• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 33
  • 8
  • 7
  • 6
  • 5
  • 2
  • 1
  • Tagged with
  • 68
  • 68
  • 23
  • 21
  • 18
  • 11
  • 11
  • 11
  • 10
  • 9
  • 9
  • 9
  • 9
  • 8
  • 8
  • 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.
61

Rôle des microARN dans la différenciation de l'épithélium respiratoire humain : caractérisation de miR-449 comme acteur central de la multiciliogenèse conservé chez les vertébrés / Role of microRNAs in human airway epithelium differentiation : characterization of miR-449 as a central player in multiciliogenesis conserved in vertebrates

Chevalier, Benoît 17 December 2013 (has links)
Chez les vertébrés, le battement coordonné des cils motiles présents par centaines à la surface apicale des cellules multiciliées (MCC) est requis pour propulser directionnellement les fluides biologiques à l’intérieur de certains organes (voies respiratoires, ventricules cérébraux, trompes utérines ou certaines structures embryonnaires). De nombreuses pathologies humaines sont associées à des défauts ciliaires ou à une perte des MCC (dyskinésies ciliaires, mucoviscidose, asthme,...). Dans ce contexte, mon travail de thèse a consisté à élucider les mécanismes complexes contrôlant la différenciation des MCC et donc la formation des cils motiles (multiciliogenèse). Par des approches de génomiques fonctionnelles à partir de deux modèles d’épithéliums multiciliés évolutivement éloignés (épithélium respiratoire humain et épiderme d’embryon de Xénope) nous avons identifié la famille des microARN (petits ARN non-codants régulateurs de l’expression génique) miR-449 comme majoritairement exprimée dans les MCC. Nous avons montré que miR-449 contrôle la multiciliogenèse i) en bloquant le cycle cellulaire, ii) en réprimant directement la voie de signalisation Notch et iii) en inhibant l’expression de la petite GTPase R-Ras. Enfin, nos travaux montrent que l’ensemble de ces mécanismes est conservé chez les vertébrés. En conclusion, miR-449 est un nouveau régulateur clé de la multiciliogenèse conservé au cours de l’évolution. Nos résultats pourraient ouvrir la voie à de nouvelles stratégies thérapeutiques utilisant des petits ARN régulateurs dans le traitement de certaines pathologies associées à des défauts ciliaires. / In vertebrates, the coordinated beating of hundreds of motile cilia present at the apical surface of multiciliated cells (MCC) is required for propel directionally flow of biological fluids inside some organs (airways, cerebral ventricles, fallopian tubes or some embryonic structures). Many human diseases are associated with ciliary defects or loss of MCC (ciliary dyskinesia, cystic fibrosis, asthma ...). In this context, my thesis has sought to elucidate the complex mechanisms that control the differentiation of MCC and thus the formation of motile cilia (multiciliogenesis). By functional genomic approaches from two evolutionarily distant models of multiciliated epithelia (human respiratory epithelium and epidermis of Xenopus embryo) we identified the miR-449 family of microRNAs (small non-coding RNAs regulating gene expression) as mainly expressed in MCC. Then, we showed that miR-449 controlled multiciliogenesis by i) blocking the cell cycle ii) directly suppressing the Notch pathway and iii) by inhibiting the expression of the small GTPase R-Ras. Finally, we have demonstrated that all these mechanisms were conserved in vertebrates. In conclusion, miR-449 is a new key and conserved regulator of multiciliogenesis. Our findings could pave the way for new therapeutic strategies using small regulatory RNAs in the treatment of several diseases associated with ciliary defects.
62

Rôle de la voie de signalisation Notch dans la réponse lymphocytaire T CD8 suite à une infection aiguë ou chronique

Duval, Frédéric 12 1900 (has links)
No description available.
63

Rôle(s) de la protéine O-fucosyltransférase 1 au cours de la différenciation myogénique / Role(s) of protein O-fucosyltransferase 1 during myogenic differentiation

Der Vartanian, Audrey 11 February 2015 (has links)
Au cours de la myogenèse post-natale, la voie de signalisation de Notch participe au développement et à la régénération du muscle squelettique chez les mammifères. Elle permet le maintien de l'état prolifératif des myoblastes, contrôle la quiescence des cellules satellites in vivo et préserve une sous-population de cellules de réserve indifférenciées in vitro. L' activation de la voie et l'interaction du récepteur Notch avec ses ligands est dépendante de leur entité glucidique, notamment de leurs O-fucosylglycannes. La synthèse de ces derniers est initiée par la protéine O-fucosyltransférase 1 (Pofut1) qui greffe un O-fucose sur des domaines peptidiques particuliers appelés EGF-like. Bien que les acteurs moléculaires de la différenciation myogénique aient été largement étudiés par la communauté scientifique, la contribution de la glycosylation des protéines dans ce processus reste peu documentée. Une approche expérimentale in vitro basée sur l'utilisation de la lignée myoblastique murine C2C12 nous a permis d'identifier une expression importante de Pofut1 dans les cellules de réserve tandis qu' elle est restreinte dans les myotubes durant la différenciation myogénique. Plusieurs lignées de cellules C2C12 ont été générées pour qu' elles expriment de manière stable et différentielle Pofut1. Elles permettent ainsi d' évaluer l' importance du niveau d' expression de Pofut1 sur la différenciation myogénique.La sous-expression de Pofut1 réduit l' activation de la voie de signalisation de Notch conduisant à une entrée précoce des myoblastes dans le programme myogénique. Ceci a pour conséquence la dépletion des cellules de réserve Pax7+/MyoD- au profit d' une augmentation du nombre de myotubes. Des études morphométriques ont révélé un défaut d' accrétion nucléaire dans les myotubes sous-exprimant Pofut1, caractéristique d' une altération de la fusion secondaire. Ces observations sont accompagnées d' une diminution significative de l' expression du récepteur à l' interleukine 4 dans les cellules de reserve sous-exprimant Pofut1. Les lignées cellulaires ré-exprimant Pofut1 présentent une activation de la voie de signalisation de Notch et un processus de fusion myoblastique correctement restaurés.Ces travaux de thèse ont mis en exergue pour la première fois le rôle essentiel de Pofut1 dans le devenir cellulaire et la fusion des myoblastes au cours de la différenciation myogénique. / During post-natal myogenesis, Notch signaling pathway is involved in the development and regeneration of skeletal muscle in mammals. It maintains progenitor cell properties during the development of the myogenic lineage and controls the transition of satellite cells from a quiescent to an active state and preserves a subpopulation of reserve cells, in cell culture, in an undifferentiated state. The interaction between Notch and its ligands and the activation of this signaling is mainly controlled by the activity of protein O-fucosyltranferase 1 (Pofut1) and thus by the O-fucosylation state of the EGF-like repeats.Although the molecular players in myogenic differentiation have been extensively studied by the scientific community, the contribution of glycosylated proteins in this process remains poorly documented. An experimental in vitro study based on the C2C12 mouse myoblast cell line allowed us to identify a high expression of Pofut1 in reserve cells while a low expression was found in myotubes during myogenic differentiation. Several C2C12 cell lines were generated to express Pofut1 at different levels. They were used to evaluate the contribution of Pofut1 expression to the myogenic differentiation.The knockdown of Pofut1 repressed Notch signaling pathway activation leading to an earlier entrance of myoblasts in myogenic program. This resulted in the depletion of reserve cells Pax7+/MyoD- and an increase in the number of myotubes. Morphometric analysis revealed a nuclear accretion defect in Pofut1 knockdown myotubes. A significant decrease in the expression of the interleukin-4 receptor in Pofut1 knockdown reserve cells was also observed. Cell lines re-expressing correctly Pofut1 restored Notch signaling pathway and subsequently myoblast fusion process.This thesis work highlights, for the first time, the crucial role of Pofut1 in the cell fate decision and the fusion of myoblasts during myogenic differentiation.
64

Contribution de la protéine O-fucosyltransférase 1( POFUT1) à la différenciation myogénique et à la tumorigenèse colorectale / Contribution of O-fucosyltransferase 1 (POFUT1) protein to myogenic differentiation and colorectal tumorigenesis

Chabanais, Julien 06 December 2019 (has links)
La protéine O-fucosyltransférase 1 (POFUT1) réticulaire, dont le gène est localisé dans la région chromosomique 20q11.21 chez l’Homme, catalyse le transfert d’un fucose qui sera O-lié sur la sérine ou la thréonine présente dans la séquence consensus (C2X4S/TC3), portée par un domaine EGF-like d’une glycoprotéine membranaire ou sécrétée. Le knockdown de Pofut1 (Po -) dans la lignée myoblastique murines C2C12 conduit à la formation de myotubes allongés et minces, à faible nombre de noyaux ainsi qu’à une sous-expression du marqueur myogénique tardif Myf6, suggérant des défauts significatifs dans la fusion secondaire. La signalisation NFATc2/IL-4 est décrite comme la voie principale associée à cette étape. Nous montrons que la moindre expression de Nfatc2 dans les myotubes Po - est corrélée à une baisse de l'IL-4 sécrétée et à une faible quantité de son récepteur (IL-4Rα) présent chez les cellules de réserve qui doivent participer à la fusion avec les myotubes naissants. La neutralisation de l’IL-4Rα sur les C2C12 sauvages provoque des défauts d'accrétion myonucléaire, semblables à ceux observés pour les Po -. Ainsi, POFUT1 pourrait être un nouveau médiateur de la croissance des myotubes au cours du processus myogénique, notamment par la signalisation NFATc2/IL-4. La glycoprotéine WIF1, cible potentielle de POFUT1, est un antagoniste de la signalisation WNT via sa fixation aux protéines WNT. Cette voie est connue pour être impliquée dans la prolifération et la différenciation des myoblastes. Néanmoins, aucune donnée ne concerne le rôle de WIF1 dans le processus myogénique. Par un apport exogène de WIF1, nous avons montré l’augmentation de la prolifération et l’altération de la différenciation myoblastique des C2C12. Lors de la prolifération, une augmentation de l’expression de Myf5 et une diminution de MyoG sont observées. A 7 jours de différenciation, les myotubes Po - ont un diamètre plus petit que les myotubes sauvages et ils sont plus nombreux à avoir un faible nombre de noyaux, traduisant des défauts de fusion. Nous démontrons pour la première fois, l’implication de la protéine WIF1 dans le processus myogénique. Récemment, POFUT1 a aussi été proposé comme nouveau biomarqueur pour certains cancers, mais pas évalué dans le cancer colorectal (CCR). Nous avons donc collecté des données issues de 626 tumeurs et 51 tissus adjacents non tumoraux disponibles dans FireBrowse, celles de lignées cellulaires cancéreuses colorectales et de prélèvements tumoraux provenant du Centre de Ressources Biologiques du CHU de Limoges. Une surexpression de POFUT1 est observée dès le stade I, majoritairement due à une amplification de la région 20q11.21. Elle est significativement associée aux adénocarcinomes non mucineux et à une localisation rectale. De plus, l’expression de POFUT1 est corrélée à celles des récepteurs NOTCH ainsi qu’au processus métastatique, probablement par activation de la voie NOTCH. A ce titre, POFUT1 pourrait être considéré comme un nouveau biomarqueur pour le diagnostic du CCR. / The ER protein O-fucosyltransferase 1 (POFUT1), whose gene is located at the 20q11.21 chromosomic region in humans, catalyzes O-linked fucose addition to serine or threonine present in the consensus sequence (C2X4S/TC3) carried by EGF-like domain of membrane or secreted glycoprotein. Pofut1 knockdown (Po -) in murine myoblast C2C12 cell line leads to formation of elongated and thin myotubes, with a low number of nuclei and to downexpression of the late myogenic marker Myf6, suggesting significant defects in secondary fusion. NFATc2/IL-4 signaling is described as the main pathway associated to this step. We showed that the slightest expression of Nfatc2 in Po - myotubes is correlated with a decrease in IL-4 secretion and a lower quantity of IL 4Rα in reserve cells, which had to fuse with nascent myotubes. IL-4Rα neutralization on wild-type C2C12 causes myonuclear accretion defects, similar to those observed in Po -. Then, POFUT1 could be a new mediator of myotube growth during myogenic process, particularly through NFATc2/IL-4 signaling. The glycoprotein WIF1, potential POFUT1 target, is an antagonist of WNT signaling via its binding to WNT proteins. This pathway is involved in proliferation and differentiation of myoblasts. However, no data are available on WIF1 role in the myogenic process. Through exogenous WIF1 treatment, we showed a proliferation increase and a myoblast differentiation impairment in C2C12. During proliferation, increase in Myf5 and decrease in MyoG expressions are observed. At 7 days of differentiation, Po - myotubes have a smaller diameter than wild-type ones and are more numerous to have a small number of nuclei, reflecting fusion defects. For the first time, we demonstrate the involvement of WIF1 in the myogenic process. Recently, POFUT1 was proposed to be a new biomarker for several cancers, but not evaluated in colorectal cancer (CRC). We used data from 626 tumors and 51 adjacent non-tumor tissues available at FireBrowse, colorectal cancer cell lines and tumor samples from the Biological Resource Centre of Limoges hospital. A POFUT1 overexpression is observed from stage I, mainly due to amplification of 20q11.21 region. It is significantly associated to non-mucinous adenocarcinoma and to rectum location. Moreover, POFUT1 expression is correlated with those of NOTCH receptors as well as the metastatic process, probably by activation of the NOTCH pathway. POFUT1 could therefore be considered as a new biomarker for CRC diagnosis.
65

Rôle de la voie de signalisation Notch dans la différenciation des lymphocytes T CD8

Ennajimi, Myriam 12 1900 (has links)
Au pic de la réponse effectrice des LT CD8, on retrouve deux sous-populations, soit les effecteurs à demi-vie courte (SLEC) ou les effecteurs précurseurs de cellules mémoires (MPEC). La phase de contraction de la réponse effectrice implique l’apoptose des SLEC et la survie des MPEC, qui se différencient en cellules mémoires pour protéger contre une réinfection. La voie Notch est impliquée dans les choix de différenciation binaire et l’interaction ligand-récepteur mène au clivage du domaine intracellulaire de Notch (NICD), qui migrera au noyau afin d'induire l’expression de gènes cibles. Dans un modèle où l’expression des récepteurs Notch1 et Notch2 est absente uniquement dans les LT CD8 (N1N2∆/∆), le laboratoire a démontré que l’absence du signal Notch favorisait la différenciation en MPEC et affectait l’expression de 217 gènes. Cette étude visait à 1) identifier les gènes cibles de Notch contrôlant la différenciation SLEC-MPEC, et 2) évaluer si l’absence du signal Notch permet un meilleur contrôle tumoral par les LT CD8. Nous avons priorisé les 217 gènes en fonction de différents critères et identifié Il2ra comme une cible importante en aval de la voie Notch. Toutefois, nous avons établis que lors d’une infection aiguë, la surexpression rétrovirale de Il2ra dans les LT CD8 N1N2∆/∆ n’influençait pas la différenciation SLEC-MPEC. Nous avons également déterminé qu’une thérapie adoptive de LT CD8 N1N2∆/∆ limitait le contrôle de la croissance tumorale et impliquait une diminution des fonctions effectrices des LT CD8 N1N2∆/∆, qui étaient moins terminalement différenciés. Une meilleure compréhension du rôle de Notch dans la réponse des LT CD8 permettra de développer de nouvelles stratégies de vaccination et de traitement du cancer. / In response to acute infections, effector CD8 T cells differentiate into short-lived effector cells (SLECs) and memory precursor effector cells (MPECs) capable of generating long-lived memory CD8 T cells. The Notch signaling pathway is a key regulator of cell fate decision. Following ligand- receptor interaction, the Notch intracellular domain (NICD) is cleaved and migrates to the nucleus in order to induce the expression of target genes. In a model in which Notch1 and Notch2 expression is inhibited only in mature CD8 T cells, our team has established that Notch deficiency favors MPEC differentiation in CD8 T cells and influences the expression of 217 gens. This study aims to: 1) identify target genes of the Notch pathway regulating SLEC-MPEC differentiation, 2) evaluate if Notch deficiency can augment tumor control by CD8 T cells. We have prioritized the list of genes differentially expressed in the absence of Notch signaling according to various criteria. We hence identified Il2ra as a target gene, but during an acute infection, overexpression of Il2ra in Notch deficient CD8 T cell was insufficient to modulate SLEC-MPEC differentiation. In addition, we have established that adoptive therapy with Notch deficient CD8 T cell impaired tumor control and implicated a diminution of effector function in Notch deficient CD8 T cell, which were less terminally differentiated. A better understanding of Notch signaling pathway’s role in the CD8 T cell response will allow for improvement of vaccinal strategies and cancer treatment.
66

Unfolding the Mechanism of Notch1 Receptor Activation : Implications in Cancer Stem Cell Targeting

Sharma, Ankur January 2013 (has links) (PDF)
Notch receptors and ligands are single-pass transmembrane proteins which play important roles in cell-cell communication. Notch in ‘harmony’ with other signaling pathways regulate the entire diversity of metazoan life (Artavanis-Tsakonas & Muskavitch, 2010). These signaling pathways also play key roles in regulatingseveral developmental processes. Given the importance of Notch signaling in various developmental decisions, it is not surprising that aberrant gain or loss-of-function of Notch pathway leads to several human diseases including cancer (Ranganathan et al, 2011). Notch signaling has also been implicated in various human cancers, most notably in T-cell acute lymphoblastic leukemia (T-ALL) (Weng et al, 2004). In view of the importance of Notch signaling in cancers, therapeutic molecules targeting this pathway are making their way into clinical trials (Rizzo et al, 2008). This underscores the importance of understanding the mechanism of Notch receptor activation in normal and patho-physiological conditions. In this thesis, antibodies against different domains of human Notch1 receptor have been used as tools to understand the mechanism of receptor activation. This work has provided insights into the role of Notch1 extracellular domain in ligand-dependent receptor activation. Further, the mechanism of ligand-independent receptor activation in T-ALL associated mutant Notch1 has also been investigated. This understanding of ligand-dependent and independent receptor activation facilitated development of mechanistic inhibitors of Notch signaling for therapeutic targeting of the cancer stem cells (CSCs) across the pectrum of cancers. The thesis is divived into two parts. Part-I focuses on understanding the role of Notch1 extracellular domain in receptor-ligand interactions using antibodies as a tool. In part-II, implications of these antibodies in therapeutic targeting of CSCs has been investigated. Part-I Unfolding the Mechanism of Notch1 Receptor Activation The extracellular domain of Notch1 receptor consists of 36 EGF-like repeats that contribute to ligand binding (Kopan & Ilagan, 2009). Despite extensive studies on the downstream consequences of Notch signaling, the initial events of ligandreceptor interactions have not been clearly elucidated. In the absence of structural insights into the receptor-ligand interactions, it was important to decipher the roles of various receptor domains in ligand-binding and consequent signaling. In this study, antibodies have been employed as tools for in-depth analyses of Notch receptorligand, interactions. Studies in Drosophila Notch receptor suggest that EGF-like repeats 11-12 are necessary and sufficient for ligand binding (Rebay et al, 1991). However, the role of these repeats in human Notch1 receptor-ligand interaction(s) was not clearly elucidated. Antibodies were generated against Notch1 EGF-like repeats 11-15. Further, these antibodies were characterized for their specificity for Notch1 receptor in various ligand-binding and signaling assays. The results suggest that the monoclonal antibodies (MAbs) against EGF-like repeats 11-12 were more potent inhibitors of ligand-binding compared to the antibodies against EGF-like repeats 13-15. As a part of these investigations, the Notch ligands Jagged1 and Jagged2, Delta-like1 and Delta-like4 were purified and characterized in various assays. Ability of these ligands to interact with Notch1 EGF-like repeat 11-15 was determined using Surface Plasmon Resonance. The Jagged family of ligands demonstrated higher affinity for this recept or fragment when compared to the Delta family of ligands. The relatively low affinities (μM) of all the ligands suggested possibile involvement of other EGF-like repeats in ligand-binding. This was further investigated using antibodies against other EGF-like repeats of Notch1. In Drosophila Notch EGF-like repeats 24-29 have been implicated in the ligand-dependent gain-of-function phenotype, suggesting a plausible involvement of this region in receptor activation (Pei & Baker, 2008). Therefore, role of human Notch1 EGF-like repeats 21-30 in ligand-binding and signaling was investigated. These EGF-like repeats demonstrated specific interaction with the ligand-binding domain (EGF-like repeats 11-15). This suggested that in the absence of the ligand, these inter-domain interactions keep the receptor in an auto-inhibited conformation. Further, ligand binding to EGF-like repeats 11-15 dissociated pre-formed interdomain interactions. These results suggested that, the binding of ligand to EGF-like repeat 11-12 overcomes the negative constraint imposed by the intra-domain interactions which might lead to receptor activation. Next, to understand the role of EGF-like repeats 21-30 in ligand binding, polyclonal antibodies were generated against the same and extensively characterized in various solid-phase and cell-based assays. These antibodies demonstrated partial inhibition of ligand-binding. Further, using immunoaffinity purified antibodies it was demonstrated that antibodies against EGF-like repeats 25-26 were most potent inhibitors of ligand-binding compared to antibodies against EGF-like repeats 21-24 and 27-30. These results provided novel insights into Notch1 receptor activation. The model proposed on the basis of these results suggested that ligand-binding to EGF-like repeats 11-12 competes with the inter-domain interaction, in turn dissociating EGF-like repeats 21-30 from the ligandbinding domain. It emerged that this altered conformation of the receptor creates a secondary ligand-binding site at EFG-like repeats 25-26. Overall these results provided novel insight into the mechanism of Notch receptor-ligand interaction(s). Part-II Implication in Cancer Stem Cell Targeting Recent studies have suggested existence of the CSC population in various cancers (Clevers, 2011). Notch signaling plays an important role in maintenance of these CSCs (Pannuti et al, 2010). Thus, targeting Notch signaling may provide a potential therapeutic tool for CSC targeting. Several studies have indicated that Notch1 receptor and ligands are overexpressed in breast cancer cells compared to the normal breast epithelium (Mittal et al, 2009; Reedijk et al, 2005; Reedijk et al, 2008). Moreover, it has been suggested that Notch1 signaling plays a key role in breast carcinogenesis (Stylianou et al, 2006). Monoclonal antibodies (MAbs) were used as mechanistic inhibitors of aberrant Notch1 signaling for therapeutic targeting of CSCs. One such antibody, MAb 602.101, against Notch1 ligand-binding domain (EGF-like repeat 11-12) inhibited proliferation and depleted breast CSCs. This MAb also modulated genes associated with stemness and epithelial to mesenchymal transition (EMT). Furthermore, MAb 602.101 irreversibly inhibited the sphere-forming potential of breast cancer cells by modulating long-term self renewing capacity of breast CSCs. Inhibition of Notch1 signaling by the MAb also depleted the chemoresistant CD44Hi/CD24Low sub-population in breast cancer cells. Interestingly, antibody treatment led to elevated expression of genes associated with myoepithelial lineage, which suggested that inhibition of Notch1 signaling might induce a differentiation program leading to reduction in the CSC population. This study demonstrated the importance of Notch1 signaling in CSCs and effectiveness of antibodies as a tool for specific targeting of individual Notch receptors in cancer therapeutics. While aberrant expression of receptors and ligands leads to breast cancer (Reedijk et al, 2005), gain-of-function mutations are associated with 40-50% of TALL\ patients (Weng et al, 2004). These mutations lead to ligand-independent receptor activation (Malecki et al, 2006). Despite several attempts of successful antibodymediated therapeutic targeting of Notch1 (Aste-Amézaga et al, 2010; Wu et al, 2010), specific antibodies recognizing T-ALL associated mutant Notch1 remains elusive. Using homology modeling, the mutation induced conformational change in T-ALL associated mutant Notch1 was predicted. These results suggested that mutation led to conformational changes in the Notch1 negative regulatory region (NRR) This conformation change might result in the constitutive activation of Notch1 signaling leading to pathogenesis. Next, MAbs were generated against the wild-type Notch1 NRR and characterized in flow-cytometry based assays for identification of conformation specific antibodies. These antibodies were classified as either wild-type specific, mutant specific or unbiased to receptor conformations. One such mutant specific MAb 604.107 demonstrated higher binding to mutant Notch1 in flowcytometer and SPR based experiments. This MAb also demonstrated specific inhibition of T-ALL associated mutant Notch1 signaling without affecting the wildtype signaling. Moreover, antibody treatment also inhibited proliferation and depleted leukemia initiating sub-population in patient derived T-ALL cells. Taken together, this study provides a novel tool for specific targeting of mutant Notch1 receptors in TALL. CSCs are inherently chemo-resistant and lead to tumor relapse (Chen et al, 2012). Recent studies have demonstrated a strong correlation between Notch1 signaling in lung CSCs and chemotherapy resistance (Hassan et al, 2013). In this study, Notch1 heterogeneity in solid tumors viz. breast and colon cancers was investigated. Using the antibodies generated previously in this study, Notch1High and Notch1Low sub-populations from MDA-MB-231 (breast cancer) and HCT-116 (colon cancer) cell lines were flow-sorted. It was demonstrated that the Notch1High subpopulation represented the sphere-forming CSCs in breast and colon cancer. The Notch1High sub-population also demonstrated chemo-resistant properties and expressed higher level of EMT and stemness markers. These results suggested explicit involvement of Notch1 signaling in EMT and maintenance of CSCs subpopulation in these cancers. The anti-Notch1 MAb also inhibited proliferation of the chemo-resistant Notch1High sub-population. Further, treatment with MAb inhibited expression of ABCC1 transporters in these drug-resistant cells leading to augmentation of chemotherapeutic response. Using mouse xenograft assays, it was demonstrated that Notch1 signaling plays an important role in the maintenacne of tumor-initiating sub-population in breast and colon cancer cells. Prior exposure of breast and colon cancer cells to MAb inhibited the tumor forming potential of these cells in xenotransplantation assays. Treatment with MAb alone or in combination with chemotherapy led to regression of pre-formed tumors in breast and colon xenograft models. These results demonstrated existence of Notch1 heterogeneity in breast and colon cancer cells and emphasised the importance of targeting Notch1 signaling to overcome drug-resistance in these cancers. The results described above have provided important insights into Notch1 receptor activation and this understanding was translated into therapeutic targeting of CSCs. This “proof-of-principle” demonstration has significant mechanistic and applied implications in Notch and cancer biology.
67

Studying normal and cancer stem cells in the kidney using 3D organoids and genetic mouse models

Myszczyszyn, Adam 17 August 2021 (has links)
Organoide aus adulten Mäusen sind vielversprechende Modelle für die Nierenforschung. Ihre Charakterisierung wurde jedoch nicht auf ein zufriedenstellendes Niveau gebracht. Hier habe ich ein langfristiges 3D-Maus-Organoid (Tubuloid)-Modell etabliert und charakterisiert, das die Erneuerung und die Reparatur sowie die Architektur und die Funktionalität der adulten tubulären Epithelien rekapituliert. In der Zukunft wird das Modell detaillierte Untersuchungen der Trajektorien selbsterneuernder Zellen sowohl zur teilweisen Wiederherstellung der Niere als auch zur malignen Transformation der Niere ermöglichen. Das klarzellige Nierenzellkarzinom (ccRCC) ist der häufigste und aggressivste Nierenkrebs. Die Inaktivierung des Tumorsuppressorgens Von Hippel-Lindau (VHL) ist der Haupttreiber des ccRCCs. Zuvor hatten wir die Hochregulation der Wnt- und Notch-Signalübertragung in den CXCR4+MET+CD44+-Krebsstammzellen (CSC) aus primären humanen ccRCC-Tumoren identifiziert. Das Blockieren von Wnt und Notch in von Patienten stammenden Xenotransplantaten, Organoiden und nicht-anhaftenden Sphären unter Verwendung von niedermolekularen Inhibitoren beeinträchtigte die Selbsterneuerung der CSC und das Tumorwachstum. Um CSC-gesteuertes humanes ccRCC in genetischen Mausmodellen nachzuahmen, begann ich mit der Erzeugung von zwei Doppelmausmutanten; β-Catenin-GOF; Notch-GOF und Vhl-LOF; β-Catenin-GOF. Sowohl die β-Catenin-GOF; Notch-GOF Mausmutante als auch die Vhl-LOF; β-Catenin-GOF Mausmutante entwickelten innerhalb einiger Monate schwere Krankheitssymptome. Überraschenderweise beobachtete ich weder Tumore oder Tumorvorläuferläsionen noch höhere Zellproliferationsraten in den mutierten Nieren. Weitere Analysen ergaben, dass die Mausmutanten Merkmale chronischer Nierenerkrankung (CKD) aufwiesen. / Adult mouse organoids are promising models for kidney research. However, their characterization has not been pushed forward to a satisfying level. Here, I have generated and characterized a long-term 3D mouse organoid (tubuloid) model, which recapitulates renewal and repair, and the architecture and functionality of the adult tubular epithelia. In the future, the model will allow detailed investigations of trajectories of self-renewing cells towards both the partial recreation and malignant transformation of the kidney. Clear cell renal cell carcinoma (ccRCC) is the most common and aggressive kidney cancer. Inactivation of the Von Hippel-Lindau (VHL) tumor suppressor gene is the major driver of ccRCC. Earlier, we identified the upregulation of Wnt and Notch signaling in CXCR4+MET+CD44+ cancer stem cells (CSCs) from primary human ccRCCs. Blocking Wnt and Notch in patient-derived xenografts, organoids and non-adherent spheres using small-molecule inhibitors impaired self-renewal of CSCs and tumor growth. To mimic CSC-governed human ccRCC in genetic mouse models, I started from the generation of two double mouse mutants; β-catenin-GOF; Notch-GOF and Vhl-LOF; β-catenin-GOF. Surprizingly, I observed neither tumors or tumor precursor lesions nor higher cell proliferation rates in the mutant kidneys. Further analyses revealed that the mutant mice displayed features of chronic kidney disease (CKD). Thus, β-catenin-GOF; Notch-GOF and Vhl-LOF; β-catenin-GOF mouse mutants did not develop kidney tumors under the given experimental conditions.
68

Hand2 function within non-cardiomyocytes regulates cardiac morphogenesis and performance

VanDusen, Nathan J. January 2014 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / The heart is a complex organ that is composed of numerous cell types, which must integrate their programs for proper specification, differentiation, and cardiac morphogenesis. During cardiac development the basic helix-loop-helix transcription factor Hand2 is dynamically expressed within the endocardium and extra-cardiac lineages such as the epicardium, cardiac neural crest cells (cNCCs), and NCC derived components of the autonomic nervous system. To investigate Hand2 function within these populations we utilized multiple murine Hand2 Conditional Knockout (H2CKO) genetic models. These studies establish for the first time a functional requirement for Hand2 within the endocardium, as several distinct phenotypes including hypotrabeculation, tricuspid atresia, aberrant septation, and precocious coronary development are observed in endocardial H2CKOs. Molecular analyses reveal that endocardial Hand2 functions within the Notch signaling pathway to regulate expression of Nrg1, which encodes a crucial secreted growth factor. Furthermore, we demonstrate that Notch signaling regulates coronary angiogenesis via Hand2 mediated modulation of Vegf signaling. Hand2 is strongly expressed within midgestation NCC and endocardium derived cardiac cushion mesenchyme. To ascertain the function of Hand2 within these cells we employed the Periostin Cre (Postn-Cre), which marks cushion mesenchyme, a small subset of the epicardium, and components of the autonomic nervous system, to conditionally ablate Hand2. We find that Postn-Cre H2CKOs die shortly after birth despite a lack of cardiac structural defects. Gene expression analyses demonstrate that Postn-Cre ablates Hand2 from the adrenal medulla, causing downregulation of Dopamine Beta Hydroxylase (Dbh), a gene encoding a crucial catecholaminergic biosynthetic enzyme. Electrocardiograms demonstrate that 3-day postnatal Postn-Cre H2CKO pups exhibit significantly slower heart rates than control littermates. In conjunction with the aforementioned gene expression analyses, these results indicate that loss of Hand2 function within the adrenal medulla results in a catecholamine deficiency and subsequent heart failure.

Page generated in 0.0744 seconds