• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 8
  • 2
  • 1
  • Tagged with
  • 19
  • 19
  • 19
  • 7
  • 6
  • 5
  • 5
  • 5
  • 3
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 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

The role of growth factors in ruminant mammary development

Winder, S. J. January 1987 (has links)
No description available.
2

A mutagenic study of functional and structural aspects of rat insulin-like growth factor binding protein-5

Song, Hyuk January 2001 (has links)
No description available.
3

The role of β1-integrin in normal and oncogene-mediated proliferation in breast epithelia

Moreno Layseca, Paulina January 2015 (has links)
Luminal epithelial cells in the mammary gland require two types of signals to proliferate: soluble signals (growth factor signals) and signals from the extracellular matrix (ECM). The composition of the ECM is sensed by adhesion receptors such as integrins. Integrins modulate cell behaviour and play a key role in cell cycle entry. Altered integrin expression and signalling has been associated with breast cancer and studies using mouse mammary epithelial cells (MECs) have shown that the absence of β1-integrin induces growth arrest. However, it is not completely understood how integrins transduce the signals from the plasma membrane to the nucleus to induce cell cycle entry. Thus, the first aim of this project was to determine how β1-integrin controls proliferation in MECs. I established a model to study the effects of depleting β1-integrin using the FSK7 mammary epithelial cell line. The proliferation defect observed in this β1-integrin knockdown model was rescued by expressing a constitutively active Rac1 or Pak. Moreover, inhibiting Rac1 or Pak prevented normal proliferation in MECs in a similar fashion as β1-integrin depletion. Furthermore, in this thesis I have identified the complex comprised of Src, paxillin and p130Cas as a potential link between β1-integrin and Rac1. These results provide an insight into the mechanism that regulates proliferation downstream of β1-integrin. During breast cancer initiation, β1-integrin signals are disrupted. This indicates that additional signals must be driving proliferation during tumorigenesis. Therefore, the second aim of this project was to test whether expression of breast oncogenes can overcome the proliferation defect present in β1-integrin null cells. In order to do so, an oncogenic ErbB2, a constitutively active form of Akt (myrAkt) and the Notch1 intracellular domain (NICD) were transfected in the β1-integrin knockdown MECs. The results showed that ErbB2 overcomes the need for β1-integrin by signalling to Pak. NICD does not require β1-integrin to drive proliferation by an unknown mechanism. Expression of myrAkt did not restore normal levels of proliferation in β1-integrin depleted MECs. This finding suggests that Akt is not sufficient to induce cell cycle entry by itself and instead, both Akt and Erk signalling are needed to exert this function. This work has further delineated the specific signals controlling proliferation downstream of β1-integrin, and has provided a model to test the dependence of oncogenes for β1-integrin to drive proliferation in MECs. These studies are important to understand the role of β1-integrin in breast cancer formation and to define the types of breast cancer where β1-integrin can be used as an effective therapeutic target.
4

ABCB5 and the regulation of p16INK4a by non-coding RNA

Braker, Paul January 2014 (has links)
p16INK4a (p16) traps the cell at the restriction point of the cell cycle by binding to cyclin-dependent kinase 4/6 thus preventing the phosphorylation of the retinoblastoma protein (pRB). As p16 accumulates the cell stops dividing and becomes senescent. This study investigates the modulation of p16 function by the putative membrane protein ABCB5 and a group of five putative oncogenic microRNAs (oncomiRs). ABCB5 is a poorly characterised member of the B-subfamily of human ATP Binding Cassette transporters. ABCB5 is reportedly transcribed into four transcripts, one of which could potentially encode a full-length transporter (ABCB5fl) whilst a second could encode a half-transporter (ABCB5β). The other two transcripts (ABCB5α and ABCB5γ) could only encode short polypeptides. Exogenous expression of ABCB5fl and ABCB5β was achieved in HEK293T cells, but the recombinant protein expressed poorly and localised to the endoplasmic reticulum. Point mutations introduced into the ATP catalytic domain failed to improve expression levels suggesting that protein function was not deleterious to the cell. Exogenous expression in HEK293T cells also allowed commercial antibodies purportedly raised against ABCB5 isoforms to be tested. Several were found not to recognise ABCB5 necessitating re-interpretation of published data. However, one antibody recognised both ABCB5fl and ABCB5β, and was subsequently used to evaluate protein expression levels in other cell types.siRNA knockdown of ABCB5 in human mammary epithelial cells (HMECs) caused a concomitant reduction in p16 expression and an increase in cellular proliferation. Differential siRNAs and RT-qPCR analyses demonstrated ABCB5β to be the relevant transcript with respect to the reduction in p16 expression; however, no native ABCB5β protein was detected in HMECs. Together these data lead to the hypothesis that the ABCB5β transcript may act as a long noncoding RNA to regulate p16. Exogenous expression of each of five distinct putative oncomiRs in HMECs was found to increase cellular proliferation and, surprisingly, increase p16 expression. These results mirror a phenotype commonly observed in p16-positive basal-like breast cancer (BLBC), an aggressive form of breast cancer with poor prognosis and few treatment options. Bioinformatic analysis of the predicted target genes for these oncomiRs identified multiple transcriptional regulators of pRB. These predictions, together with the work performed in a cellular model of p16-positive BLBC, suggest that the oncomiRs may cause unrestricted cell proliferation by indirectly reducing transcription of the pRB gene, RB1. In the absence of pRB, p16 expression is induced via a previously reported oncogeneinduced senescence-like positive feedback loop. These data, and previously published observations, suggest that a similar mechanism may explain the basis of p16-positive BLBC.
5

Epigenetic changes in breast cancer

Hinshelwood, Rebecca, Garvan Institute of Medical Research, UNSW January 2009 (has links)
Changes in the epigenetic landscape are widespread in neoplasia, with de novo methylation and histone repressive marks commonly occurring in association with gene silencing. However, understanding the dynamics of epigenetic changes is often hindered due to the absence of adequate in vitro model systems that accurately reflect events occurring in vivo. Human mammary epithelial cells (HMECs) grown under standard culture conditions enter a growth arrest termed selection, but a subpopulation is able to escape from arrest and continue to proliferate. These cells, called post-selection cells, have many of the hallmarks seen in the earliest lesions of breast cancer, including transcriptional silencing and hypermethylation of the p16INK4A tumour suppressor gene. The overall aim of my thesis was to use post-selection HMECs as model system to identify and dissect the mechanism involved in early epigenetic aberrations. Firstly, using a microarray approach, I found that multiple members of the TGF-β signalling pathway were concordantly suppressed in post-selection cells, and this was associated with functional disruption of the TGF-β pathway. Interestingly, concordant gene suppression was not associated with aberrant DNA methylation, but with repressive chromatin remodelling. Secondly, to further understand the mechanism underpinning epigenetic silencing, I demonstrated using laser capture technology, that p16INK4A silencing is a precursor to DNA methylation and histone remodelling. Thirdly, I found that individual post-selection HMEC strains during the early passages shared a common 'wave' pattern of regional-specific methylation within the p16INK4A CpG island. Interestingly, the 'wave' pattern of early de novo methylation correlated with the apparent footprint of nucleosomes within the p16INK4A CpG island. Lastly, to further characterise the properties of the HMEC culture system, I demonstrated that post-selection cells do not possess a natural tumour-inducing property when transplanted into the mammary fat pad of immunocompromised mice. However, post-selection HMECs were associated with high expression of a variety of stem/progenitor markers, as well as stem/progenitor associated polycomb genes, thus demonstrating that these cells share some common features of stem/progenitor cells. The research presented in this thesis demonstrate that epigenetic changes occur early in the growth of post-selection HMECs and many of these changes are common in breast cancer.
6

Epigenetic changes in breast cancer

Hinshelwood, Rebecca, Garvan Institute of Medical Research, UNSW January 2009 (has links)
Changes in the epigenetic landscape are widespread in neoplasia, with de novo methylation and histone repressive marks commonly occurring in association with gene silencing. However, understanding the dynamics of epigenetic changes is often hindered due to the absence of adequate in vitro model systems that accurately reflect events occurring in vivo. Human mammary epithelial cells (HMECs) grown under standard culture conditions enter a growth arrest termed selection, but a subpopulation is able to escape from arrest and continue to proliferate. These cells, called post-selection cells, have many of the hallmarks seen in the earliest lesions of breast cancer, including transcriptional silencing and hypermethylation of the p16INK4A tumour suppressor gene. The overall aim of my thesis was to use post-selection HMECs as model system to identify and dissect the mechanism involved in early epigenetic aberrations. Firstly, using a microarray approach, I found that multiple members of the TGF-β signalling pathway were concordantly suppressed in post-selection cells, and this was associated with functional disruption of the TGF-β pathway. Interestingly, concordant gene suppression was not associated with aberrant DNA methylation, but with repressive chromatin remodelling. Secondly, to further understand the mechanism underpinning epigenetic silencing, I demonstrated using laser capture technology, that p16INK4A silencing is a precursor to DNA methylation and histone remodelling. Thirdly, I found that individual post-selection HMEC strains during the early passages shared a common 'wave' pattern of regional-specific methylation within the p16INK4A CpG island. Interestingly, the 'wave' pattern of early de novo methylation correlated with the apparent footprint of nucleosomes within the p16INK4A CpG island. Lastly, to further characterise the properties of the HMEC culture system, I demonstrated that post-selection cells do not possess a natural tumour-inducing property when transplanted into the mammary fat pad of immunocompromised mice. However, post-selection HMECs were associated with high expression of a variety of stem/progenitor markers, as well as stem/progenitor associated polycomb genes, thus demonstrating that these cells share some common features of stem/progenitor cells. The research presented in this thesis demonstrate that epigenetic changes occur early in the growth of post-selection HMECs and many of these changes are common in breast cancer.
7

Regulation of tight junction proteins during engorgement of the mammary gland : a thesis presented in partial fulfilment of the requirements for the degree of Doctor of Philosophy in Animal Science at Massey University, Palmerston North, New Zealand

Cooper Phyn, Claire Vanessa January 2006 (has links)
Content removed due to copyright restriction: Appendix 6 Cooper, C. V., Stelwagen, K., Singh, K., Farr, V. C., Prosser, C. G., and Davis, S. R. (2004): Expression of the tight junction protein zonula occludens-1 during mammary engorgement. Proceedings of the New Zealand Society of Animal Production 64,43-47. Singh, K., Dobson, J., Phyn, C. V. C., Davis, S. R., Farr, V. C., Molenaar, A. J., and Stelwagen, K. (2005): Milk accumulation decreases expression of genes involved in cell-extracellular matrix communication and is associated with induction of apoptosis in the bovine mammary gland. Livestock Production Science 98,67-78. Appendix 7 McMahon, C.D., Farr, V.C., Singh, K., Wheeler, T.T. and Davis, S.R. (2004). Decreased expression of ß1-integrin and focal adhesion kinase in epithelial cells may initiate involution of mammary glands. Journal of Cellular Physiology 200, 318-325 / Extended periods of milk accumulation result in loss of secretory activity, increased apoptosis and eventually, involution of mammary glands. This process is associated with increased permeability of the tight junction (TJ) complexes between adjacent mammary epithelial cells (MECs). The change in cell shape during mammary engorgement from a cuboidal to a flattened morphology may initiate changes in protein and gene expression (mechanotransduction) that trigger these processes. Therefore, this study examined the regulation of the major TJ protein components during mammary engorgement, and in particular the role of physical distension of the mammary epithelium in the regulatory process. Expression of the integral transmembrane TJ proteins, occludin and claudin-1, and the cytoplasmic TJ protein, ZO-1, were down-regulated in both bovine and rat mammary glands during the early stages of mammary apoptosis and involution following the abrupt cessation of milk removal. In the rat, these responses were locally regulated as they occurred only in teat-sealed glands in a hemi-suckled model. Furthermore, the down-regulation of TJ proteins is consistent with a loss of TJ integrity during mammary engorgement. Induced physical distension of rat mammary glands in vivo transiently up-regulated the expression levels of occludin protein and mRNA, and ZO-1 mRNA, followed by an accelerated decrease in expression compared with the effects of milk accumulation alone. This was associated with the initiation of apoptosis, the up-regulation of the pro-apoptotic factor pSTAT3, and the down-regulation of the cell-ECM survival factor βl-integrin. An in vitro model was also developed to stretch MECs, mimicking the flattening in cell shape during mammary engorgement in vivo. While stretching MECs in vitro did not conclusively alter TJ protein expression, the overall results of this project support further investigation into the role of the TJ complex in mechanotransduction pathways. In addition, the results point to crosstalk between cell-ECM survival signalling and STAT3 death signalling as a candidate for regulation by physical distension of the mammary epithelium. In conclusion, this study supports the hypothesis that physical distension during engorgement of the mammary glands with milk is a primary trigger initiating apoptosis of MECs through changes in the regulation of gene pathways controlling cell survival and death, and the disruption of TJ function.
8

Modulation du phénotype dans les cellules HMEC / Phenotype modulation in HMEc

Abi Khalil, Amanda 28 June 2017 (has links)
Le cancer du sein est une pathologie hétérogène au plan clinique et au moins 5 sous-types moléculaires ont pu être définis sur la base de différences d’expression ARNm. Ces sous-types présentent des différences de profils d’anomalies génomiques et de méthylation des cytosines. Ces différences génétiques et épigénétiques s’expliqueraient par des types cellulaires d’origines distincts au sein de l’épithélium mammaire, toutefois, ceci n’a pas été confirmé clairement à ce jour. Alternativement, il a été proposé que l’activation de voies oncogéniques différentes pouvait avoir un impact significatif sur les modifications génétiques ou épigénétiques. Dans ce travail nous avons voulu vérifier cette hypothèse en l’appliquant à un modèle de cellules épithéliales mammaires normales primaires humaines, que nous avons isolé des à partir de glandes mammaires. Ces cellules ont été transformées en deux étapes par transduction avec (i) un shARN ciblant TP53, (ii) un oncogène. Nous avons sélectionné 3 oncogènes qui activent des voies de signalisations distinctes CCNE1, HRAS-v12 et WNT1. Nous avons établi un modèle de transformation tumorale en trois étapes, cellules normales, immortalisées et transformées, permettant de suivre les modifications moléculaires associées à chaque étape et de vérifier si l’activation de voies oncogéniques distinctes produisait des profils d’anomalies différents. Les différents modèles ont été analysés par CGH-array, RRBS, transcriptome et miRNA à des temps de culture définis.Nos résultats montrent que l’activation de la voie RAS aboutit à des profils d’anomalies génétiques et de méthylation des CpG radicalement différents de ceux obtenus après surexpression des gènes CCNE1 et WNT1. Ces différences apparaissent très rapidement après transduction des oncogènes alors que les profils des cellules CCNE1 et WNT1 divergent plus tardivement. Enfin, l’inactivation de p53 n’induit pas par elle-même une instabilité élevée, mais produit un contexte de plasticité favorable aux modifications génétiques et épigénétique.Par ailleurs, nous avons noté des différences phénotypiques entre les HMEC RAS (mésenchymateuses) et les HMEC CCNE1 et les HMEC WNT1 (épithéliales). Dans ce travail, je montre que les HMEC shp53 immortalisées présentent une plasticité phénotypique, une partie des cellules entrant en EMT spontanément, l’autre restant épithéliales. J’ai montré que la transduction RAS sélectionnait les cellules ayant effectué une EMT, alors que la transduction de CCNE1 ou WNT1 sélectionnait les cellules épithéliales. J’ai cherché à identifier les déterminants de ces changements phénotypiques et mes résultats suggèrent qu’ils résultent d’une balance entre une signalisation TGFB1/BMP1, qui favorise l’EMT, et BMP4/WNT7 qui favorise la MET. / Breast cancer is a heterogeneous pathology. Based on the differences of mRNA expression, at least five molecular subtypes have been defined. These subtypes show differences in profiles of genomic abnormalities and CpG methylation. These molecular subtypes are thought to originate from different cell lineages in the mammary gland. However, this has not yet been clearly demonstrated. Alternatively, it has been proposed that the activation of different oncogenic pathways could have a significant impact on genetic or epigenetic changes.We wanted to verify this hypothesis by applying it to a normal primary human mammary epithelial cells (HMEC) model, which we isolated from normal mammary explants. These cells were transformed in two step process by sequential transduction of (i) a shRNA targeting TP53, (ii) an oncogene. We selected 3 oncogenes that activate distinct signaling pathways CCNE1, HRAS-v12 and WNT1. Our tumor transformation model was established in three-step, normal, immortalized and transformed cells, allowing us to monitor the molecular changes associated with each step and to verify whether the activation of distinct oncogenic pathways produced different profiles of genetic and epigenetic modifications. The different models were analyzed at defined culture times by CGH-array, RRBS, transcriptome and miRNA. Our results show that genetic abnormalities and CpG methylation profiles are different between cells where the RAS pathway was activated and cells overexpressing WNT1 or CCNE1. These differences appear rapidly after oncogene transduction, whereas the profiles of the CCNE1 and WNT1 cells diverged later. Finally, inactivation of p53 by itself does not induce high instability, but produces a context of plasticity favorable to genetic and epigenetic changes.In addition, we noted phenotypic differences between HMEC RAS (mesenchymal) and HMEC CCNE1 and HMEC WNT1 (epithelial). In this work, I show that the immortalized HMEC shp53 exhibit a phenotypic plasticity, where some cells enter a spontaneous EMT and the others remain epithelial. I showed that RAS transduction selected cells that are undergoing an EMT, whereas transduction with CCNE1 or WNT1 selected the epithelial cells. I have sought to identify the determinants of these phenotypic changes and my results suggest that a balance exists between TGFβ1 / BMP1 signaling, which promotes EMT, and BMP4 / WNT7a which promotes TEM.
9

Etude de l'impact de la leptine sur le statut oxydatif et inflammatoire du tissu mammaire : approche expérimentale in vitro et in vivo - Mise en oeuvre de la technique de détection par fluorescence native. / Impact of leptin on oxidative and inflammatory status of the mammary tissue. An in-vitro and in- vivo experimental approach using the native fluorescence detection technique.

Mahbouli, Sinda 10 September 2015 (has links)
La leptine est une hormone peptidique ayant une action sur de nombreux tissus. Une dérégulation de la sécrétion de cette hormone est observée au cours de l’obésité. L’obésité est fréquemment associée à des troubles de santé dont les principaux sont le diabète de type II, l’hypertension artérielle et les maladies cardiovasculaires. Elle est également un facteur de risque du cancer du sein, particulièrement en post-ménopause favorisant la récidive et augmentant la mortalité. Ces perturbations, associées à un état de stress oxydant défini par un excès de production des espèces réactives de l’oxygène (ERO) par rapport aux systèmes de défense antioxydants, pourraient avoir un impact majeur dans le risque de carcinogenèse chez le sujet obèse. Il est clairement établi aujourd’hui que le statut oxydatif des cellules est directement corrélé aux capacités de prolifération mais aussi de survie des cellules dans leur environnement. A ce jour, très peu de données existent concernant le rôle de la leptine dans la modulation du statut oxydatif des cellules épithéliales mammaires saines et tumorales. L’objectif de cette thèse était d’étudier d'abord les mécanismes d’action et les effets de la leptine sur le statut oxydatif et inflammatoire des cellules épithéliales mammaires saines et néoplasiques ; puis dans un deuxième temps, une étude expérimentale a été conduite pour caractériser in vivo l’impact de l’obésité associée ou non à l’activité physique sur la croissance tumorale et le statut oxydatif et inflammatoire des tumeurs. Le projet avait également pour but de mettre en œuvre une nouvelle technique d’analyse basée sur la détection de fluorescence native induite par excitation laser à 224 nm afin d’évaluer la production de composés bio-actifs de la famille des éicosanoïdes, dont les isoprostanes, impliqués dans le processus inflammatoire. Nous avons exploré in vitro l’impact de la leptine sur le statut oxydatif des cellules épithéliales mammaires. Cette étude nous a permis d’établir que la réponse au signal leptinique varie en fonction du statut néoplasique de la lignée considérée, en fonction du temps de contact et non de la dose testée. Ensuite, nous avons étudié l’impact de l’obésité associée ou non à l’activité physique sur la croissance tumorale et sur le statut oxydatif et inflammatoire des tumeurs à l’aide d’un modèle de souris âgées C57BL/6 nourries avec un régime hyper-lipidique (HL) pendant 14 semaines, et hébergées soit dans un environnement enrichi (EE) pour favoriser l’activité physique et les interactions sociales, soit dans un environnement standard pendant 8 semaines, après quoi des cellules syngéniques de tumeur mammaire EO771 ont été implantées dans les quatrièmes coussinets adipeux mammaires. In vitro, la leptine a stimulé la production de ROS de façon indépendante de la dose et cette augmentation était dépendante de la production d'O2 cytosolique. Les résultats montrent une augmentation significative du poids dans les groupes recevant le régime HL à prise alimentaire journalière identique. La composition corporelle à 8 semaines montre une prise de masse grasse significative sous régime HL, majorée par l’ovariectomie et partiellement limitée par l’activité physique. Après implantation des tumeurs, le régime HL favorise la croissance tumorale et la perte de l’activité locomotrice. Par contre, l’EE prévient la perte d’activité physique des animaux. L’ensemble de ces travaux montre que la leptine contribue à l’apparition d’un stress oxydant en lien avec le statut tumoral des cellules épithéliales mammaires. Ceci peut expliquer en partie l’augmentation du risque de cancer mammaire associée à l’obésité en post-ménopause. Ces résultats permettront d'objectiver le bénéfice d'une intervention nutritionnelle ciblée afin de moduler la réponse des cellules aux stimulations des adipokines. A terme, cette étude doit contribuer à mieux comprendre l’intégration des signaux issus de l’environnement cellulaire. / Obesity is now considered, as a risk factor for developing breast cancer in postmenopausal women and for mortality in response to this pathology. Obesity, which is frequently associated with hyperleptinemia, induces cellular signalling pathways, some of which involving reactive oxygen species (ROS) as intracellular messengers. High levels of ROS contribute to oxidative stress, cellular damages and pathogenesis. Therefore, ROS production associated to obesity could be a major risk factor for mammary carcinogenesis. Furthermore, increased oxidative stress and inflammation characterised by infiltration of immune cells into adipocytes are described. This is associated with a lipid peroxidation and the production of bio-active compounds including isoprostanes.The aim of this study was to determine the impact of leptin in modulating the oxidative and inflammatory status of epithelial mammary cells and in tumor mammary tissue. Moreover, the purpose of this work was to develop a new analysis technique based on native fluorescence detection induced by laser excitation at 224 nm to evaluate the production of bio-active compounds from the family of eicosanoids, involved in the inflammatory process, including isoprostanes.Initially we identified in vitro the leptin effects on ROS production in 3 human epithelial mammary cell models which present different neoplastic status (healthy primary (HMEC) cells, MCF-7 and MDA-MB-231) in presence of two leptin concentrations (10 ng/ml close to physiological values, 100 ng/ml as obesity level). To better understand the potential involvement of adipocyte tumor microenvironment in mammary carcinogenesis, we secondly explored in vivo the impact of high fat diet (HFD) and of enriched environment (EE) on mammary tumor development. Female C57BL/6 mice were fed with a HFD versus a normo-caloric diet (NC) for 14 weeks. After 8 weeks mammary tumor syngeneic cells EO771 were implanted into the fourth mammary fat pads. Before injection, mice were housed in EE or in standard environment (ES) for 8 weeks. In vitro, leptin stimulated ROS production in dose-independent manner and this increase was dependent of cytosolic O2•- production. This ROS production contributed to a different antioxidative response depending of the neoplastic cell status. Leptin induced the antioxidative enzymes expression and activities such as heme-oxygenase or glutathione peroxidase only in HMEC cells. In neoplastic cells, these enzyme activities did not change whatever the leptin concentration used.Thus, high fat diet promoted mammary tumor development associated with a decrease in body fat and an increase in volume and weight of tumors that was not limited by physical activity. This diet induced a decrease of adiponectin and an increase of leptin plasma level compared to NC diet however, leptinemia was not influenced by EE.The native fluorescence isoprostanes determination method, turned out not to be quite sensitive. Therefore, the native fluorescence of these compounds is too low to allow their detection in biological media used. In contrast, the native fluorescence appears to be a potential cellular exploration tool.Through this work, we have shown that leptin contributes to the onset of oxidative stress linked to the status of mammary epithelial tumor cells. This may partly explained the increase of risk of breast cancer recurrence observed in situations of obesity. The results obtained in vivo eventually will support the benefit of a nutrition intervention to modulate cell response to adipokines stimulation. Ultimately, this study contributes to better understand the integration of signals from the cell environment.
10

Bovine Mastitis Resistance: Novel Quantitative Trait Loci and the Role of Bovine Mammary Epithelial Cells

Kurz, Jacqueline P. 01 May 2018 (has links)
Bovine mastitis, or inflammation of the mammary gland, has substantial economic and animal welfare implications. A genetic basis for mastitis resistance traits is recognized and can be used to guide selective breeding programs. The discovery of regions of the genome associated with mastitis resistance, and knowledge of the underlying molecular mechanisms responsible, can facilitate development of efficient mastitis control and therapeutic strategies. The objectives of this dissertation research were to identify sites of genetic variation associated with mastitis resistance, and to define the contributions of the milk-secreting epithelial cells to mammary gland immune responses and mastitis resistance. Twenty seven regions of the bovine genome potentially involved in mastitis resistance were identified in Holstein dairy cattle. Additionally, this research demonstrates a role of bovine mammary epithelial cells in mastitis resistance, and provides guidance for the use of an in vitro model for mastitis studies. Primary bovine mammary epithelial cells from mastitis-resistant cows have differential expression of 42 inflammatory genes compared with cells from mastitis-susceptible cows, highlighting the importance of epithelial cells in mastitis resistance. Bovine mammary epithelial cells display both similarities and differences in pro-inflammatory gene expression compared to fibroblasts, and their expression of inflammatory genes is influenced by administration of the enzyme phospholipase A2. The growth potential of milk-derived bovine mammary epithelial cells in vitro can be extended, facilitating their use in mastitis studies, by transfection with a viral protein. Collectively, this research contributes to current knowledge on bovine mastitis resistance and in vitro models.

Page generated in 0.0701 seconds