24 research outputs found

    SASH1, un nouveau lien potentiel entre le tabagisme et l'athérosclerose

    No full text
    Atherosclerosis is characterized by lipids accumulation in medium and big size arteries, as the result of a complex series of mechanisms leading to chronic inflammation and accumulation of macrophage-derived foam cells in the intimal space of the vessels leading to atherosclerotic plaque formation. Rupture of the plaque can lead to life threatening events, such as myocardial infarction and stroke. Our scientific work is in the frame of the Gutenberg Health Study, a population based study in the region of Mainz in Germany, which goal is to identify new biological markers and therapeutic targets, with a particular focus on atherosclerosis. These previous studies have shown through transcriptomic analyses that a number of gene expression were correlated to both smoking and atherosclerosis, opening new perspective to better characterize mechanisms linking smoking to atherosclerosis. Among those genes SASH1, a tumor suppressor was the most correlated to smoking and was also correlated to plaques. Another gene of interest, SLC39A8 showed the strongest correlation to plaques. This thesis project aimed at exploring the role of the tumor suppressor SASH1 and the metallic ion transporter in vitro to determine the cellular and molecular mechanisms by which they could affect plaque formation during atherosclerosis...L’athĂ©rosclĂ©rose est caractĂ©risĂ©e par l’accumulation de lipides dans les artĂšres de gros et moyen calibre. Cette accumulation est due Ă  une sĂ©rie de mĂ©canismes complexes aboutissant a une rĂ©action inflammatoire chronique et l’accumulation de cellules spumeuse dans l’espace neointimale de la paroi vasculaire. Les complications liĂ©es Ă  cette pathologie peuvent entraĂźner des Ă©vĂ©nements vasculaires graves, tels que l’infarctus du myocarde ou les accidents vasculaires cĂ©rĂ©braux. Nos travaux de recherches s’inscrivent dans le cadre de la Gutenberg Health Study, une Ă©tude de population dans la rĂ©gion de Mayence (Mainz) en Allemagne, dont le but est d’identifier de nouveaux marqueurs biologiques et cibles thĂ©rapeutiques liĂ©es aux maladies cardiovasculaires, avec un accent particulier sur l’athĂ©rosclĂ©rose. Nos prĂ©cĂ©dents travaux ont dĂ©montrĂ© que l’expression de certains gĂšnes dans les monocytes circulants Ă©tait corrĂ©lĂ©e Ă  la fois au tabagisme et Ă  l’athĂ©rosclĂ©rose, ouvrant ainsi de nouvelles perspectives pour expliquer les mĂ©canismes par lesquels le tabagisme accĂ©lĂšre la formation des plaques d’athĂ©rosclĂ©rose. Parmi ces gĂšnes, SASH1, un gĂšne suppresseur de tumeur Ă©tait le plus corrĂ©lĂ© au tabagisme, tout en Ă©tant Ă©galement corrĂ©lĂ© au nombre de plaques. Un autre gĂšne, SLC39A8, montrait la plus forte corrĂ©lation avec le nombre de plaque. Mon travail de thĂšse a consistĂ© Ă  explorer le rĂŽle de SASH1, un gĂšne suppresseur de tumeur, et SLC39A8, un symporteur HCO3-/ion mĂ©tallique divalent, in vitro pour tenter de dĂ©terminer par quels mĂ©canismes cellulaires et molĂ©culaires ils pouvaient affecter la formation de la plaque d’athĂ©rosclĂ©rose. L’étude sur SASH1 porte en particulier sur la paroi vasculaire oĂč SASH1 a Ă©tĂ© dĂ©tectĂ©e dans toutes les cellules prĂ©sentes (cellules endothĂ©liales, cellules musculaires lisses, monocytes et macrophages). De plus, des mesures en RT-qPCR ont montrĂ© que l’expression de SASH1 Ă©tait plus Ă©levĂ©e dans les carotides de fumeurs que dans celles des non-fumeurs et ex-fumeurs, confirmant ainsi les observations dĂ©jĂ  faites dans les monocytes circulants humains..

    Factor XII-Driven Inflammatory Reactions with Implications for Anaphylaxis

    No full text
    Anaphylaxis is a life-threatening allergic reaction. It is triggered by the release of pro-inflammatory cytokines and mediators from mast cells and basophils in response to immunologic or non-immunologic mechanisms. Mediators that are released upon mast cell activation include the highly sulfated polysaccharide and inorganic polymer heparin and polyphosphate (polyP), respectively. Heparin and polyP supply a negative surface for factor XII (FXII) activation, a serine protease that drives contact system-mediated coagulation and inflammation. Activation of the FXII substrate plasma kallikrein leads to further activation of zymogen FXII and triggers the pro-inflammatory kallikrein–kinin system that results in the release of the mediator bradykinin (BK). The severity of anaphylaxis is correlated with the intensity of contact system activation, the magnitude of mast cell activation, and BK formation. The main inhibitor of the complement system, C1 esterase inhibitor, potently interferes with FXII activity, indicating a meaningful cross-link between complement and kallikrein–kinin systems. Deficiency in a functional C1 esterase inhibitor leads to a severe swelling disorder called hereditary angioedema (HAE). The significance of FXII in these disorders highlights the importance of studying how these processes are integrated and can be therapeutically targeted. In this review, we focus on how FXII integrates with inflammation and the complement system to cause anaphylaxis and HAE as well as highlight current diagnosis and treatments of BK-related diseases

    The Tumor Suppressor SASH1 Interacts With the Signal Adaptor CRKL to Inhibit Epithelial–Mesenchymal Transition and Metastasis in Colorectal CancerSummary

    Get PDF
    Background & Aims: The tumor-suppressor sterile α motif– and Src-homology 3–domain containing 1 (SASH1) has clinical relevance in colorectal carcinoma and is associated specifically with metachronous metastasis. We sought to identify the molecular mechanisms linking decreased SASH1 expression with distant metastasis formation. Methods: SASH1-deficient, SASH1-depleted, or SASH1-overexpressing HCT116 colon cancer cells were generated by the Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated 9-method, RNA interference, and transient plasmid transfection, respectively. Epithelial-mesenchymal transition (EMT) was analyzed by quantitative reverse-transcription polymerase chain reaction, immunoblotting, immunofluorescence microscopy, migration/invasion assays, and 3-dimensional cell culture. Yeast 2-hybrid assays and co-immunoprecipitation/mass-spectrometry showed V-Crk avian sarcoma virus CT10 oncogene homolog-like (CRKL) as a novel interaction partner of SASH1, further confirmed by domain mapping, site-directed mutagenesis, co-immunoprecipitation, and dynamic mass redistribution assays. CRKL-deficient cells were generated in parental or SASH1-deficient cells. Metastatic capacity was analyzed with an orthotopic mouse model. Expression and significance of SASH1 and CRKL for survival and response to chemotherapy was assessed in patient samples from our department and The Cancer Genome Atlas data set. Results: SASH1 expression is down-regulated during cytokine-induced EMT in cell lines from colorectal, pancreatic, or hepatocellular cancer, mediated by the putative SASH1 promoter. Deficiency or knock-down of SASH1 induces EMT, leading to an aggressive, invasive phenotype with increased chemoresistance. SASH1 counteracts EMT through interaction with the oncoprotein CRKL, inhibiting CRKL-mediated activation of SRC kinase, which is crucially required for EMT. SASH1-deficient cells form significantly more metastases in vivo, depending entirely on CRKL. Patient tumor samples show significantly decreased SASH1 and increased CRKL expression, associated with significantly decreased overall survival. Patients with increased CRKL expression show significantly worse response to adjuvant chemotherapy. Conclusions: We propose SASH1 as an inhibitor of CRKL-mediated SRC signaling, introducing a potentially druggable mechanism counteracting chemoresistance and metastasis formation. Keywords: EMT, Tumor Suppressor, SRC-Kinase, Chemoresistanc

    Role of lipid phosphate phosphatase 3 in human aortic endothelial cell function

    No full text
    International audienceAims: Lipid phosphate phosphatase 3 (LPP3; PPAP2B) is a transmembrane protein dephosphorylating and thereby terminating signalling of lipid substrates including lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P). Human LPP3 possesses a cell adhesion motif that allows interaction with integrins. A polymorphism (rs17114036) in PPAP2B is associated with coronary artery disease, which prompted us to investigate the possible role of LPP3 in human endothelial dysfunction, a condition promoting atherosclerosis. Methods and results: To study the role of LPP3 in endothelial cells we used human primary aortic endothelial cells (HAECs) in which LPP3 was silenced or overexpressed using either wild type or mutated cDNA constructs. LPP3 silencing in HAECs enhanced secretion of inflammatory cytokines, leukocyte adhesion, cell survival and migration and impaired angiogenesis, whereas wild-type LPP3 overexpression reversed these effects and induced apoptosis. We also demonstrated that LPP3 expression was negatively correlated with VEGF expression. Mutations in either the catalytic or the RGD domains impaired endothelial cell function and pharmacological inhibition of S1P or LPA restored it. LPA was not secreted in HAECs under silencing or overexpressing 2 LPP3. However the intra-and extracellular levels of S1P tended to be correlated with LPP3 expression, indicating that S1P is probably degraded by LPP3. Conclusions: We demonstrated that LPP3 is a negative regulator of inflammatory cytokines, leukocyte adhesion, cell survival and migration in HAECs, suggesting a protective role of LPP3 against endothelial dysfunction in humans. Both the catalytic and the RGD functional domains were involved and S1P, but not LPA, might be the endogenous substrate of LPP3

    Role of lipid phosphate phosphatase 3 in human aortic endothelial cell function.

    No full text
    AIMS: Lipid phosphate phosphatase 3; type 2 phosphatidic acid phosphatase ÎČ (LPP3; PPAP2B) is a transmembrane protein dephosphorylating and thereby terminating signalling of lipid substrates including lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P). Human LPP3 possesses a cell adhesion motif that allows interaction with integrins. A polymorphism (rs17114036) in PPAP2B is associated with coronary artery disease, which prompted us to investigate the possible role of LPP3 in human endothelial dysfunction, a condition promoting atherosclerosis. METHODS AND RESULTS: To study the role of LPP3 in endothelial cells we used human primary aortic endothelial cells (HAECs) in which LPP3 was silenced or overexpressed using either wild type or mutated cDNA constructs. LPP3 silencing in HAECs enhanced secretion of inflammatory cytokines, leucocyte adhesion, cell survival, and migration and impaired angiogenesis, whereas wild-type LPP3 overexpression reversed these effects and induced apoptosis. We also demonstrated that LPP3 expression was negatively correlated with vascular endothelial growth factor expression. Mutations in either the catalytic or the arginine-glycine-aspartate (RGD) domains impaired endothelial cell function and pharmacological inhibition of S1P or LPA restored it. LPA was not secreted in HAECs under silencing or overexpressing LPP3. However, the intra- and extra-cellular levels of S1P tended to be correlated with LPP3 expression, indicating that S1P is probably degraded by LPP3. CONCLUSIONS: We demonstrated that LPP3 is a negative regulator of inflammatory cytokines, leucocyte adhesion, cell survival, and migration in HAECs, suggesting a protective role of LPP3 against endothelial dysfunction in humans. Both the catalytic and the RGD functional domains were involved and S1P, but not LPA, might be the endogenous substrate of LPP3

    SASH1, a new potential link between smoking and atherosclerosis

    No full text
    International audienceObjective: We have previously reported that SASH1 expression is increased in circulating human monocytes from smokers and was positively correlated with the number of carotid atherosclerotic plaques. The aim of this study was to further validate the link between smoking, SASH1 and atherosclerosis within the vascular wall and to assess the impact of SASH1 expression on endothelial cell functions. Method: Human carotids with atherosclerotic plaques were obtained from 58 patients (45 of them with known smoking status: smoker, non-smoker, ex-smokers), and were processed for gene expression analyses and immunostaining. To investigate its function, SASH1 was silenced in human aortic endothelial cells (HAECs) using two different siRNA and subcellular localization of SASH1 was determined by immunostaining and subcellular fractionation. Subsequently the transcriptomic analyses and functional experiments (wound healing, WST-1 proliferation or Matrigel assays) were performed to characterize SASH1 function. Results: SASH1 was expressed in human vascular cells (HAECs, smooth muscle cells) and in monocytes/macrophages. Its tissue expression was significantly higher in the atherosclerotic carotids of smokers compared to non-smokers (p < 0.01). In HAECs, SASH1 was expressed mostly in the cytoplasm and SASH1 knockdown resulted in an increased cell migration, proliferation and angiogenesis. Transcriptomic and pathway analyses showed that SASH1 silencing results in a decreased CYP1A1 expression possibly through the inhibition of TP53 activity. Conclusion: We showed that SASH1 expression is increased in atherosclerotic carotids in smokers and its silencing affects endothelial angiogenic functions; therefore we provide a potential link between smoking and atherosclerosis through SASH1 expression

    Graphical Modeling of Gene Expression in Monocytes Suggests Molecular Mechanisms Explaining Increased Atherosclerosis in Smokers

    No full text
    <div><p>Smoking is a risk factor for atherosclerosis with reported widespread effects on gene expression in circulating blood cells. We hypothesized that a molecular signature mediating the relation between smoking and atherosclerosis may be found in the transcriptome of circulating monocytes. Genome-wide expression profiles and counts of atherosclerotic plaques in carotid arteries were collected in 248 smokers and 688 non-smokers from the general population. Patterns of co-expressed genes were identified by Independent Component Analysis (ICA) and network structure of the pattern-specific gene modules was inferred by the PC-algorithm. A likelihood-based causality test was implemented to select patterns that fit models containing a path “smoking→gene expression→plaques”. Robustness of the causal inference was assessed by bootstrapping. At a FDR ≀0.10, 3,368 genes were associated to smoking or plaques, of which 93% were associated to smoking only. <em>SASH1</em> showed the strongest association to smoking and <em>PPARG</em> the strongest association to plaques. Twenty-nine gene patterns were identified by ICA. Modules containing <em>SASH1</em> and <em>PPARG</em> did not show evidence for the “smoking→gene expression→plaques” causality model. Conversely, three modules had good support for causal effects and exhibited a network topology consistent with gene expression mediating the relation between smoking and plaques. The network with the strongest support for causal effects was connected to plaques through <em>SLC39A8</em>, a gene with known association to HDL-cholesterol and cellular uptake of cadmium from tobacco, while smoking was directly connected to <em>GAS6</em>, a gene reported to have anti-inflammatory effects in atherosclerosis and to be up-regulated in the placenta of women smoking during pregnancy. Our analysis of the transcriptome of monocytes recovered genes relevant for association to smoking and atherosclerosis, and connected genes that before, were only studied in separate contexts. Inspection of correlation structure revealed candidates that would be missed by expression-phenotype association analysis alone.</p> </div

    Characteristics of the Gutenberg Health Study population.

    No full text
    †<p>p-values calculated from a χ<sup>2</sup> test for smoking and diabetes (number of subjects), and from an F test for all others. Standard errors or percents of individuals are in parenthesis.</p
    corecore