8 research outputs found

    Etude des mécanismes moléculaires controlant la biogenÚse des granules de sécrétion : Role de la chromogranine A, du complexe actomyosine et des lipides de la membrane golgienne

    No full text
    Neuroendocrine cells exhibit the constitutive secretory pathway which is common all cell types and allows the continuous renewal of the plasma membrane and the extracellular matrix, and the regulated secretory pathway, which is specific to secretory cells and allows hormone secretion following cell stimulation. The organelles supporting the latter pathway are dense-core secretory granules (SG), which store hormones and soluble glycoproteins, called granins. Among these, chromogranin A (CgA) plays a major role in the biogenesis of SG but the molecular mechanisms underlying this process are not clearly understood. Using non-endocrine COS7 cell line (which are devoid of granins and regulated secretory pathway), my host team has demonstrated that the CgA expression induces the formation of vesicles with structural and functional characteristic of SG. The proteome analysis of purified SG from a COS7 cell line stably expressing CgA (COS7-CgA) revealed the presence of cytoskeleton- and calcium-binding proteins. During my thesis, we focused our attention on myosin 1b (myo1b), actin and actin nucleation complex Arp2/3 due to their ability to induce the budding of post-Golgi compartments in non-endocrine cells. We have shown (i) that myo1b controls SG formation as welle as the regulated secretion in COS7-CgA and PC12 neuroendocrine cells, (ii) that myo1b and Arp2/3 complex are required to recruit fibrillar actin (F-actin) to the Golgi region and to SG formation. These results highlight for the first time the involvement of the actomyosin complex in SG formation. In order to identify the molecular link between luminal CgA and Cytosolic myo1b, we investigated the potential interactions of CgA with lipids of the trans-Golgi network (TGN) membrane. We showed (i) that CgA interacts with phosphatidic acid (PA), (ii) that the predominant PA species are common in Golgi and granular membranes, (iii) that Cg Ais able to interact specifically with these PA species included in artificial membranes, and (iv) that inhibition of PA production at the Golgi level significantly alters SG formation and regulated secretion in neuroendocrine cells. All these results obtained during my thesis suggest that the interaction between CgA and PA is crucial for SG biogenesis from the TGN membrane. We suggest that this interaction is at the origin of the formation of PA-enriched microdomains that could control the curvature of the TGN membrane and the recruitment of the actomyosin complex.Les cellules neuroendocrines possĂšdent d’une part la voie de sĂ©crĂ©tion constitutive, existant dans tous les types cellulaires, qui permet le renouvellement continu de la membrane plasmique et de la matrice extracellulaire, et d’autre part la voie de sĂ©crĂ©tion rĂ©gulĂ©e, spĂ©cifique aux cellules sĂ©crĂ©trices, qui permet la sĂ©crĂ©tion d’hormones suite Ă  la stimulation de la cellule. Les organites impliquĂ©s dans cette derniĂšre voie sont des granules de sĂ©crĂ©tion Ă  cƓur dense (GS), sui stockent les hormones ainsi que les glycoprotĂ©ines solubles, les granines. Parmi ces derniĂšres, la chromogranine A (CgA) joue un rĂŽle majeur dans la biogĂ©nĂšse des GS mais les mĂ©canismes molĂ©culaires ne sont pas clairement dĂ©finis. Dans une lignĂ©e de cellules non-endocrines COS7 (dĂ©pourvues de granines et donc de voie de sĂ©crĂ©tion rĂ©gulĂ©e), mon Ă©quipe d’accueil a dĂ©montrĂ© que l’expression de la CgA induit la formation de vĂ©sicules prĂ©sentant une structure et des fonctions caractĂ©ristiques des GS. L’analyse du protĂ©ome des GS purifiĂ©s Ă  partir d’une lignĂ©e de cellules COS7 exprimant de maniĂšre stable la CgA (COS7-CgA) a rĂ©vĂ©lĂ© la prĂ©sence de protĂ©ines liant les Ă©lĂ©ments du cytosquelette et le calcium. Durant ma thĂšse, nous avons focalisĂ© notre attention sur la myosine 1b (myo1b), l’actine et le complexe de nuclĂ©ation de l’actine Arp2/3 du fait de leur capacitĂ© Ă  induire le bourgeonnement de compartiments post-golgiens dans des cellules non-endocrines. Nous avons montrĂ© (i) que la myo1b contrĂŽle la formation des GS ainsi que la sĂ©crĂ©tion rĂ©gulĂ©e au sein des cellules COS7-CgA et des cellules neuroendocriniennes PC12, et (ii) que la myo1b et le complexe Arp2/3 permettent le recrutement d’actine fibrillaire dans la rĂ©gion golgienne et la formation des GS. Ces travaux montrent pour la premiĂšre fois l’implication du complexe actomyosine dans la formation des GS. Afin d’identifier le lien molĂ©culaire entre la CgA luminale et la myo1b cytosolique, nous avons recherchĂ© les interactions potentielles de la CgA avec les lipides de la membrane du rĂ©seau trans-golgien (TGN). Nous avons montrĂ© (i) que la CgA interagit avec l’acide phosphatidique (PA), (ii) que les espĂšces de PA prĂ©dominantes sont communes dans les membranes golgienne et granulaire, (iii) que la CgA est capable d’interagir spĂ©cifiquement avec des espĂšces de PA intĂ©grĂ©es avec des membranes artificielles et (iv) que l’inhibition de la production du PA au niveau golgien altĂšre significativement la formation des GS et la sĂ©crĂ©tion rĂ©gulĂ©e dans les cellules neuroendocrines. L’ensemble des rĂ©sultats obtenus dans le cadre de ma thĂšse suggĂšre que l’interaction entre la CgA et le PA est cruciale pour la biogenĂšse de GS Ă  partir de la membrane du TGN. Nous Ă©mettons l’hypothĂšse que cette interaction est Ă  l’origine de la formation de microdomaines enrichis en PA qui contrĂŽleraient la courbure de la membrane du TGN et le recrutement du complexe actomyosine

    Study of the molecular mechanisms controlling the biogenesis of secretory granules : Role of chromogranin A, actomyosin complex and lipids of the Golgi membrane

    No full text
    Les cellules neuroendocrines possĂšdent d’une part la voie de sĂ©crĂ©tion constitutive, existant dans tous les types cellulaires, qui permet le renouvellement continu de la membrane plasmique et de la matrice extracellulaire, et d’autre part la voie de sĂ©crĂ©tion rĂ©gulĂ©e, spĂ©cifique aux cellules sĂ©crĂ©trices, qui permet la sĂ©crĂ©tion d’hormones suite Ă  la stimulation de la cellule. Les organites impliquĂ©s dans cette derniĂšre voie sont des granules de sĂ©crĂ©tion Ă  cƓur dense (GS), sui stockent les hormones ainsi que les glycoprotĂ©ines solubles, les granines. Parmi ces derniĂšres, la chromogranine A (CgA) joue un rĂŽle majeur dans la biogĂ©nĂšse des GS mais les mĂ©canismes molĂ©culaires ne sont pas clairement dĂ©finis. Dans une lignĂ©e de cellules non-endocrines COS7 (dĂ©pourvues de granines et donc de voie de sĂ©crĂ©tion rĂ©gulĂ©e), mon Ă©quipe d’accueil a dĂ©montrĂ© que l’expression de la CgA induit la formation de vĂ©sicules prĂ©sentant une structure et des fonctions caractĂ©ristiques des GS. L’analyse du protĂ©ome des GS purifiĂ©s Ă  partir d’une lignĂ©e de cellules COS7 exprimant de maniĂšre stable la CgA (COS7-CgA) a rĂ©vĂ©lĂ© la prĂ©sence de protĂ©ines liant les Ă©lĂ©ments du cytosquelette et le calcium. Durant ma thĂšse, nous avons focalisĂ© notre attention sur la myosine 1b (myo1b), l’actine et le complexe de nuclĂ©ation de l’actine Arp2/3 du fait de leur capacitĂ© Ă  induire le bourgeonnement de compartiments post-golgiens dans des cellules non-endocrines. Nous avons montrĂ© (i) que la myo1b contrĂŽle la formation des GS ainsi que la sĂ©crĂ©tion rĂ©gulĂ©e au sein des cellules COS7-CgA et des cellules neuroendocriniennes PC12, et (ii) que la myo1b et le complexe Arp2/3 permettent le recrutement d’actine fibrillaire dans la rĂ©gion golgienne et la formation des GS. Ces travaux montrent pour la premiĂšre fois l’implication du complexe actomyosine dans la formation des GS. Afin d’identifier le lien molĂ©culaire entre la CgA luminale et la myo1b cytosolique, nous avons recherchĂ© les interactions potentielles de la CgA avec les lipides de la membrane du rĂ©seau trans-golgien (TGN). Nous avons montrĂ© (i) que la CgA interagit avec l’acide phosphatidique (PA), (ii) que les espĂšces de PA prĂ©dominantes sont communes dans les membranes golgienne et granulaire, (iii) que la CgA est capable d’interagir spĂ©cifiquement avec des espĂšces de PA intĂ©grĂ©es avec des membranes artificielles et (iv) que l’inhibition de la production du PA au niveau golgien altĂšre significativement la formation des GS et la sĂ©crĂ©tion rĂ©gulĂ©e dans les cellules neuroendocrines. L’ensemble des rĂ©sultats obtenus dans le cadre de ma thĂšse suggĂšre que l’interaction entre la CgA et le PA est cruciale pour la biogenĂšse de GS Ă  partir de la membrane du TGN. Nous Ă©mettons l’hypothĂšse que cette interaction est Ă  l’origine de la formation de microdomaines enrichis en PA qui contrĂŽleraient la courbure de la membrane du TGN et le recrutement du complexe actomyosine.Neuroendocrine cells exhibit the constitutive secretory pathway which is common all cell types and allows the continuous renewal of the plasma membrane and the extracellular matrix, and the regulated secretory pathway, which is specific to secretory cells and allows hormone secretion following cell stimulation. The organelles supporting the latter pathway are dense-core secretory granules (SG), which store hormones and soluble glycoproteins, called granins. Among these, chromogranin A (CgA) plays a major role in the biogenesis of SG but the molecular mechanisms underlying this process are not clearly understood. Using non-endocrine COS7 cell line (which are devoid of granins and regulated secretory pathway), my host team has demonstrated that the CgA expression induces the formation of vesicles with structural and functional characteristic of SG. The proteome analysis of purified SG from a COS7 cell line stably expressing CgA (COS7-CgA) revealed the presence of cytoskeleton- and calcium-binding proteins. During my thesis, we focused our attention on myosin 1b (myo1b), actin and actin nucleation complex Arp2/3 due to their ability to induce the budding of post-Golgi compartments in non-endocrine cells. We have shown (i) that myo1b controls SG formation as welle as the regulated secretion in COS7-CgA and PC12 neuroendocrine cells, (ii) that myo1b and Arp2/3 complex are required to recruit fibrillar actin (F-actin) to the Golgi region and to SG formation. These results highlight for the first time the involvement of the actomyosin complex in SG formation. In order to identify the molecular link between luminal CgA and Cytosolic myo1b, we investigated the potential interactions of CgA with lipids of the trans-Golgi network (TGN) membrane. We showed (i) that CgA interacts with phosphatidic acid (PA), (ii) that the predominant PA species are common in Golgi and granular membranes, (iii) that Cg Ais able to interact specifically with these PA species included in artificial membranes, and (iv) that inhibition of PA production at the Golgi level significantly alters SG formation and regulated secretion in neuroendocrine cells. All these results obtained during my thesis suggest that the interaction between CgA and PA is crucial for SG biogenesis from the TGN membrane. We suggest that this interaction is at the origin of the formation of PA-enriched microdomains that could control the curvature of the TGN membrane and the recruitment of the actomyosin complex

    Myosin 1b and F-actin are involved in the control of secretory granule biogenesis

    No full text
    Hormone secretion relies on secretory granules which store hormones in endocrine cells and release them upon cell stimulation. The molecular events leading to hormone sorting and secretory granule formation at the level of the TGN are still elusive. Our proteomic analysis of purified whole secretory granules or secretory granule membranes uncovered their association with the actomyosin components myosin 1b, actin and the actin nucleation complex Arp2/3. We found that myosin 1b controls the formation of secretory granules and the associated regulated secretion in both neuroendocrine cells and chromogranin A-expressing COS7 cells used as a simplified model of induced secretion. We show that F-actin is also involved in secretory granule biogenesis and that myosin 1b cooperates with Arp2/3 to recruit F-actin to the Golgi region where secretory granules bud. These results provide the first evidence that components of the actomyosin complex promote the biogenesis of secretory granules and thereby regulate hormone sorting and secretion.This work was supported by Institut National de la SantĂ© et de la Recherche MĂ©dicale, the University of Rouen Normandy, the Conseil RĂ©gional de Normandie and the MinistĂšre de l’Enseignement SupĂ©rieur et de la RecherchePeer Reviewe

    Chromogranin A preferential interaction with Golgi phosphatidic acid induces membrane deformation and contributes to secretory granule biogenesis

    No full text
    International audienceChromogranin A (CgA) is a key luminal actor of secretory granule biogenesis at the trans‐Golgi network (TGN) level but the molecular mechanisms involved remain obscure. Here, we investigated the possibility that CgA acts synergistically with specific membrane lipids to trigger secretory granule formation. We show that CgA preferentially interacts with the anionic glycerophospholipid phosphatidic acid (PA). In accordance, bioinformatic analysis predicted a PA‐binding domain (PABD) in CgA sequence that effectively bound PA (36:1) or PA (40:6) in membrane models. We identified PA (36:1) and PA (40:6) as predominant species in Golgi and granule membranes of secretory cells, and we found that CgA interaction with these PA species promotes artificial membrane deformation and remodeling. Furthermore, we demonstrated that disruption of either CgA PABD or phospholipase D (PLD) activity significantly alters secretory granule formation in secretory cells. Our findings show for the first time the ability of CgA to interact with PLD‐generated PA, which allows membrane remodeling and curvature, key processes necessary to initiate secretory granule budding
    corecore