• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 116
  • 65
  • 59
  • 23
  • 10
  • 6
  • 2
  • 2
  • 1
  • 1
  • 1
  • Tagged with
  • 337
  • 59
  • 50
  • 48
  • 46
  • 45
  • 41
  • 41
  • 36
  • 33
  • 31
  • 30
  • 27
  • 26
  • 22
  • 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.
321

Chronic Dietary Supplementation of Branched-Chain Amino Acids Does Not Attenuate Muscle Torque Loss in a Mouse Model of Duchenne Muscular Dystrophy

Sperringer, Justin Edward 12 September 2019 (has links)
Duchenne Muscular Dystrophy (DMD) is an X-linked recessive, progressive muscle-wasting disease characterized by mutations in the dystrophin gene. Duchenne muscular dystrophy is the most common and most severe form of inherited muscle diseases, with an incidence of 1 in 3,500 male births1,2. Mutations in the dystrophin gene result in non-functional dystrophin or the complete absence of the protein dystrophin, resulting in necrosis and fibrosis in the muscle, loss of ambulation, cardiomyopathies, inadequate or failure of respiratory function, and decreased lifespan. Although there has been little research for effective nutritional strategies, dietary intervention may be effective as an adjuvant treatment. In this study, wild type (WT) and mdx animals were provided either a control or elevated branched chain amino acid (BCAA) diet nocturnally for 25 weeks to determine if the elevated BCAAs would attenuate muscle torque loss. Twenty-five weeks of chronic, elevated BCAA supplementation had no impact on muscle function measures. Interestingly, mdx and WT animals had the same torque responses in the low stimulation frequencies (1 Hz – 30 Hz) compared to higher stimulation frequencies. Tetanus was reached at a much lower stimulation frequency in mdx animals compared to WT animals (100 Hz vs +150 Hz). The mdx mouse consistently had more cage activity in the light cycle X- and Y-planes. Interestingly, animals on the BCAA diet increased X-, Y-, and Z-plane activity in the dark cycles at four weeks while animals on the control diet more Z-plane activity at 25 weeks, although not significant. All three BCAAs were elevated in the plasma at 25 weeks, although only Leu was significantly elevated. The BCAAs had no effect on. The diaphragm and skeletal muscle masses were larger in mdx animals, and WT animals had a significantly larger epididymal fat pad. The active state of BCKDC determined by phosphorylation of the E1α enzyme was greater in WT animals in white skeletal muscle, but not red skeletal muscle. Protein synthesis effectors of the mTORC1 signaling pathway and autophagy markers were similar among groups. Wild type animals had increased mTORC1 effectors and animals on the BCAA diet had decreased autophagy markers, although not significant. Although BCAAs did not affect muscle function, fibrosis, or protein synthesis effectors, this study illustrates the functionality of mdx muscles over time. It would be interesting to see how the different muscle fiber types are affected by DMD, noting the differences between the diaphragm, heart, red muscle, and white muscle fibrosis markers. Although there was no increase in mTORC1 effectors with an elevated BCAA diet, it would be interesting to determine muscle protein synthesis, myofibrillar protein synthesis, and total protein turnover in the mdx mouse with an elevated BCAA diet, although the dietary intervention started when mice arrived at 4 weeks of age, earlier intervention may be beneficial early in the disease process. / Doctor of Philosophy / Duchenne Muscular Dystrophy (DMD) is an X-linked recessive, progressive muscle-wasting disease characterized by mutations in the dystrophin gene. Duchenne muscular dystrophy is the most common and most severe form of inherited muscle diseases, with an incidence of 1 in 3,500 male births1,2. Mutations in the dystrophin gene result in non-functional dystrophin or the complete absence of the protein dystrophin, resulting in necrosis and fibrosis in the muscle, loss of movement and walking ability, cardiomyopathies, inadequate or failure of respiratory function, and decreased lifespan. Although there has been little research for effective nutritional strategies, dietary intervention may be effective as an adjuvant treatment and palliative care. The branched chain amino acids (BCAAs) are known to directly stimulate muscle protein synthesis by direct activation of the mechanistic target of rapamycin complex 1 (mTORC1). This study aimed to illustrate the differences between diseased and healthy mice and determine if BCAAs can reduce muscle torque loss. Twenty-five weeks of chronic, elevated BCAA supplementation had no impact on muscle function measures. Interestingly, mdx and WT animals had the same torque responses in the low stimulation frequencies (1 Hz – 30 Hz) compared to higher stimulation frequencies. Tetanus was reached at a much lower stimulation frequency in mdx animals compared to WT animals (100 Hz vs +150 Hz). The mdx mouse consistently had more cage activity in the light cycle X- and Y-planes. Interestingly, animals on the BCAA diet increased X-, Y-, and Z-plane activity in the dark cycles at four weeks while animals on the control diet more Z-plane activity at 25 weeks, although not significant. All three BCAAs were elevated in the plasma at 25 weeks, although only Leu was significantly elevated. The BCAAs had no effect on. The diaphragm and skeletal muscle masses were larger in mdx animals, and WT animals had a significantly larger epididymal fat pad. The active state of BCKDC determined by phosphorylation of the E1α enzyme was greater in WT animals in white skeletal muscle, but not red skeletal muscle. Protein synthesis effectors of the mTORC1 signaling pathway and autophagy markers were similar among groups. Wild type animals had increased mTORC1 effectors and animals on the BCAA diet had decreased autophagy markers, although not significant. Although BCAAs did not affect muscle function, fibrosis, or protein synthesis effectors, this study illustrates the functionality of mdx muscles over time. It would be interesting to see how the different muscle fiber types are affected by DMD, noting the differences between the diaphragm, heart, red muscle, and white muscle fibrosis markers. Although there was no increase in mTORC1 effectors with an elevated BCAA diet, it would be interesting to determine muscle protein synthesis, myofibrillar protein synthesis, and total protein turnover in the mdx mouse with an elevated BCAA diet, although the dietary intervention started when mice arrived at 4 weeks of age, earlier intervention may be beneficial early in the disease process.
322

Virus host interactome du polyomavirus à cellules de Merkel / Merkel cell polyomavirus virus host interactome

Ferté-Chaudoy, Marion 15 September 2017 (has links)
Le polyomavirus à cellules de Merkel est aujourd’hui reconnu comme l’agent étiologique du carcinome à cellules de Merkel (CCM). Le cycle viral et les mécanismes de l’oncogenèse viro-induite sont peu connus et les connaissances se basent essentiellement sur les études menées notamment sur le polyomavirus SV40. L’objectif des travaux de thèse était d’identifier les interactions entre les protéines virales et les protéines cellulaires lors de l’infection ou dans le contexte du carcinome à cellules de Merkel (CCM). Pour identifier ces interactions, nous avons réalisé des cribles double hybride en levures sur les oncogènes du MCPyV et du BKPyV. Afin valider les interactions obtenues en levures, nous avons utilisé une méthode orthogonale de validation par complémentation en cellules de mammifères reposant sur la restauration de la luciférase de Gaussia princeps. La combinaison de ses deux techniques nous a permis de valider des interactions avec des partenaires cellulaires impliqués dans la régulation du cycle cellulaire ou encore de la voie Akt-mTOR. Les précédents travaux du laboratoire, qui portaient sur l’interactome des protéines mineures de capsides VP2/VP3, avaient également permis d’identifier des interactions avec des protéines de la voie NF-kB. Nous avons alors testé les interactions entre les oncogènes et la protéine mineure de capside VP2 avec des protéines cellulaires impliquées dans cette voie. Ces travaux nous ont conduits à tester l’activation de la voie, l’expression des gènes sous le contrôle de NF-kB et la régulation de l’apoptose. Les résultats obtenus montrent une action de la protéine VP2 sur l’activation de la voie NF-kB et une induction de l’apoptose. / The Merkel cell polyomavirus is now recognized as the etiologic agent of Merkel cell carcinoma (MCC). The viral cycle and viro-induced oncogenesis mechanisms are not fully understood and the knowledge is mainly based on the studies carried out particularly on the SV40 polyomavirus. The aim of our work is to identify interactions between viral proteins and cellular proteins during productive infection or in MCC context. To identify these interactions, we performed yeast two hybrid screens on MCPyV and BKPyV oncogenes, as control. To validate the interactions obtained in yeasts, we used an orthogonal method of validation by complementation in mammalian cells based on the restoration of Gaussia princeps luciferase. The combination of these two orthogonal techniques allowed us to validate interactions with cellular partners involved in cell cycle regulation or Akt-mTOR pathway. Previous lab work on VP2/VP3 minor capsid proteins allowed the identification of interactions with NF-kB pathway involved proteins. We examined the interactions between oncogenes, VP2, with the cellular proteins involved in this pathway. This work led us to evaluate pathway activation, genes expression under the control of NF-kB and apoptosis regulation. These results evidenced an action of the VP2 protein on the activation of NF-kB pathway and an induction of apoptosis.
323

Identification of Therapeutic Targets for Oral Squamous Cell Carcinoma

Avinash, Pradhan Shalmali January 2013 (has links) (PDF)
Oral squamous cell carcinoma (OSCC) is the most common head and neck cancer, with a worldwide incidence of 275,000 new cases annually (Warnakulasuriya, 2009). Globally, the head and neck carcinoma represents a major cause of morbidity and mortality and is the sixth most commonly occurring cancer (Warnakulasuriya, 2009). A majority (>90%) of the head and neck cancers are squamous in origin and thus are linguistically referred to as head and neck squamous cell carcinoma (HNSCC) (Warnakulasuriya, 2009). HNSCC includes cancers of the oral cavity, larynx and pharynx; oral cancer being the most common (Warnakulasuriya, 2009). Although, HNSCC is the sixth most common cancer globally (Warnakulasuriya, 2009), the Indian scenario is graver. According to GLOBOCAN 2008 (http://globocan.iarc.fr), the worldwide age standardized incidence rate (ASR) for HNSCC (and thus OSCC) is 5.3 and 2.5 per 100,000 males and females respectively (Ferlay et al., 2010). In India, the ASR is 9.8 and 5.2 per 100,000 males and females respectively, clearly demonstrating a remarkably high incidence rate of OSCC (Ferlay et al., 2010; http://globocan.iarc.fr). OSCC is a peculiar cancer which is largely preventable and rarely presents as a familial disorder. The most common etiological factors associated with OSCC include tobacco and alcohol consumption (Johnson, 2001). Additionally, high risk human papillomaviruses (HPV strains 16 and 18) as well as genetic predispositions have been implicated. The treatment of OSCC mainly relies on surgical resection of the tumor. The site, size, depth of infiltration and proximity to the bone of the tumor determine whether a combination of surgery with radiation therapy or chemotherapy would be advised (Scully and Bagan, 2009). The concomitant chemo-radiation therapy is the most commonly used strategy in locally advanced cancer. Taxanes (e.g., paclitaxel and docetaxel) and platinum-based induction chemotherapy (e.g., cisplatin) are the options in the treatment of locally advanced cancer. Epidermal growth factor receptor (EGFR) targeted with cetuximab in combination with radiotherapy has been successfully tested in a large randomized trial and thus is currently a new option (Scully and Bagan, 2009). The success of cetuximab has paved the path for the development and implementation of molecules targeting various signaling pathways. Despite extensive research on oral squamous cell carcinoma (OSCC), the five-year survival rate has not changed in several decades with the exception of the targeted treatment strategies involving cetuximab as discussed above. The current chemotherapeutic approaches lack selectivity and are flagitious. Thus, effective treatment of OSCC requires the identification of molecular targets to design appropriate therapeutic strategies. To this end, the present study took three distinct approaches in order to validate the use of existing targets and to reveal novel prognostic biomarkers and therapeutic targets. 1) Targeting the PI3K-AKT-MTOR pathway in OSCC and identification of determinants of its sensitivity. 2) Gene expression analysis of ectopically overexpressed TSC2 to identify new therapeutic targets and prognostic biomarkers as well as to elucidate the genes regulated by it. 3) Expression profiling of CYP1B1 in order to validate the use of CYP1B1 based prodrug therapy in OSCC. Investigations pertaining to the changes in gene and protein expression profiles in malignant as well as pre-malignant lesions have documented the deregulation of the PI3K-AKT-MTOR (phosphoinositide 3-kinase-AKT-mechanistic target of rapamycin) and EGFR (epidermal growth factor receptor) pathways in OSCC which are being widely targeted in many therapeutic strategies (Molinolo et al., 2007; Chakraborty et al., 2008; Matta and Ralhan, 2009; Molinolo et al., 2009; Stransky et al., 2011). The PI3K-AKT-MTOR pathway is a central hub for controlling cellular proliferation and growth in response to various intracellular as well as extracellular stimuli. Crucial signaling cascades including WNT, RAS, HIF-1α and AMPK cross-talk with the PI3K-AKT-MTOR pathway at a variety of molecular junctions. Thus, making this pathway sensitive to perceiving various growth modulatory conditions, ranging from the presence of growth factors to hypoxia and nutrient deprivation (Sengupta et al., 2010; Yang and Guan, 2007). The aberrant expression of the PI3K-AKT-MTOR pathway in OSCC advocated the targeting of this coveted pathway (Chakraborty et al., 2008). In various cancers, the monotherapeutic treatments with inhibitors like LY294002 (PI3K inhibitor) and rapamycin (MTOR inhibitor) demonstrated reduced efficacies. Such reduced efficacies were attributed to the drug toxicity and non-specific action of LY294002 (Davies et al., 2000; Sun et al., 2005; Ikezoe et al., 2007; Wang et al., 2008; Liu et al., 2009), or the ablation of a feedback inhibition loop leading to the reactivation of the PI3K-AKT-MTOR pathway by rapamycin (O'Reilly et al., 2006; Carracedo et al., 2008). Thus, rapamycin or its analogues demonstrated mediocre efficacy due to cytostatic effects in clinical trials, primarily due to the paradoxical activation of major survival kinases namely MAPK and AKT (O'Reilly et al., 2006; Carracedo et al., 2008). The present study aimed at increasing the efficacy of these drugs by incorporating a combinatorial approach. The MTT assay demonstrated that prolonged monotherapeutic treatments with rapamycin led to a modest growth inhibition in three OSCC (KB, SCC131 and SCC084) and HeLa cell lines. Western blot analysis of the phosphorylation status of AKT and RPS6KB1 revealed that monotherapeutic treatments with rapamycin for 96 hr led to the reactivation of the PI3K-AKT-MTOR pathway. Thus, the modest growth inhibitory effect of rapamycin was attributed to the reactivation of the PI3K-AKT-MTOR pathway. A combinatorial treatment approach was hence believed to circumvent this problem in order to increase the efficacy of targeting the PI3K-AKT-MTOR pathway. The PI3K inhibitor LY294002 was used combinatorially with rapamycin. This prolonged dual combinatorial treatment regime was distinctly more efficacious than either of the drugs alone and led to a reduction in cellular viability accompanied by increased sub-G1 population, indicating marked cell death that was characterized as caspase-3 dependent apoptosis. The differential sensitivity of the cell lines towards this combinatorial treatment revealed a novel determinant of the sensitivity, the transactivation of EGFR. The cell lines (SCC131 and SCC084) that were capable of transactivating EGFR were relatively resistant to the dual targeting of PI3K and MTOR in comparison to cell lines that did not transactivate EGFR (HeLa and KB). Further, targeting PI3K, MTOR and EGFR simultaneously was more efficacious in the presence of EGFR transactivation than dually targeting PI3K and MTOR. The results conclusively proved that the combinatorial therapeutic approach dually targeting PI3K and MTOR is a promising treatment strategy as compared to a monotherapeutic treatment and a major factor determining the sensitivity towards this treatment is the status of autophosphorylation of EGFR (Tyr1173) which governs the potential for EGFR transactivation by the combinatorial treatment. Thus, this study demonstrated that the status of EGFR autophosphorylation (Tyr1173) can be used as a biomarker to predict the sensitivity towards the combinatorial targeting of PI3K and MTOR in OSCC. The PI3K-AKT-MTOR pathway is negatively regulated by TSC2 (tuberous sclerosis complex 2; tuberin) (Tee et al., 2002). The importance of the TSC2 gene in the regulation of cell growth and proliferation is irrefutable. TSC2 facilitates the crosstalk between a variety of cellular signals, making it a crucial hub where many cellular networks integrate like AKT, MAPK and AMPK (Clements et al., 2007; Rosner et al., 2007; Rosner et al., 2008). It is a tumor suppressor gene and is downregulated in many cancers including OSCC (Chakraborty et al., 2008). In order to identify the genes regulated by TSC2 in OSCC, we stably overexpressed TSC2 in KB cells and the changes in the gene expression profiles caused by this ectopic overexpression were observed using a whole genome expression microarray. The results showed differential regulation of 268 genes (107 genes were upregulated and 161 genes were downregulated, p<0.05, fold change ≥ 1.5). A majority of these genes were functionally associated with transcription, cell growth and proliferation, apoptosis, cell cycle and neurogenesis. Functional annotation and network analysis was performed by using the DAVID v6.7 and IPA version 8.7 softwares. The microarray data revealed a novel aspect in the crosstalk between WNT signaling and TSC2, namely the transcriptional regulation of WNT signaling by TSC2. Further, in the context of therapeutic applications, the microarray analysis revealed multiple genes that were functionally categorized to be involved in response to radiation, UV and drugs (e.g., SERPINB13 and IL1B). Future studies on the regulation of such genes that are involved in responses to drugs and radiation may give insights into the role of TSC2 in resistance or sensitivity towards chemotherapy and radiation therapy. Moreover, EREG, a member of the epidermal growth factor family, was found to be the most downregulated gene in the microarray analysis. Previous reports have documented elevated levels of EREG in tuberous sclerosis lesions and its association with poor clinical prognosis in OSCC patients (Li et al., 2008; Shigeishi et al., 2008), making its regulatory aspects intriguing. Additionally, published data on the transcriptional functions of TSC2 instigated us to analyze the role of TSC2 in the regulation of EREG. TSC2 has been shown to modulate the transcription mediated by members of the steroid receptor superfamily of genes (Henry et al., 1998) and was shown to bind specifically to ERα and inhibit estrogen induced proliferation (Finlay et al., 2004). Also, TSC2 has been shown to possess C-terminal transcriptional activation domains (Tsuchiya et al., 1996). We have therefore attempted to investigate the transcription related functional aspects of TSC2 by exploiting the observed transcriptional repression of EREG. The physiological roles of TSC1 and TSC2 that are independent of the PI3K-AKT-MTOR pathway have been termed as ‘non-canonical’ (Neuman and Henske, 2011). The repression of EREG by TSC2 was observed to be insensitive to rapamycin, suggesting that it was independent of MTORC1 and thus a non-canonical function of TSC2. To determine whether the repression in EREG was at the level of the promoter, we performed a dual luciferase reporter assay. The results showed that the EREG promoter was repressed by stable as well as transient overexpression of TSC2. In order to elucidate the mechanism of transcriptional regulation by TSC2, we performed the ChIP analysis to observe the in vivo binding of TSC2 to the EREG promoter. In the ChIP analysis with the anti-TSC2 antibody, we observed that TSC2 did not bind to the EREG promoter between the regions -857 bp to -302 bp or -325 bp to +165 bp. Further, in silico analysis revealed an interesting trend among the transcription factors that were differentially regulated by TSC2 and had putative binding sites on the EREG promoter. A majority of these transcription factors (17/21) were downregulated by the overexpression of TSC2. This observation suggested that the repression of EREG could be an indirect effect due to repression of transcription factors caused by overexpression of TSC2. On the whole, this study revealed novel functions of TSC2 in OSCC with implications in determining novel biomarkers and therapeutic targets. As discussed previously, OSCC has a very flagitious treatment regime. A prodrug approach is thought to aid in targeting chemotherapy (Rooseboom et al., 2004). CYP1B1, a member of the cytochrome P450 family, has been implicated in chemical carcinogenesis (Bandiera et al., 2005; Sliwinski et al., 2010). There exists a general accordance that this protein is overexpressed in a variety of cancers (e.g., colon, lung, renal, bladder, prostate, breast, endometrial and esophageal cancers), making it an ideal candidate for a prodrug therapy (McFadyen et al., 1999; Murray et al., 2001; McFadyen et al., 2004; Sissung et al., 2006; Wen and Walle, 2007; Sliwinski et al., 2010). The activation of the prodrug facilitated by CYP1B1 would enable the targeting of chemotherapy to tumor tissues in which CYP1B1 is specifically overexpressed as a result reducing the non-specific side effects that the current chemotherapy elicits (Rooseboom et al., 2004). This study was aimed at validating the use of CYP1B1 as a target for the prodrug therapy in OSCC. The expression profile of CYP1B1 was analysed in a panel of 51 OSCC tumors, their corresponding normal tissues, an epithelial dysplasia lesion and its matched normal tissue by qRT-PCR, Western blotting and Immunohistochemistry. Counterintuitively, CYP1B1 was found to be downregulated in 77.78% (28/36) tumor tissues in comparison to their corresponding normal tissues as well as in the epithelial dysplasia lesion compared to its matched normal tissue at the transcriptional level, and in 92.86% (26/28) of tumor tissues at the protein level. This clearly demonstrated the downregulation of CYP1B1 at the transcriptional and translational levels in tumor tissues in comparison to their corresponding normal tissues. These observations indicate that caution should be observed as this therapy may not be applicable universally to all cancers. Since CYP1B1 has been shown to be involved in the activation of pro-carcinogens (Murray et al., 2001; Bandiera et al., 2005; Sissung et al., 2006), its inhibition could facilitate the development of a prophylactic therapy for oral cancer. Overall, this study has identified the transactivation of EGFR as a determinant of sensitivity towards combinatorial targeting of PI3K and MTOR in OSCC and has demonstrated that the autophosphorylation of EGFR (Tyr1173) can be used as a marker to judge the sensitivity towards this treatment. In the clinical perspective, the identification of such markers would aid in predicting the efficacy of targeted therapies. Such investigations would enable the strategic treatment of OSCC patients, thus decreasing the time lost in trial and errors for determining the appropriate treatment. Additionally, this study elucidated a novel role of TSC2 in the transcriptional repression of EREG, a prognostic biomarker for OSCC. Further, the study revealed potential prognostic biomarkers as well as therapeutic targets that are regulated by TSC2 by using a whole genome expression microarray. Moreover, the counterintuitive downregulation of CYP1B1 in OSCC tumors suggested the possibility of a prophylactic therapy for oral cancer but also advised a precautionary note for the application of prodrug treatments based on CYP1B1 overexpression in OSCC.
324

Modification of ion channel auxiliary subunits in cardiac disease

Al Katat, Aya 10 1900 (has links)
L’infarctus du myocarde (IM) survenant après l’obstruction de l’artère coronaire est la cause principale des décès cardiovasculaires. Après l’IM, le coeur endommagé répond à l’augmentation du stress hémodynamique avec une cicatrice et une hypertrophie dans la région non-infarcie du myocarde. Dans la région infarcie, la cicatrice se forme grâce au dépôt du collagène. Pendant formation de la cicatrice, les cardiomyocytes ventriculaires résidant dans la région non-infarcie subissent une réponse hypertrophique après l’activation chronique due au système sympathique et à l’angiotensine II. La cicatrisation préserve l’intégrité structurale du coeur et l'hypertrophie des cardiomyocytes apporte un support ionotropique. Le canal CaV1.2 joue un rôle dans la réponse hypertrophique après l’IM. L’activation du CaV1.2 déclenche la signalisation dépendante de Ca2+ induisant l’hypertrophie. Cependant, il est rapporté que l’ouverture des canaux potassiques (KATP) ATP sensitifs joue un rôle sélectif dans l’expansion de la cicatrice après IM. Malgré leur expression dans les coeurs mâles, les KATP fournissent une cardioprotection sexe dépendante limitant l’expansion de la cicatrice chez les femelles. L’administration de rapamycine aux rates ayant subi un infarctus produit l’expansion de la cicatrice, soutenant la relation possible entre la cible de rapamycine, mTORC1 et les KATP dans la cardioprotection sexe spécifique. Effectivement, dans les cellules pancréatiques α, la signalisation mTORC1 était couplée à l'activation du KATP. Cependant, le lien entre mTORC1 et les canaux KATP dans le coeur reste inconnu. L'objectif de la thèse est d’examiner le rôle des canaux ioniques dans le remodelage cardiaque post-IM, surtout des canaux calciques dans l'hypertrophie et d'élucider la relation entre les KATP et mTORC1. L’hypothèse première teste que l’hypertrophie médiée par le système sympathique des cardiomyocytes ventriculaires des rats néonataux (NRCM) produit une augmentation de l’influx calcique après une augmentation des sous-unités du CaV1.2. Le traitement de norépinéphrine (NE) quadruple l’amplitude du courant calcique type L et double l’expression protéique des sous unités de CaVα2δ1 et CaVβ3. L’hypertrophie des NRCM au NE s’associe à une augmentation de la phosphorylation de la Kinase ERK 1/2. Le β1-bloqueur metoprolol et l’inhibiteur ii de ERK1/2 diminuent l’effet de NE sur CaVα2δ1. Cependant, l’augmentation de CaVβ3 et de la réponse hypertrophique persiste. Ainsi, le signal β1-adrenergique à travers ERK augmente les sous-unités CaVα2δ1 outre l’hypertrophie. L’autre hypothèse examine la spécificité du sexe sur l’expansion cicatricielle médiée par rapamycine et l’influence de mTOR sur l’expression de KATP. Rapamycin augmente la surface de la cicatrice et inhibe la phosphorylation de mTOR chez les coeurs de femelles. Dans les coeurs des deux sexes, la phosphorylation de mTOR et l’expression de KATP, Kir6.2 et SUR2A sont similaires. Cependant, une grande inactivation de la tubérine et une faible expression de raptor sont détectées chez les femelles. Le traitement à l’ester de phorbol des NRCM induit l’hypertrophie, augmente la phosphorylation de p70S6K et l’expression SUR2A. Le prétraitement par Rapamycine atténue chacune des réponses. Rapamycin démontre un patron d’expansion cicatriciel sexe spécifique et une régulation de phosphorylation de mTOR dans IM. Aussi, l’augmentation de SUR2A dans les NRCM traités par PDBu révèle une interaction entre mTOR et KATP. / Myocardial infarction (MI) secondary to the obstruction of the coronary artery is the main cause of cardiovascular death. Following MI, the damaged heart adapts to the increased hemodynamic stress via formation of a scar and a hypertrophic response of ventricular cardiomyocytes in the non-infarcted myocardium. In the infarcted region, a scar is formed via the rapid deposition of collagen. With ongoing scar formation, ventricular cardiomyocytes in the non-infarcted myocardium undergo a hypertrophic response secondary to the chronic activation by the sympathetic system and angiotensin II. Collectively, scar formation and cardiomyocyte hypertrophy preserve the structural integrity of the heart and provide inotropic support, respectively. CaV1.2 channels play a significant role in the hypertrophic response post-MI. Notably, the activation of CaV1.2 channel triggers Ca2+-dependent signaling that induces hypertrophy. By contrast, the opening of ATP-sensitive potassium (KATP) channels was shown to partake in selective scar expansion following MI. Notwithstanding its expression in male hearts, KATP channels endow a sex-dependent cardioprotection limiting scar expansion selectively in females. Moreover, administration of the macrolide rapamycin to the infarcted female rat heart led to scar expansion, supporting the possible relationship between the target of rapamycin, mTORC1 and KATP channels in providing sex-specific cardioprotection. Indeed, in pancreatic-α cells, mTORC1 signaling was coupled to KATP channel activation. However, whether mTORC1 targets KATP channels in the heart remains unknown. Thus, the AIM of the thesis was to explore the role of ion channels in cardiac remodeling post-MI by specifically addressing the role of Ca channels in cardiomyocyte hypertrophy and elucidate the potential relationship between KATP channels and mTORC1 signaling. The first study tested the hypothesis that hypertrophied neonatal rat ventricular cardiomyocytes (NRVMs) following sympathetic stimulation translated to an increase in calcium influx secondary to the augmentation of CaV1.2 channel subunits. NE treatment led to a 4-fold increase of L-type Ca2+ peak current associated with a 2-fold upregulation of CaVα2δ1 and CaVβ3 protein subunits in hypertrophied NRVMs. The hypertrophic response of NNVMs to NE was associated with the increased phosphorylation of extracellular regulated kinase (ERK1/2). The β1-blocker metoprolol and the ERK1/2 inhibitor suppressed NE-mediated protein upregulation of CaVα2δ1 whereas CaVβ3 upregulation and the hypertrophic response persisted. Therefore, sympathetic mediated β1-adrenergic signaling via ERK selectively upregulated the CaVα2δ1 subunit independent of NRVM hypertrophy. The second study tested the hypothesis that rapamycin-mediated scar expansion was sexspecific and mTOR influenced KATP channel subunit expression. Rapamycin administration translated to scar expansion and inhibited mTOR phosphorylation exclusively in females. In normal adult male and female rat hearts, mTOR phosphorylation and protein levels of KATP channel subunits Kir6.2 and SUR2A were similar. However, greater tuberin inactivation and reduced raptor protein levels were detected in females. NRVMs treated with a phorbol ester induced hypertrophy, increased p70S6K phosphorylation and SUR2A protein levels and rapamycin pretreatment attenuated each response. Thus, rapamycin administration to MI rats unmasked a sex-specific pattern of scar expansion and highlighted the disparate regulation of mTOR phosphorylation. Moreover, rapamycin-dependent upregulation of SUR2A in PDButreated NRVMs revealed a novel interaction between mTOR and KATP channel subunit expression
325

Regulation of UV-Protective Pathways Downstream of the Melanocortin 1 Receptor in Melanocytes

Wolf Horrell, Erin M. 01 January 2016 (has links)
Malignant cutaneous melanoma is the deadliest form of skin cancer, and a majority of melanoma diagnoses are a result of exposure to ultraviolet (UV) radiation. UV radiation causes DNA damage, which if not repaired correctly via nucleotide excision repair (NER) can result in mutations and melanomagenesis. The melanocortin 1 receptor (MC1R) is a Gs protein coupled receptor located on melanocyte plasma membranes and is involved in protecting the skin from UV induced damage. MC1R signaling results in the activation of two protective pathways: 1) induction of eumelanin synthesis downstream of micropthalmia-associated transcription factor (MITF) and 2) acceleration of NER downstream of ataxia telangiectaseia mutated and Rad3 related (ATR). MC1R signaling, however, also promotes melanocyte proliferation, therefore, the activation of the MC1R pathway must be regulated. The overall hypothesis of this dissertation is that the pathways downstream of MC1R can be manipulated to protect against UV induced damage. Chapter 2 investigates the regulation of the MC1R neutral antagonist human β-defensin 3 (βD3). UV damage did not induce βD3 mRNA expression in ex vivo human skin explants. The induction of βD3 expression instead correlated with inflammatory cytokines including TNF. Chapter 3 investigates the interdependence and cross talk between the two protective pathways downstream of MC1R. We directly tested the effect of MITF on the acceleration of NER and the effect of ATR on the induction of eumelanin synthesis following MC1R activation. MITF was not required for the acceleration of NER as mediated by ATR, however, the induction of transcription of enzymes involved in eumelanin synthesis was dependent upon ATR kinase activity. Finally, Chapter 4 investigates the mechanism by which MC1R promoted proliferation and whether the two UV protective pathways downstream of MC1R could be selectively activated without the risk of melanocyte proliferation. MC1R signaling resulted in activation of the mechanistic target of rapamycin complex 1 (mTORC1), a major regulator of cell growth and proliferation. Inhibition of mTORC1 signaling via rapamycin prevented MC1R induced proliferation in vitro. Rapamycin, however, did not prevent MC1R induced eumelanin synthesis or the acceleration of NER in vitro or in vivo suggesting it is possible to selectively activate the beneficial signaling pathways without the risk of melanocyte proliferation. The results of this dissertation suggest that MC1R signaling could be augmented in individuals to prevent UV induced damage.
326

Rôles des facteurs de croissance dans la prolifération de la cellule β-pancréatique en réponse à un excès de nutriments : étude du facteur de croissance HB-EGF et du récepteur à l’EGF

Benterki, Isma 04 1900 (has links)
Le diabète de type 2 (DT2) résulte d’une résistance à l’insuline par les tissus périphériques et par un défaut de sécrétion de l’insuline par les cellules β-pancréatiques. Au fil du temps, la compensation des îlots de cellules β pour la résistance à l’insuline échoue et entraine par conséquent une baisse progressive de la fonction des cellules β. Plusieurs facteurs peuvent contribuer à la compensation de la cellule β. Toutefois, la compréhension des mécanismes cellulaires et moléculaires sous-jacents à la compensation de la masse de la cellule β reste à ce jour inconnue. Le but de ce mémoire était d’identifier précisément quel mécanisme pouvait amener à la compensation de la cellule β en réponse à un excès de nutriments et plus précisément à l’augmentation de sa prolifération et de sa masse. Ainsi, avec l’augmentation de la résistance à l’insuline et des facteurs circulants chez les rats de six mois perfusés avec du glucose et de l’intralipide, l’hypothèse a été émise et confirmée lors de notre étude que le facteur de croissance HB-EGF active le récepteur de l’EGF et des voies de signalisations subséquentes telles que mTOR et FoxM1 impliquées dans la prolifération de la cellule β-pancréatique. Collectivement, ces résultats nous permettent de mieux comprendre les mécanismes moléculaires impliqués dans la compensation de la masse de la cellule β dans un état de résistance à l’insuline et peuvent servir de nouvelles approches thérapeutiques pour prévenir ou ralentir le développement du DT2. / Type 2 diabetes (T2D) results from insulin resistance in peripheral tissues and impaired insulin secretion from the pancreatic β-cell. Over the time, compensation of the β cell islets for insulin resistance fails, and therefore leads to a gradual decline in β-cell function. Several factors may contribute to β-cell compensation. However, the cellular and molecular mechanisms underlying β-cell compensation remain unknown. The purpose of this thesis was to identify what mechanism could lead to β cell compensation in response to nutrients excess and specifically the increase in proliferation and β-cell mass. Thus, with increasing insulin resistance and circulating factors in the 6 month rats infused with glucose + intralipid, the hypothesis was made and confirmed in our study that the growth factor HB-EGF would activate the EGF receptor, and subsequent signaling pathways such as mTOR and FoxM1, both involved in the proliferation of the pancreatic beta-cell. Collectively, these results allow us to understand better the molecular mechanisms involved in the β cell compensation in the insulin resistance state and may serve as a potential new therapeutic approach to prevent or delay T2D development.
327

Studies on Signal Transduction Mechanisms in Rhabdomyosarcoma

Durbin, Adam 06 August 2010 (has links)
Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma of childhood, with two predominant histologic subtypes: embryonal and alveolar. These histologies display distinct clinical courses, and despite refinements in dose and duration of multimodality therapy, the 5-year overall survival of patients diagnosed with metastatic RMS remains <30%. Thus, there is an urgent need to define novel targets for therapeutic intervention. Interrogation of cancer cell signal transduction pathways that regulate the pathogenic behaviours of tumor cells has been successful in defining targets in numerous tumor types. These have ultimately yielded clinically-relevant drugs that have improved the disease-free and overall survival of patients diagnosed with cancer. Work contained in this thesis describes the interrogation of several potential targets for inhibition in RMS. Interruption of RMS cell proliferation, survival and apoptosis is examined through disruption of the protein kinase integrin-linked kinase (ILK) and the nuclear receptor estrogen-receptor β. ILK, in particular, is demonstrated to have dual competing functions through the regulation of c-jun amino-terminal kinase (JNK) signaling: an oncogene in alveolar, and a tumor suppressor in embryonal RMS. These findings are recapitulated in other tumor cell lines, indicating that expression levels of JNK1 correlate with ILK function in a broad spectrum of tumor types. Furthermore, interruption of rhabdomyosarcoma cell migration as a surrogate marker of metastasis is examined through disruption of the stromal-cell derived factor 1α/chemokine (CXC)receptor 4 signaling network, as well as through cooperative interactions between ILK and the mammalian target of rapamycin. Finally, we demonstrate that the insulin-like growth factor pathway is a potential target for therapeutic inhibition, which also distinguishes tumors of embryonal and alveolar histology. These studies provide a rationale for the development of novel agents, as well as the use of established drugs targeting these pathways in rhabdomyosarcoma.
328

Studies on Signal Transduction Mechanisms in Rhabdomyosarcoma

Durbin, Adam 06 August 2010 (has links)
Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma of childhood, with two predominant histologic subtypes: embryonal and alveolar. These histologies display distinct clinical courses, and despite refinements in dose and duration of multimodality therapy, the 5-year overall survival of patients diagnosed with metastatic RMS remains <30%. Thus, there is an urgent need to define novel targets for therapeutic intervention. Interrogation of cancer cell signal transduction pathways that regulate the pathogenic behaviours of tumor cells has been successful in defining targets in numerous tumor types. These have ultimately yielded clinically-relevant drugs that have improved the disease-free and overall survival of patients diagnosed with cancer. Work contained in this thesis describes the interrogation of several potential targets for inhibition in RMS. Interruption of RMS cell proliferation, survival and apoptosis is examined through disruption of the protein kinase integrin-linked kinase (ILK) and the nuclear receptor estrogen-receptor β. ILK, in particular, is demonstrated to have dual competing functions through the regulation of c-jun amino-terminal kinase (JNK) signaling: an oncogene in alveolar, and a tumor suppressor in embryonal RMS. These findings are recapitulated in other tumor cell lines, indicating that expression levels of JNK1 correlate with ILK function in a broad spectrum of tumor types. Furthermore, interruption of rhabdomyosarcoma cell migration as a surrogate marker of metastasis is examined through disruption of the stromal-cell derived factor 1α/chemokine (CXC)receptor 4 signaling network, as well as through cooperative interactions between ILK and the mammalian target of rapamycin. Finally, we demonstrate that the insulin-like growth factor pathway is a potential target for therapeutic inhibition, which also distinguishes tumors of embryonal and alveolar histology. These studies provide a rationale for the development of novel agents, as well as the use of established drugs targeting these pathways in rhabdomyosarcoma.
329

Caractérisation des processus moléculaires impliqués dans l’activation de la protéine IKKbeta par l'angiotensine II et son rôle dans la réponse phénotypique des CMLV

Doyon, Priscilla 12 1900 (has links)
No description available.
330

Identification et activation des cellules souches neurales quiescentes dans le cerveau adulte et durant le vieillissement

Cochard, Loïc 12 1900 (has links)
La neurogenèse est maintenue dans le cerveau adulte dans des régions restreintes du cerveau appelées niches neurogéniques. L’une des niches principales est la zone ventriculaire/sous-ventriculaire (V-SVZ) dans laquelle résident des cellules souches neurales (NSCs). Les NSCs sont à l’origine de la formation des nouveaux neurones en donnant naissance aux progéniteurs puis aux neuroblastes. Les études récentes sur la neurogenèse ont mis en évidence l’existence des NSCs quiescentes (qNSCs, aussi appelées cellules B1) et des NSCs actives (aNSCs). Le modèle actuel de la neurogenèse adulte place les qNSCs B1 en amont des aNSCs. L’hypothèse étant que cette population dormante constitue une « réserve », afin de maintenir les aNSCs tout au long de la vie. Les techniques actuelles ne permettent pas de cibler les qNSCs spécifiquement in vivo et donc, d’analyser leurs propriétés biologiques, leurs mécanismes d’activation ainsi que leur relation avec les aNSCs. Cette compréhension est nécessaire pour la mise au point de stratégies thérapeutiques pouvant utiliser le potentiel des cellules souches pour restaurer la neurogenèse dans les contextes de vieillissement et de maladies neurodégénératives. Afin de caractériser les qNSCs de la V-SVZ, nous avons utilisé l’électroporation de plasmides dans un modèle de souris rapportrice Rosa26-stop-EYFP. Dans celle-ci, la séquence codant pour la protéine EYFP précédée par un codon STOP floxé, est inséré au locus Rosa26. L’excision du codon STOP par une recombinase permet l’expression du rapporteur dans les cellules électroporées ainsi que leur descendance. Cette technique nous a permis de cibler spécifiquement une population d’astrocytes en contact avec le ventricule et d’étudier leur contribution à la neurogenèse adulte. À la différence des approches virales et transgéniques, l’électroporation peut cibler les cellules quiescentes et l’expression du plasmide est limité aux cellules en contact avec le ventricule. Grâce à cette technique nous avons mis en évidence des éléments surprenants : i) cette population est majoritairement quiescente et ne contribuent à la neurogenèse que de manière minimale, ii) cette population ne participe pas à la régénération de la niche in vivo, iii) elles ne génèrent pas les aNSCs à l’origine des neurosphères in vitro et iv) son activité neurogénique peut être augmentée en exprimant le gène pro-neural Mash1. Ensuite, nous nous sommes intéressés au rôle de la signalisation EGFR dans la régulation de l’activité des cellules souches/progéniteurs neuraux (NSPCs). Dans cette seconde étude, nous montrons que i) la signalisation EGFR est réduite avec l’âge, ii) PI3K/AKT, MEK/ERK et mTOR régulent différemment la prolifération, la différenciation et la survie des NSPCs et iii) l’activation d’EGFR dans les qNSCs permet d’augmenter la neurogenèse sous ventriculaire à 3 mois, mais pas à 6 mois ou dans un modèle de la maladie d’Alzheimer. Nos données suggèrent donc que les qNSCs représentent une population hétérogène et/ou présentant 2 voies neurogéniques distinctes. De plus, nous avons montré que les voies de signalisation associées à EGFR exercent un contrôle différentiel sur l’activité des NSPCs. Enfin, nos résultats indiquent que les facteurs présents dans la niche sous-ventriculaire lors du vieillissement inhibent de manière dominante l’activation des NSCs. / Neurogenesis is maintained in restricted regions of the adult brain called neurogenic niches. One of the main neurogenic niches is the ventricular-subventricular zone (V-SVZ) in which neural stem cells (NSCs) reside. NSCs produce neurons through the generation of transit amplifying progenitors and neuroblasts. Recent studies on adult neurogenesis revealed the existence of quiescent NSCs (qNSCs, also called B1) and activated NSCs (aNSCs). The current model of adult neurogenesis places qNSCs (B1) upstream of aNSCs in the lineage. The hypothesis is that the qNSC population constitutes a “reserve” pool to maintain aNSC pool throughout life. So far, the techniques used do not allow specific targeting of the qNSCs in vivo. Therefore, it is not possible to analyze their biological properties, activation mechanisms and relationship with aNSCs. This understanding is also necessary to establish therapeutic strategies that could utilize the potential of stem cells to restore neurogenesis in contexts of aging and neurodegenerative diseases. In order to characterize qNSCs in the ventricular zone, we took advantage of plasmid electroporation in a reporter mouse model, Rosa26-stop-EYFP. In this model, the sequence coding for EYFP preceded by a floxed STOP codon is inserted at the Rosa26 locus. Excision of the STOP codon by a recombinase enables expression of the reporter in electroporated cells and their progeny. This technique enabled the specific targeting of a ventricle-contacting astrocytes population and to study their contribution to adult neurogenesis. Unlike transgenic or viral approaches, electroporation can target quiescent cells and the expression of the plasmid is restricted to the ventricle-contacted cells. Using this approach, we made surprising observations: i) this population is mostly quiescent and only minimally contributes to adult neurogenesis, ii) this population does not participate in niche regeneration in vivo and iv) their neurogenic output can be increased by expressing the pro-neural gene Mash1. Next, we investigated the role of EGFR signaling in the regulation neural stem and progenitor cells (NSPCs) activity. In this second study, we show that i) EGFR signaling decreases during aging, ii) PI3K/AKT, MEK/ERK and mTOR exert different regulation proliferation, differentiation and survival of NSPCs and iii) activation of EGFR in the qNSCs increases V-SVZ neurogenesis in 3-months-old animals but not in 6-months-old or Alzheimer’s disease model animals. Our data suggests that the NSC population is heterogeneous, with variable neurogenic output from the different sub-populations, as well as different activation modalities. We also showed that EGFR-associated signalling pathways differentially regulate NSPCs activity. Finally, our results indicate that the factors present in the V-SVZ niche during aging dominantly inhibit activation of NSCs.

Page generated in 0.0534 seconds