solution; however, only a heart transplant can fully resolve heart failure, and donor organ shortages severely limit this therapy. C-kit+ human cardiac stem cells (hCSCs) offers a viable alternative therapy to treat cardiovascular disease by replacing damaged cardiac tissue; however, low cell viability, low retention/engraftment, and uncontrollable in vivo differentiation after transplantation has limited the efficacy of stem cell therapy. Tissue engineering solutions offer potential tools to overcome current limitations of stem cell therapy. Materials derived from natural sources such as keratin from human hair offers innate cellular compatibility, bioactivity, and low immunogenicity. Keratin proteins extracted using oxidative chemistry known as keratose (KOS) have shown therapeutic potential in a wide range of applications including cardiac regeneration. My studies utilize KOS hydrogels to modulate c-kit+ hCSC differentiation, and explore the capability of differentiated cells to regenerate vascular tissue. In the first Chapter, we reviewed literature relevant to keratin-based biomaterials and their biomedical applications, the use of stem cells in cardiovascular research, and the differentiation of vascular smooth muscle cells (VSMCs). The section on biomedical applications of keratin biomaterials focuses on the oxidized form of keratin known as keratose (KOS), because this was the material used for our research. Since we planned to use this material in conjunction with c-kit+ hCSCs, we also briefly explored the use of stem cells in cardiovascular research. Additionally, we examined some key signaling pathways, developmental origins, and the cell phenotype of VSMCs for reasons that will become clear after observing results from chapters 2 and 3. Based upon our review of the literature, KOS biomaterials and c-kit+ hCSCs were determined to be promising as a combined therapeutic for the regeneration of cardiac tissue. Research in Chapter 2 focused on characterizing the effects of KOS hydrogel on c-kit+ hCSC cell viability, proliferation, morphology, and differentiation. Results demonstrated that KOS hydrogels could maintain hCSC viability without any observable toxic effects, but it modulated cell size, proliferation, and differentiation compared to standard tissue culture polystyrene cell culture (TCPS). KOS hydrogel produced gene and protein expression consistent with a VSMC phenotype. Further, we also observed novel "endothelial cell tube-like" microstructures formed by differentiated VSMCs only on KOS hydrogel, suggesting a potential capability of the hCSC-derived VSMCs for in vitro angiogenesis. Results from this study lead us to question what signaling pathways might be responsible for the apparent VSMC differentiation pattern we observed on KOS hydrogels. Research in Chapter 3 explored the time course of VSMC differentiation, cell contractility, inhibition of VSMC differentiation, and measured protein expression of transforming growth factor beta 1 (TGF-β1) and its associated peptides for hCSCs cultured on KOS hydrogels, tissue culture polystyrene, and collagen hydrogels. A review of VSMC differentiation signaling pathways informed our decision to investigate the role of TGF-β1 in VSMC differentiation. Results demonstrated that KOS hydrogel differentiated hCSCs significantly increased expression for all three vascular smooth muscle (VSM) markers compared to TCPS differentiated cells. Additionally, KOS differentiated hCSCs were significantly more contractile than cells differentiated on TCPS. Recombinant human (rh) TGF-β1 was able to induce VSM differentiation on TCPS. VSM differentiation was successfully inhibited using TGF-β NABs and A83-01. Enzyme-Linked Immunosorbent Assay (ELISA) analysis revealed that both TCPS and KOS hydrogel differentiated cells produced TGF-β1, with higher levels being measured at early time points on TCPS and later time points on KOS hydrogels. Results from supplementing rhTGF-β1 to TCPS and KOS hydrogels revealed that KOS seems to interact with TGF-β to a greater extent than TCPS. Western blot results revealed that latency TGFβ binding protein (LTBP-1) and latency associated peptide (LAP) had elevated levels early during differentiation. Further, the levels of LTBP-1 and LAP were higher on KOS differentiated hCSCs than TCPS hCSCs. This study reaffirms previous results of a VSM phenotype observed on KOS hydrogels, and provides convincing evidence for TGF-β1 inducing VSM differentiation on KOS hydrogels. Additionally, results from ELISA and western blot provide evidence that KOS plays a direct role in this pathway via interactions with TGF-β]1 and its associated proteins LTBP-1 and LAP. Results from chapter 2 and 3 offered significant evidence that our cells exhibited a VSMC phenotype, and that TGF-β1 signaling was a key contributor for the observed phenotype, but we still needed an animal model to explore the therapeutic potential of our putative VSMCs. Research in Chapter 4 investigated a disease model to test the ability of KOS hydrogel differentiated cells to regenerate vascular tissue. To measure vascular regenerative capability, we selected a murine model of critical limb ischemia (CLI). CLI was induced in 3 groups (n=15/group) of adult mixed gender NSG mice by excising the femoral artery and vein, and then treated the mice with either PBS (termed as PBS-treated), Cells differentiated on TCPS (termed as Cells from TCPS), or KOS hydrogel-derived VSMCs (termed as Cells from KOS). Blood perfusion of the hind limbs was measured immediately before and after surgery, then 14, and 28 days after surgery using Laser Doppler analysis. Tissue vascularization, cell engraftment, and skeletal muscle regeneration were measured using immunohistochemistry, 1,1'-Dioctadecyl3,3,3',3'-Tetramethylindocarbocyanine Perchlorate (DiL) vessel painting, and hematoxylin and eosin (HandE) pathohistological staining. During the 4-week period, both cell treatment groups showed significant increases in blood perfusion compared to the PBS-treated control, and at day 28 the Cells from KOS group had significantly better blood flow than the Cells from TCPS group. Additionally, the Cells from KOS group demonstrated a significant increase in the ratio of DiL positive vessels, capillary density, and a greater density of small diameter arterioles compared to the PBS-treated group. Further, both cell-treated groups had similar levels of engraftment into the host tissue. We conclude that Cells from KOS therapy increases blood perfusion in an NSG model of CLI, but does not lead to increased cell engraftment compared to other cell based therapies. Overall, the results from this dissertation demonstrated that KOS hydrogels produce VSMC differentiation from c-kit+ hCSCs mediated by TGF-β1 signaling, and that the differentiated cells are able to increase blood perfusion in a CLI model by increasing capillary density, suggesting enhanced angiogenesis. Future studies should explore potential protein-protein interactions between KOS, TGF-β1 and its associated proteins. Additionally, we should plan animal studies that examine the efficacy of our cells to regenerate cardiac tissue following an acute myocardial infarction (AMI). / PHD
547

Semi-analytical Investigation on the Transmural Alignment of Vascular Smooth Muscle Cells

Wollner, Maximilian Peter 11 April 2024 (has links)
The apoptosis and dysfunction of vascular smooth muscle cells in the human descending thoracic aorta is often associated with cardiovascular diseases like aortic dissection and aneurysms. Knowledge of the mechanical effects of contractile smooth muscle cells plays a crucial role in the understanding these potentially lethal conditions. Located in the medial layer, vascular smooth muscle cells are arranged in the so-called herringbone pattern. In regards to the mechanics of the aorta, the consequences of this type of anisotropy have not been fully discussed in literature so far. In this end, a novel hyperelastic constitutive law is proposed which accounts for the dispersive, transmural alignment of vascular smooth muscle cells and their characteristic length-tension behaviour. The model is calibrated with experimental data and is applied to the simulation of an aortic ring under in vivo conditions. By approximating the geometry of the aorta as a layered, thick-walled cylinder, the corresponding quasistatic, mechanical boundary value problem is solved semi-analytically. It is shown that the herringbone pattern induces shear deformation and equalises the normal stress gradients in the aortic wall. Since arterial vessels are able to actively adapt and alter the alignment and activity of smooth muscle cells, the existence of the herringbone pattern is in accordance with Fung's principle of optimal operation.
548

Autoregulation of the Human Cerebrovasculature by Neurovascular Coupling

Farr, Hannah Abigail January 2013 (has links)
Functional hyperaemia is an important mechanism by which increased neuronal activity is matched by a rapid and regional increase in blood supply. This mechanism is facilitated by a process known as “neurovascular coupling” – the orchestrated communication system involving the cells that comprise the neurovascular unit (neurons, astrocytes and the smooth muscle and endothelial cells lining arterioles). Blood flow regulation and neurovascular coupling are altered in several pathological states including hypertension, diabetes, Alzheimer’s disease, cortical spreading depression and stroke. By adapting and extending other models found in the literature, we create, for the first time, a mathematical model of the entire neurovascular unit that is capable of simulating two separate neurovascular coupling mechanisms: a potassium- and EET-based and a NO-based mechanism. These models successfully account for several observations seen in experiment. The potassium/EET-based mechanism can achieve arteriolar dilations similar in magnitude (3%) to those observed during a 60-second neuronal activation (modelled as a release of potassium and glutamate into the synaptic cleft). This model also successfully emulates the paradoxical experimental finding that vasoconstriction follows vasodilation when the astrocytic calcium concentration (or perivascular potassium concentration) is increased further. We suggest that the interaction of the changing smooth muscle cell membrane potential and the changing potassium-dependent resting potential of the inwardly rectifying potassium channel are responsible for this effect. Furthermore, our simulations demonstrate that the arteriolar behaviour is profoundly affected by depolarization of the astrocytic cell membrane, and by changes in the rate of perivascular potassium clearance or the volume ratio between the perivascular space and astrocyte. In the modelled NO-based neurovascular coupling mechanism, NO exerts its vasodilatory effects via neuronal and endothelial cell sources. With both sources included, the model achieves a 1% dilation due to a 60-second neuronal activation. When the endothelial contribution to NO production is omitted, the arteriole is more constricted at baseline. Without the endothelial NO contribution, the arteriolar change in diameter during neuronal activity is greater (6%). We hypothesize that NO has a dual purpose in neurovascular coupling: 1) it dixxxvi rectly mediates neurovascular coupling through release by neuronal sources, and 2) it indirectly modulates the size of the neurovascular coupling response by determining the baseline tone. Our physiological models of neurovascular coupling have allowed us to replicate, and explain, some of the phenomena seen in both neurovascular coupling-oriented and clinicallyoriented experimental research. This project highlights the fact that physiological modelling can be used as a tool to understand biological processes in a way that physical experiment cannot always do, and most importantly, can help to elucidate the cellular processes that induce or accompany our most debilitating diseases.
549

Modulation de l'expression de Sirt-1 induite par l'endothéline-1 dans les cellules musculaires lisses vasculaires

Mir, Ahmed 08 1900 (has links)
Au cours des maladies cardiovasculaires (MCV), il peut se produire divers problèmes de santé, telle que l’insuffisance cardiaque ou encore l’HTA. Ces phénomènes se caractérisent, entre autres, par une augmentation de synthèse d’endotheline-1 (ET-1), un neuropeptide synthétisé par les cellules endothéliales ayant un effet vasoconstricteur sur les cellules musculaires lisses vasculaires (CMLV). Ainsi, la surexpression de ce vasopeptide, mène à terme, au maintien de l’HTA aggravée des sujets, précédée ou concomitante à l’athérosclérose ou à la resténose, cliniquement illustrées par une prolifération et une migration anormale des CMLV de la media vers l’intima des vaisseaux sanguins. Parallèlement, il a été observé que la protéine sirtuine-1 (Sirt-1), membre de la famille des protéines histones déacétylases (HDAC), présente des propriétés anti-athérosclérotiques par sa capacité d’atténuer la prolifération et la migration des CMLV. Des travaux récents ont aussi montré qu’au cours de l’HTA la protéine Sirt-1 est faiblement exprimée dans les CMLV. Son implication dans le développement des pathologies vasculaires semble apparente, mais des études demeurent nécessaires pour décrire son rôle exact dans la pathogenèse des MCV. Dans cette optique, l’objectif de cette étude a été d’observer la variation d’expression de Sirt-1 dans les CMLV, isolées de l’aorte ascendante de rat, en réponse à l’ET-1. On a remarqué qu’une heure de stimulation des CMLV avec l’ET-1 induit une diminution de l’expression de Sirt-1 via l’activation des récepteurs ETA. Ces résultats suggèrent que la capacité d’ET-1 à atténuer l’expression de Sirt-1 serait un éventuel mécanisme d’action avec des effets favorisant les MCV. / Cardiovascular diseases (CVD) are associated with several vascular dysfunctions such as heart failure and hypertension. These phenomena cause increased synthesis of endothelin-1 (ET-1), a neuropeptide, synthesized by endothelial cells which has vasoconstrictor action on vascular smooth muscle cells (VSMC). Overexpression of this vasopeptide leads eventually to hypertension (HTA). This usually happen after atherosclerosis or restenosis, leading to proliferation and migration of VSMC from media to intima. It was shown that during atherosclerosis, the protein sirtuin-1 (Sirt-1), a member of protein histone deacetylases (HDAC), has an anti-atherosclerotic effect due to its ability to diminish proliferation and migration of VSMC. It has also been observed that during hypertension, Sirt-1 was poorly expressed in VSMC. Its role in vascular pathophysiology remains sparsely studied, therefore it’s essential to explore it. In the present study we investigated the expression of Sirt-1 in VSMC isolated from the ascending aorta of rats, in response to ET-1 stimulation. We observed that Sirt-1 expression decreases after 1 hour of stimulation by ET-1 via ETA receptors. In summary, these results suggest that the ability of ET-1 to attenuate Sirt-1 expression in VSMC, may be a potential mechanism for promoting CVD.
550

Contribution différentielle du tissu adipeux mâle et femelle dans l’établissement du diabète de type 2 et des altérations cardiovasculaires : rôle de l’apport lipidique

El Akoum, Souhad 09 1900 (has links)
Au cours des dernières années, il est devenu évident que les sociétés des pays industrialisés sont à haut risque de maladies métaboliques. Une alimentation riche en énergie (lipide/glucide), combinée à une sédentarité accrue, est un facteur environnemental contribuant à l'augmentation de la prévalence de maladies reliées spécifiquement à des troubles endocriniens comme l'obésité et le diabète. Le traitement de ces désordres métaboliques doit donc passer par la connaissance et la compréhension des mécanismes moléculaires qui contrôlent ces désordres et le développement de traitements ciblés vers les facteurs responsables. Le tissu adipeux est une glande endocrine qui sécrète des substances, regroupées sous le terme d'adipokines, qui contrôlent l'homéostasie énergétique. L'augmentation de la masse adipeuse est responsable du développement de dérégulation hormonale qui mène à des dysfonctions physiologiques et métaboliques. Pour contrecarrer le développement démesuré du tissu adipeux, la signalisation insulinique ainsi que l’apport énergétique, responsables de la différenciation adipocytaire, doivent être inhibés. In vivo, la leptine, adipokine dont la concentration est corrélée à la masse adipeuse, présente des actions pro ou anti-insuliniques dans l’organisme pour réguler ce phénomène. Elle favorise l’effet inhibiteur de l’insuline sur la synthèse hépatique de glucose alors qu’elle s’oppose à son action sur l’expression des enzymes glucokinase et phosphoénol-pyruvate carboxykinase. La leptine influence aussi le taux circulant de triglycérides en diminuant sa concentration plasmatique. D'autre part, l'adiponectine, adipokine insulino- sensibilisante, voit sa sécrétion diminuée avec la prise de poids. La sensibilité à l'insuline est ainsi diminuée au fur et à mesure que le débalancement de ces deux adipokines s'accentue. La résistance à l'insuline s'installe alors pour s'opposer au stockage énergétique et à la prise illimitée de poids et la glycémie augmente. L'augmentation du glucose sanguin stimule la sécrétion d'insuline au niveau des cellules pancréatiques. C'est le diabète caractérisé par une hyperglycémie et une résistance à l'insuline. Le diabète, une des premières causes de mortalité dans le monde, est plus répandu sous sa forme non insulinodépendante (diabète de type 2, DT2) liée à l'obésité. Récemment, différents facteurs de transcription ont été identifiés comme régulateurs de l'expression d'une panoplie de gènes impliqués dans le métabolisme glucidique et lipidique. Parmi eux, les récepteurs des inducteurs de la prolifération des peroxysomes (PPAR, Peroxisome Proliferator-Activated Receptor), appartenant à la famille des récepteurs nucléaires. Les PPAR ont été démontrés comme ayant un rôle central dans le contrôle de la transcription des gènes codants pour des protéines impliquées dans le métabolisme : les adipokines. PPARg, en plus de son implication dans le contrôle de l'homéostasie glucidique et lipidique, est reconnu comme étant un facteur de transcription pivot régulant l'adipogenèse du fait de son expression majeure dans le tissu adipeux. D'autre part, il est bien établi maintenant que l'obésité et le diabète sont des facteurs contribuant au développement du processus inflammatoire vasculaire caractéristique de l’athérosclérose. En effet, les cellules endothéliales et musculaires lisses, principales composantes de la média de l’artère, sont très sensibles aux altérations métaboliques. Une diminution de la sensibilité à l’insuline entraine une réduction de la disponibilité du glucose et l’utilisation des acides gras comme alternatif par ces cellules. Ceci induit l’accumulation des acides gras oxydés dans l’intima et leur filtration dans la média pour former un core lipidique. Bien que l’induction de la dysfonction endothéliale soit impliquée très précocement, certaines études pointent l’accumulation lipidique dans les cellules musculaires lisses vasculaires (CML) et leur dysfonction comme déclencheurs de l’athérosclérose. Ce travail visait donc, dans un premier temps, à développer un modèle d'altérations métaboliques liées à la modulation de l'activité du tissu adipeux via une alimentation riche en lipides. Dans un second temps, cette étude tentait d'évaluer l’impact des adipocytes de souris sur les CML vasculaires et sur la modulation de leurs fonctions dans ce modèle d'altérations métaboliques et DT2 liés à l'alimentation et à l'obésité. Ainsi, par le biais de deux diètes pauvres en cholestérol à profil lipidique différent, nous avons développé un modèle murin présentant divers stades d'altérations du métabolisme allant jusqu'au DT2 en lien avec l'obésité chez les mâles et chez les femelles. D’autre part, des signes de cardiomyopathie ainsi qu’une modulation du taux des adipokines sont reliés à ces mêmes diètes. Parallèlement, l’activité de PPAR!2 est modulée chez les souris sous diètes enrichies en gras. Ensuite, nous avons démontré que les adipocytes, provenant de souris alimentées avec une diète enrichie en gras, modulaient la migration et la prolifération des CML comparativement au groupe contrôle. Ces modulations dépendaient en grande partie de la nature de la diète consommée, mais également du sexe de la souris. Par ailleurs, les altérations fonctionnelles des CML, couplées à des modulations géniques, sont associées aux changements du profil de sécrétion des adipokines mesurées chez les adipocytes. L’ensemble de ces travaux suggère une action directe de la nature de la stimulation du tissu adipeux blanc dans la modulation du profil de sécrétion des adipokines et l'induction du DT2 in vivo. Ces altérations de la physiologie adipocytaire se reflètent in vitro où le tissu adipeux contribue aux altérations physiopathologiques des CML liées au DT2. Ainsi, cette étude est l'une des premières à établir un lien direct entre les modulations adipocytaires et les effets de leurs sécrétions sur la physiologie des CML. Ces observations peuvent être exploitées cliniquement dans un développement futur d’outils thérapeutiques visant à prévenir et à traiter les troubles métaboliques et le DT2, en ciblant le tissu adipeux comme entité métabolique et endocrine. / Obesity is recognized as a risk factor to a variety of chronic diseases linked to the metabolic syndrome like atherosclerosis and type 2 diabetes (T2D), and is a major cause of increased risk of morbidity and mortality worldwide. High fat diets (HFD) coupled with sedentarity in the industrialized societies contribute to the raise of metabolic alterations prevalence specifically linked to endocrine troubles. Treatment of these latter should include the comprehension of the molecular mechanisms underlying these disorders in order to appropriately target factors responsible for the disease establishment. Adipose tissue is no longer considered as a passive organ which only stores lipids, but also works as an active gland that secretes several bioactive substances called adipokines. Among them, there are key factors known to play a pivotal role in the regulation of glucose and lipid homeostasis, lipid storage, adipogenesis. They are also recognized for their control of a wide range of cell type like adipocytes, hepatocytes and skeletal myocytes. Accumulation of adipose tissue in obesity, linked with the type as much as the amount of dietary lipids, is due to hyperplasia and hypertrophy of adipocytes. These changes are associated with modification in their secretion and inflammatory profile. To counteract excessive fat tissue development, insulin signalling known for its role in adipogenesis is inhibited. Thus, leptin is secreted by adipocytes to inhibit insulin action and the insulin sensitizer adipokine, adiponectin, is down regulated. The two factors are correlated to weight gain and their respective secretion profile is upregulated for leptin and down regulated for adiponectin. Insulin resistance is developed to prevent energetic storage and unlimited weight gain but glycemic control fails and glycaemia raises. Hyperglycaemia stimulates more insulin secretion, a characteristic of T2D linked to obesity. An estimated 80% of those who develop T2D are obese. Obesity induces important and complex changes, not only in glycemic homeostasis but also in the adipocytes. Following fatty acids (FA) stimulation, the main ligand-activated transcriptional factor that controls adipose tissue metabolism and adipokine secretion, peroxisome proliferator-activated receptor gamma (PPARg), is activated. This nuclear receptor subtype regroups two isoforms: PPARg1, expressed in many tissues (adipose tissue, muscle, heart and liver) where it controls glucose and lipid homeostasis, and PPARg2, the adipocyte’s specific form, which further governs preadipocyte differentiation, up-regulation of genes involved in lipogenesis and the expression of adipokines. Recent advances showed that increased FA and glycaemia trigger vascular alterations that lead to atherosclerosis. In fact, endothelial cells (EC) and smooth muscle cells (SMC), the main arterial components, are sensitive to metabolic alterations. A lack in insulin sensitivity, leading to lower glucose availability, forces arterial cells to use FA as alternative energy source. Thus, in atheroprone regions susceptible to plaque formation, EC and SMC are subjected to metabolic modifications that lead to oxidized low-density lipoprotein (oxLDL) accumulation in the intima and the progression of vascular disease. Many studies confirmed that the presence of SMC in the atherosclerotic plaque originates from the vascular wall but are showing a distinct phenotype. Even if the role of these cells in atherogenesis is not clear, trans-differentiation of SMC into foam cells has been reported in vitro. Thus, the present study aims at studying a HFD-induced obesity mouse model, developed to evaluate the impact of FA nature on the adipokine secretion profile of adipocytes. We also intended to determine gender-specific impact on modulation of metabolic disorders in response to those diets. On the other hand, we aim to determine the role of adipocytes in the development of obesity-linked atherosclerosis. For that, the second part of this study targeted the effect of adipocytes isolated from mice fed with HFD on SMC physiology. We focused our investigation on the effects of adipocytes regardless of the impact of other cell types in the adipose tissue. To reach our goal, we developed a HFD-fed mouse preparation demonstrating different stages of metabolic disorders leading to T2D. This model allowed us to generate adipocytes with different alteration status, reflected by the modulation of their adipokine secretion profile. Modifications in adipokine secretions were associated with PPAR!2 modulation. These results, reported in both genders, were delayed in female who expressed higher levels of estrogen receptor alpha (ER"). Then, the adipocytes were used to produce conditioned cultured media. To decipher the mechanistic contributions of HFD in adipokines modulation, the potential of adipocytes to induce SMC pathophysiologic disorders was evaluated in SMC stimulated by conditioned cultured media. This protocol enables the transposition of diet-induced fat cell modifications into extended alterations in the physiology of vascular SMC. These results strongly support pro-atherogenic effects of abdominal adipocytes on an important vascular component function through paracrine actions. Thus, adipocytes can be recognized as a link between the pathogenic potential of obesity and the impairments of SMC functions. A better understanding of the pathogenic effects of the adipose tissue on other tissues and organ systems might assist to develop better strategies in treating obesity- induced cardiovascular disease and metabolic syndrome.

Page generated in 0.2362 seconds