• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 5
  • 2
  • Tagged with
  • 5
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 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.
1

Implication de réseaux de récepteurs membranaires dans la signalisation du NGF et de son précurseur dans les cellules cancéreuses de sein / Involvement of membrane receptors networks in NGF and proNGF signaling in breast cancer cells

Aubert, Léo 08 December 2014 (has links)
Notre laboratoire a montré que le NGF et les neurotrophines sont des facteurs déterminants de la cancérogenèse mammaire, et plus particulièrement de la croissance et de l’invasion des cellules cancéreuses de sein. Les effets du NGF dépendent, entre autres, de l’activation du récepteur à activité tyrosine kinase TrkA. Récemment, nous avons décrit que le précurseur du NGF, le proNGF, est également produit et sécrété par les cellules cancéreuses de sein pour induire leur invasion via la formation d’un complexe sortiline/TrkA. Néanmoins, les différentes signalisations induites par le NGF et le proNGF ne sont pas encore parfaitement décryptées dans le cancer du sein. Au cours de ma thèse, j’ai démontré que le NGF et le proNGF induisent des complexes de récepteurs distincts. Ainsi, alors que le NGF conduit à la formation d’un complexe membranaire TrkA/CD44, le proNGF recrute quant à lui la sortiline, TrkA et EphA2. En outre, les études in vivo, réalisées dans un modèle de souris SCID, montrent que la croissance tumorale est dépendante de la formation de ces complexes. Ainsi, l’administration conjointe d’un inhibiteur de l’activité kinase de TrkA (CEP-701) et d’un ARN interférent dirigé soit contre CD44, soit contre EphA2, diminue significativement le volume tumoral en comparaison à l’utilisation en monothérapie du CEP-701 ou d’un siEphA2 ou du siCD44. L’ensemble de mes résultats a permis d’approfondir les mécanismes d’action du NGF et de son précurseur dans les cellules cancéreuses mammaires et suggère l’existence de mécanismes de résistance à travers les coopérations TrkA/CD44 et TrkA/EphA2, ouvrant ainsi l’opportunité à de nouvelles stratégies thérapeutiques dans les cancers. / We have shown that autocrine stimulation of breast tumor cells by NGF increased both cell growth and migration/invasion. These effects are mediated by TrkA, a receptor tyrosine kinases. Recently, it has been described that proNGF, the NGF precursor, is also produced and secreted by breast cancer cells to induce invasion through activation of sortilin/TrkA complex. Nevertheless, NGF and proNGF signaling are still unclear in breast cancer. During my thesis, I have demonstrated that the pro-invasive signaling of (pro)NGF requires the formation of NGF/TrkA/CD44 and proNGF/sortilin/TrkA/EphA2 complexes. I have showed that each of these receptor complexes leads to the induction of two separate transduction pathways, one dependent on TrkA phosphorylation, and the other independently. Furthermore, by using a SCID mouse xenograft model, we have demonstrated the involvement of TrkA/CD44 and TrkA/EphA2 complexes in tumor growth. Interestingly, the co-administration of an inhibitor of the TrkA kinase activity (CEP-701) and a siRNA directed either against CD44 or against EphA2, decreases significantly the tumor burden in comparison to the use of CEP-701, siEphA2 or siCD44 alone. Thus, our findings have improved the knowledge of NGF and proNGF signaling in breast cancer cells and suggest the presence of resistance mechanisms mediated by TrkA/CD44 and TrkA/EphA2 interactions, thereby giving the opportunity for new therapeutic modulations in breast cancer.
2

Impact de la transactivation des récepteurs membranaires par le (pro)NGF dans les cancers / Impact of (pro)NGF-dependent transactivation of cell membrane receptors in cancers

Guilbert, Matthieu 09 September 2015 (has links)
Le laboratoire INSERM U908 a montré que le Nerve Growth Factor (NGF) et son précurseur, le proNGF, sont impliqués dans l’agressivité des tumeurs mammaires via des effets sur la croissance, l’angiogenèse ou encore la migration/invasion. Pour autant, aucunes thérapies ciblées n’a à ce jour été approuvée suite à des essais cliniques de traitements visant à inhiber les effets du (pro)NGF et de leurs récepteurs dans les cancers. Ces résultats indiquent que la phosphorylation de TrkA est nécessaire mais pas suffisante pour expliquer le(s) mécanisme(s) par lequel(s) le NGF ou son précurseur participe(nt) au développement tumoral. L’obtention de lignées tumorales résistantes au lestaurtinib est un phénomène rapide, nous avons donc émis l’hypothèse que la signalisation de TrkA indépendante de sa phosphorylation existe de façon « innée » dans les cellules tumorales. Par l’analyse protéomique, nous avons ainsi découvert que le NGF induit le recrutement de CD44 et d’une cascade de signalisation p115RhoGEF/RhoA/ROCK1 (Aubert L*, Guilbert M* et al, Oncotarget, 2015 ; *co-premier auteur). Nous avons ainsi montré que le NGF via le complexe TrkA/CD44 induit la migration et l’invasion des cellules cancéreuses de sein in vivo, et augmente la tumorigénicité in vivo. Quant au proNGF, j’ai pu observer qu’il induit l’internalisation de l’EGFR par la phosphorylation du résidu Y1068. Il en résulte une diminution des effets prolifératif et pro-invasif de l’EGF dans les cellules tumorales. Ces résultats fondamentaux sont tout à fait intéressants même s’ils nécessitent leurs consolidations et doivent permettre de démontrer le caractère pronostic de la détection de TrkA et ses corécepteurs dans le cancer. Ainsi nos études permettront le développement de thérapie ciblée par des firmes pharmaceutiques. / The INSERM U908 unit has showed that Nerve Growth factor (NGF) and its precursor (ProNGF) are implicated in tumor agressivness via their effects on growth, angiogenesis or migration/invasion and metastasis. Nevertheless, (pro)NGF and their receptors targeted therapies failed to demonstrate efficiency and clinical trial are disappointing. These results indicate that TrkA phosphorylation is not sufficient to explain molecular mechanisms of (pro)NGF effects on tumors. Indeed, we obtained lestaurtinib resistant cell lines within 3 weeks of treatment which indicated that resistant mechanisms are innate. So, by functional proteomics analyses, we described that NGF induced the formation of TrkA/CD44 complex and then the recruitment of p115RhoGEF/RhoA/ROCK1 signalling cascade (Aubert L*, Guilbert M* et al, Oncotarget, 2015 ; *equally contributed to this work). We showed that CD44 mediated effects participate to invasive effects of NGF in vitro, and in vivo, we demontrated that CD44 increases NGF induced tumoriginicity. In a second part, I observed that the proNGF regulated EGFR turn over through its phosphorylation on Y1068. This effect on EGFR decreased proliferative and pro-invasive effects of EGF in cancer cells. These fundamental results are interesting and need to be consolidated to ensure development of prognosis or targeted therapies.
3

Biological Activity of a Neurotrophin Precursor and Mechanism of Neurotrophin Dysregulation in Neurodenerative Diseases

Masoudi, Raheleh 09 1900 (has links)
Neurotrophins, such as nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF), are key factors in neuronal survival and function. In Alzheimer's disease (AD), there is a change in the normal level of these neurotrophins and their precursors (proNGF and proBDNF). The mechanism/s underlying changes in the levels of these neurotrophins in AD is not fully understood. According to the amyloid cascade hypothesis, amyloid-β is the original insult in AD and tau pathology is a downstream event. Amyloid-β interferes with axonal transport and reduces BDNF levels. However, it is not clear if amyloid-β affects neurotrophin levels directly or through tau hyperphosphorylation. If tau is responsible for changes in the level of neurotrophins in AD, we expect to observe the same alteration in neurotrophin levels in other diseases with tau dysfunction such as tauopathies. We tested the levels of BDNF mRNA and proNGF protein in subjects with tauopathies. We observed significant decrease in BDNF mRNA levels in subjects with Corticobasal degeneration. Our result suggests that BDNF may be down-regulated by tau hyperphosphorylation. Moreover, we showed that there was a significant increase in the level of proNGF in Pick's disease (PiD). Interestingly, AD and PiD share common tau modifications. Our result demonstrates a role for tau dysfunction in changes in the level of proNGF. Therefore, study of the levels of NGF and BDNF in non-AD tauopathies has shed light on the mechanisms underlying neurotrophin dysregulation in AD. How do increased levels of proNGF impact the brain in AD or PiD? Is neuronal degeneration in AD or PiD due to the lack of neurotrophic support of proNGF or do increased levels of apoptotic proNGF cause neurodegeneration? Lee et al. (2001b) and Fahnestock et al. (2004a) produced two different cleavage-resistant proNGFs with opposite activities (apoptotic versus neurotrophic). Structural and procedural differences between the two cleavage resistant proNGFs and different bioassays can cause opposite activities. We showed that proNGF from Lee's lab was neurotrophic when it was expressed in the expression system used by Fahnestock et al. or when it was purified. ProNGF expressed in a different expression system was also neurotrophic. ProNGF was neurotrophic in all bioassays except the serum withdrawal assay. We conclude that proNGF is normally neurotrophic but may be apoptotic when cell survival is already compromised. We propose that in AD, cells undergo degeneration due to the lack of neurotrophic support of proNGF (impaired transport). Moreover, TrkA is downregulated in AD which compromises cell survival and may lead to apoptosis induced by increased levels of proNGF. / Thesis / Doctor of Philosophy (PhD)
4

THE EFFECTS OF AGING AND ALZHEIMER’S DISEASE ON RETROGRADE NEUROTROPHIN TRANSPORT IN BASAL FOREBRAIN CHOLINERGIC NEURONS / RETROGRADE NEUROTROPHIN TRANSPORT IN BASAL FOREBRIAN NEURONS

Shekari, Arman January 2021 (has links)
Basal forebrain cholinergic neurons (BFCNs) are critical for learning and memory. Profound and early BFCN degeneration is a hallmark of aging and Alzheimer’s disease (AD). BFCNs depend for their survival on the retrograde axonal transport of neurotrophins, proteins critical for neuronal function. Neurotrophins like brain derived neurotrophic factor (BDNF) and pro-nerve growth factor (proNGF) are retrogradely transported to BFCNs from their synaptic targets. In AD, neurotrophin levels are increased within BFCN target areas and reduced in the basal forebrain, implicating dysfunctional neurotrophin transport in AD pathogenesis. However, neurotrophin transport within this highly susceptible neuronal population is currently poorly understood. We began by establishing protocols for the accurate quantification of axonal transport in BFCNs using microfluidic culture. We then determined the effect of age on neurotrophin transport. BFCNs were left in culture for up to 3 weeks to model aging in vitro. BFCNs initially displayed robust neurotrophin transport, which diminished with in vitro age. We observed that the levels of proNGF receptor tropomyosin-related kinase-A (TrkA) were reduced in aged neurons. Additionally, neurotrophin transport in BFCNs derived from 3xTg-AD mice, an AD model, was also impaired. Next, we sought to determine a mechanism for these transport deficits. First, we determined that proNGF transport was solely contingent upon the levels of TrkA. We then found that elevation of oxidative stress, an established AD contributor, significantly reduced both TrkA levels and proNGF retrograde transport. TrkA levels are partially regulated by protein tyrosine phosphatase-1B (PTP1B), an enzyme whose activity is reduced by oxidation. PTP1B antagonism significantly reduced TrkA levels and proNGF retrograde transport in BFCNs. Treatment of BFCNs with PTP1B-activating antioxidants rescued TrkA levels, proNGF transport, and proNGF-mediated axonal degeneration. Our results suggest that oxidative stress contributes to BFCN degeneration in aging and AD by impairing retrograde neurotrophin transport via oxidative PTP1B-mediated TrkA loss. / Thesis / Doctor of Philosophy (PhD) / During aging and Alzheimer’s disease (AD), the connections between neurons, a type of brain cell, break down, causing memory loss. This breakdown begins in a brain area called the basal forebrain. Basal forebrain neurons rely upon the transport of nutrients along their connections with other neurons, called axons, for proper function. This transport process becomes impaired in AD. Our goal was to understand why this happens. First, we determined that axonal transport was impaired with age and in basal forebrain neurons of mice genetically predisposed to develop AD. We recreated these impairments by increasing the levels of harmful molecules called reactive oxidative species (ROS). ROS levels increase with age and become abnormally high during AD. We found that increased ROS impair axonal transport and contribute to the breakdown of basal forebrain neurons. Our work suggests that reducing ROS will help prevent the breakdown of basal forebrain neurons in AD.
5

ALTERED NEUROTROPHIN EXPRESSION IN AGED PERIPHERAL NEURONS AND TARGETS

Bierl, Michael A. 13 July 2005 (has links)
No description available.

Page generated in 0.0237 seconds