• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 2
  • 1
  • Tagged with
  • 4
  • 4
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 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

Defining the functional role of laminin isoforms in the regulation of the adult hepatic progenitor cell

Williams, Michael John January 2015 (has links)
During chronic and severe acute liver injury, regeneration is thought to occur through hepatic progenitor cells (HPCs). Understanding the regulation of HPCs may offer therapeutic opportunities to enhance liver regeneration. HPCs are associated with an increase in laminins in the extracellular matrix. Laminins are heterotrimeric proteins, composed of an alpha, beta and gamma chain. There are 5 alpha chains with different distributions and functions, but the relative contributions of these in HPC-mediated liver regeneration are not known. My aims were to describe the laminin alpha chains associated with the HPC response and to define the functional effects of specific laminin chains on HPCs. I examined the laminin alpha chains in two mouse models of HPC activation: a transgenic model using conditional deletion of Mdm2 in hepatocytes, and a dietary model using 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). The laminin alpha 5 (Lama5) chain is significantly upregulated in both models and forms a basement membrane which surrounds the progenitor cells. I have also demonstrated Lama5 expression in the ductular reaction seen in human liver disease. Using primary mouse cell cultures, I have shown that Lama5 is produced predominantly by the HPCs themselves, rather than by stellate cells. The HPCs express the cell surface receptor alpha-6 beta-1 integrin, a binding partner of Lama5. I then studied the functional effects of matrix on cell behaviour in vitro using recombinant laminins and a line of spontaneously immortalised mouse HPCs. Compared to other laminin chains, Lama5 selectively promotes HPC adhesion and spreading. These effects are partially blocked by antibodies against beta-1 integrin. Lama5 also significantly enhances HPC migration, resulting in an increase in cell migration. Furthermore, only Lama5 enhances HPC survival in serum-free medium, with an increase in cell viability. Culturing HPCs on HPCs maintained in culture on plastic synthesise Lama5 chain. Knock-down of endogenous Lama5 production using siRNA results in reduced proliferation and increased hepatocytic differentiation, with increased albumin production. I then studied the effects in vivo using transgenic Cre-lox mouse strains that allow conditional knock-out of either laminin alpha 5 or beta-1 integrin in HPCs. The effects of gene deletion were examined in healthy mice and two dietary models of HPC activation: the DDC diet and a choline-deficient, ethionine-supplemented (CDE) diet. Although these experiments were limited by a low number of experimental animals and low recombination rates, there was a suggestion of impaired HPC expansion associated with loss of laminin alpha 5. There was also a significant increase in hepatocellular injury and fibrosis in response to the DDC diet seen with loss of laminin alpha 5 expression. Laminin alpha 5-containing matrix is deposited around HPCs during liver regeneration and supports progenitor cell attachment, migration and maintenance of an undifferentiated phenotype. This work identifies a novel target for enhancing liver regeneration.
2

Role of interferon α and γ in the hepatic progenitor (oval) cell response

Lim, Rebecca January 2007 (has links)
[Truncated abstract] Hepatic progenitor cells (HPC) are becoming increasingly recognized as facultative stem cells capable of regenerating the liver during chronic liver injury and also as targets of malignant transformation. Similar markers are expressed by hepatocellular carcinoma (HCC) and HPC, and a precursor-product relationship is well established. This thesis focuses on the ways in which the HPC population can be controlled under circumstances of chronic liver injury, and in this manner, reduce the risk of progression to HCC reduced. The major aim of Chapters 3 to 5 was to elucidate the effect of interferon α (IFNα) therapy on HPC. Chronic hepatitis C affects approximately 250 million individuals world wide. Approximately 80% of infections progress to chronicity, which places the individuals at greater risk of developing HCC. The gold standard of treatment of chronic hepatitis C is a combination of pegylated IFNα and ribavirin. ...The results were surprising. While IFNγ exerted a pro-apoptotic and antiproliferative effect on HPC in vitro, administration of IFNγ to CDE-fed mice for 14 days increased fibrosis, enhanced inflammatory infiltration and exacerbated the HPC response, with concurrent hepatocyte cell death. In addition, increased morbidity and mortality were observed in the IFNγ-treated mice compared to control. IFNγ treatment was found to prime the liver for the HPC response by recruiting inflammatory cells and altering the hepatic cytokine profile, both of which may facilitate an increased HPC response. Numbers of activated HSC were also increased in the IFNγ-treated, CDE-fed mice, correlating with the increased fibrosis seen in these animals. This data contradicts the current experimental use of IFNγ for treatment of fibrosis. Based on our results, we suggest that IFNγ promotes HPC proliferation in the CDE model, by encouraging inflammatory infiltration and hepatocyte damage and this initiates pro-fibrotic events. Concurrent proliferation of HPC and activated HSC further supports the view that there is a close relationship between the two cell types, and thus, a link between the HPC response and fibrosis. In conclusion, findings documented in this thesis suggest that administration of IFNα and IFNγ can contribute to shaping the HPC response. IFNα therapy may reduce HCC risk in chronic hepatitis C patients by bringing the HPC population under control. In contrast, IFNγ treatment can exacerbate the HPC response, liver fibrosis and parenchymal damage, illustrating the need to approach this method of fibrosis treatment with caution.
3

Effect of selective COX-2 inhibitors on hepatic progenitor cells and the pathologies of experimental hepatocarcinogenesis

Davies, Richard January 2007 (has links)
[Truncated abstract] Hepatocellular carcinoma (HCC) is the major malignancy complicating chronic liver disease. New therapies for the prevention of HCC are required due to the limited success and high tumour recurrence rates of existing treatments. Emerging evidence suggests that HCC arise from the transformation of adult liver progenitor cells (LPCs), which have the capacity to differentiate into hepatocytes and biliary cells during liver regeneration. LPC activation precedes neoplasia in experimental hepatocarcinogenesis. LPCs share antigenic epitopes with HCCs, including α-fetoprotein (AFP) and M2- pyruvate kinase (M2PK). In animal models of hepatocarcinogenesis, attenuation of the LPC response reduces the incidence of HCC following prolonged liver injury via a tumour necrosis factor (TNF) dependent mechanism. As TNF is a pro-inflammatory cytokine, these data suggest that anti-inflammatory agents may be effective in inhibiting LPC activation and hepatocarcinogenesis. Cyclo-oxygenase-2 (COX-2) is an inducible enzyme that mediates the production of many prostaglandins during inflammation and carcinogenesis. Recent investigations show that the administration of selective COX-2 inhibitors (SC2Is) may reduce the incidence of a variety of tumours including breast, colon and skin. The broad aim of this thesis was to conduct a series of detailed studies on the effects of a SC2I on LPC activation and the hepatic pathologies associated with hepatocarcinogenesis in order to test the hypothesis that S2CIs may be a beneficial therapy that can reduce liver injury and pre-neoplastic changes in the choline-deficient, ethionine supplemented (CDE) murine model of hepatocarcinogenesis. Administration of a SC2I (SC-236) significantly inhibited a variety of hepatic cell populations that expand during the first month of the CDE mouse model of hepatocarcinogenesis (a choline deficient, ethionine supplemented diet). Numbers of M2PK-positive LPCs (which are more hepatocytic in morphology and are also COX-2 positive) and inflammatory cells were all significantly reduced by SC-236. In contrast, numbers of A6-positive LPCs (which are more biliary cell-like in morphology and do not express COX-2) were unchanged. ... In summary, these data suggest that COX-2 inhibitors such as SC-236 inhibit LPC activation and a variety of pre-neoplastic liver pathologies as a result of COX-2 dependent and independent mechanisms that may be mediated through inhibition of Akt phosphorylation and induction of apoptosis. Moreover, SC2Is may be useful as preventative treatment strategies for HCC in patients with chronic liver disease.
4

Papel de IRS2 en la regulación de la comunicación a través de FGFs en el nicho de células progenitoras hepáticas

Arámbul Anthony, María José 28 October 2022 (has links)
[ES] La resistencia a la insulina se define como un aumento en la cantidad de insulina necesaria para conseguir la homeostasis de glucosa. Una de las complicaciones más comunes de la resistencia a la insulina es el defecto en la reparación de herida. El sustrato 2 del receptor de la insulina (IRS2) es un mediador clave para la señalización de insulina en hígado que actúa de puente entre los receptores de la insulina y del factor de crecimiento insulínico (IGF-1) y sus cascadas de señalización. Tanto la resistencia a la insulina como cambios en los niveles de expresión de IRS2 han sido asociados con el desarrollo y la progresión de enfermedades hepáticas graves. El daño hepático crónico generado por algunos factores derivados de la resistencia a la insulina ha sido establecido como determinante en la patofisiología de las enfermedades hepáticas. Sin embargo, sigue siendo una incógnita cómo el daño hepático generado por la resistencia a la insulina escapa de la extraordinaria capacidad de regeneración del hígado. Durante el daño hepático crónico, la reparación epitelial está mediada por las células progenitoras hepáticas (CPH) que se expanden y diferencian hasta hepatocitos o células biliares rodeadas de un nicho estromal formado por un conjunto de células estrelladas hepáticas (CEH), células inflamatorias, componentes de la matriz extracelular (ECM) y factores de crecimiento. El factor de crecimiento de fibroblastos 7 (FGF7), expresado por las CEH, resulta crítico para la respuesta de las CPH y para la regeneración hepática. Durante el daño hepático crónico las CEH se activan transdiferenciandose desde células quiescentes a células fibrogénicas (CEHa) denominadas miofibroblastos que depositan ECM para reemplazar el tejido dañado. Para alcanzar una correcta regeneración hepática se requiere la resolución de la activación ("reversión fibrogénica") de las CEH. Empleando el modelo de ratón Irs2-/- durante el daño hepático crónico con la dieta DDC 0.1% y los modelos in vitro humanos de CPH (HepaRG) y de CEH (CEH primarias y la línea celular LX-2), los resultados de este trabajo demuestran que la señalización de insulina-IRS2 es necesaria para la epitelización dirigida por la comunicación paracrina a través de FGF7 en el nicho de CPH. Por un lado, IRS2 es necesario en la población de CEH para permitir su supervivencia durante la reversión fibrogénica, un proceso que según nuestros resultados induce un aumento en la expresión de FGF7. Nuestros datos descubren un potencial mecanismo de regulación mediante el que IRS2 induce la expresión de FGF7 en CEH a través de la remodelación en la ECM mediada por NRF2 y el integrante de la ECM SERPINE1 durante las etapas tempranas de la reversión fibrogénica. El eje NRF2-SERPINE1 ha sido descrito anteriormente en fibroblastos de piel como esencial para la reepitelización de herida. NRF2 es el principal factor de transcripción de respuesta frente al estrés oxidativo (ruta canónica). En hepatocitos, la activación de NRF2 también puede inducirse a través de una ruta no canónica mediada por la proteína cargo de la autofagia P62. A pesar de que nuestros datos descubren a P62 como capaz de inducir la actividad de NRF2 en CPH, también demuestran que IRS2 activa a NRF2 en CPH y en CEH de manera independiente a la ruta no canónica mediada por P62. Por otro lado, demostramos que la señalización de insulina-IRS2, por promover la producción de FGF7, permite un novedoso bucle de inducción positiva mediante el que la respuesta a FGF7 en CPH promueve la expresión de su receptor, FGFR2b, favoreciendo su propia sensibilidad y sosteniendo la reparación epitelial. Futuras estrategias para potenciar en hígado la actividad de NRF2 y la señalización de FGF7 podrían servir para mejorar el pronóstico de los pacientes con resistencia a la insulina, diabetes o enfermedad metabólica por promover la reparación epitelial en hígado y reducir su riesgo de desarrollar patologías hepáticas graves con elevada tasa de mortalidad. / [CAT] La resistència a la insulina es defineix com un augment en la quantitat d'insulina necessària per a aconseguir l'homeòstasi de glucosa. Una de les complicacions més freqüents de la resistència a la insulina és el defecte en la reparació de ferida. El substrat 2 del receptor de la insulina (IRS2) és un mediador clau per a la senyalització d'insulina en fetge que actua de pont entre els receptors de la insulina i del factor de creixement insulínic (IGF-1) i les seues cascades de senyalització. Tant la resistència a la insulina com els canvis en els nivells d'expressió d'IRS2 han sigut associats amb el desenvolupament i la progressió de malalties hepàtiques greus. El mal hepàtic crònic generat per alguns factors derivats de la resistència a la insulina s'ha establert com a determinant en la patofisiologia de les malalties hepàtiques. No obstant això, els motius pels quals el mal hepàtic generat per la resistència a la insulina escapa a l'extraordinària capacitat de regeneració del fetge són encara una incògnita. Durant el mal crònic, la reparació epitelial està mediada per les cèl·lules progenitores hepàtiques (CPH) que s'expandeixen i diferencien fins a hepatòcits o cèl·lules biliars envoltades d'un nínxol estromal format per un conjunt de cèl·lules estavellades hepàtiques (CEH), cèl·lules inflamatòries, components de la matriu extracelul·lar (ECM) i factors de creixement. El factor de creixement de fibroblasts 7 (FGF7), expressat en fetge per les CEH, resulta crític per a la resposta de les CPH i per a la regeneració hepàtica. Durant el mal hepàtic crònic les CEH s'activen transdiferenciant-se des de cèl·lules quiescents a cèl·lules fibrogèniques denominades miofibroblasts (CEH activades) que depositen ECM per a reemplaçar el teixit danyat. Per a aconseguir una correcta regeneració hepàtica es requereix la resolució de l'activació ("reversió fibrogènica") de les CEH. A partir de l'ús del model de ratolí Irs2-/- durant el mal hepàtic crònic amb la dieta DDC 0.1% i dels models in vitro humans de CPH (HepaRG) i de CEH (CEH primàries i la línia cel·lular LX-2), els resultats d'aquest treball demostren que la senyalització d'insulina-IRS2 és necessària per a l'epitelització dirigida per la comunicació paracrina mitjançant FGF7 en el nínxol de CPH. D'una banda, IRS2 és necessari en la població de CEH per a permetre la seua supervivència durant la reversió fibrogènica, que comporta un augment en l'expressió de FGF7. Les nostres dades descobreixen un potencial mecanisme de regulació mitjançant el qual IRS2 indueix l'expressió de FGF7 en CEH a través de la remodelació en la ECM mediada per NRF2 i per l'integrant de la ECM SERPINE1, que ocorre en les etapes primerenques de la reversió fibrogènica. L'eix NRF2-SERPINE1 ha sigut identificat anteriorment en fibroblasts de pell com a essencial per a la reparació de ferida. NRF2 és el principal factor de transcripció de resposta davant de l'estrés oxidatiu (ruta canònica). En hepatòcits, l'activació de NRF2 també pot induir-se a través d'una ruta no canònica mediada per la proteïna de càrrega de l'autofàgia P62. A pesar que les nostres dades indiquen que P62 és capaç d'induir l'activitat de NRF2 en CPH, també demostren que IRS2 activa NRF2 en CPH i CEH de manera independent a la ruta no canònica mediada per P62. D'altra banda, demostrem que la senyalització d'insulina-IRS2, per promoure la producció de FGF7, permet un nou bucle d'inducció positiva mitjançant el qual la resposta a FGF7 en CPH promou l'expressió del seu receptor, FGFR2b, afavorint la seua pròpia sensibilitat i sostenint la reparació epitelial. Futures estratègies per a potenciar en fetge l'activitat de NRF2 i la senyalització de FGF7 podrien servir per a millorar el pronòstic dels pacients amb resistència a la insulina, diabetis o malaltia metabòlica, per promoure la reparació epitelial en fetge i reduir, per tant, el seu risc de desenvolupar patologies hepàtiques greus amb elevada taxa de mortalitat. / [EN] Insulin resistance is defined as an increase in the amount of insulin that is necessary to achieve glucose homeostasis. One of the most prevalent complications of insulin resistance is the wound healing defect. Insulin receptor factor 2 (IRS2) is a key mediator of the insulin signaling in liver, which acts as a bridge between insulin and insulin growth factor 1 (IGF-1) receptors and its downstream molecular pathways. Both, insulin resistance and changes in the expression levels of IRS2, have been associated with the development and progression of severe liver diseases. Chronic liver injury produced by insulin resistance has been stablished as crucial in the pathophysiology of liver disease. However, it remains unknown how the chronic liver injury produced by insulin resistance escapes from the extraordinary ability of the liver to regenerate. During chronic liver injury, epithelial repair is mediated by liver progenitor cells (LPC), that expand and differentiate into hepatocytes and cholangiocytes surrounded by a stromal niche that consist of hepatic stellate cells (HSC), inflammatory cells, extracellular matrix (ECM) components and growth factors. Fibroblast growth factor 7 (FGF7), expressed by HSC, is critic for LPC response and liver regeneration. During chronic liver injury, HSC transdifferentiate from quiescent to active fibrogenic HSC (aHSC) called myofibroblast. aHSC deposit ECM to replace damaged tissue. A successful regeneration requires the resolution of the HSC activation, i.e., the "fibrogenic reversion" of HSC. Using the Irs2-/- mice model during chronic liver damage induced by 0.1% DDC diet and the human in vitro models of LPC (HepaRG) and HSC (primary HSC and the cell line LX-2), our results reveal a new role of insulin-IRS2 in the modulation of the paracrine FGF7 crosstalk in the LPC niche that drives LPC epithelization. On the one hand, IRS2 is necessary in HSC to allow survival during fibrogenic reversion, a process that according to our data induces an increase in FGF7 expression. Our results reveal a potential mechanism by which IRS2 promotes FGF7 expression in HSC through an ECM remodeling process that is mediated by NRF2 and the ECM constituent SERPINE1 during the early stages of fibrogenic reversion. NRF2-SERPINE1-mediated ECM remodeling has been previously identified in skin fibroblast as essential to promote re-epithelialization of wounds. NRF2 is a transcription factor that is activated in response to oxidative stress (canonical pathway). NRF2 activation in hepatocytes can be also induced by a non-canonical pathway mediated by the autophagy cargo protein P62. Although our data discovers a new ability of P62 to induce NRF2 activity in LPC, we also demonstrate that IRS2 activates NRF2 in LPC and HSC in a P62-independent manner. On the other hand, we demonstrate that insulin-IRS2 signaling, by promoting FGF7 production, enables a novel positive induction loop whereby FGF7 response in LPC promotes the expression of its receptor, FGFR2b, favoring its own sensitivity and sustaining epithelial repair. Future strategies to enhance NRF2 activity or FGF7 signaling in liver might be useful to improve the prognosis of insulin resistance, diabetic, and metabolic disease patients because of its uncovered ability to promote epithelial repair, thus, preventing the development of severe liver pathologies with high mortality risk. / Arámbul Anthony, MJ. (2022). Papel de IRS2 en la regulación de la comunicación a través de FGFs en el nicho de células progenitoras hepáticas [Tesis doctoral]. Universitat Politècnica de València. https://doi.org/10.4995/Thesis/10251/188913

Page generated in 0.1222 seconds