• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 56
  • 14
  • 7
  • 4
  • 4
  • 3
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • Tagged with
  • 109
  • 109
  • 29
  • 23
  • 19
  • 18
  • 15
  • 14
  • 14
  • 13
  • 12
  • 11
  • 10
  • 10
  • 10
  • 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.
91

Role of Bone Morphogenetic Proteins for Catecholaminergic Neurons in Vivo : Use of the Tyrosine Hydroxylase Locus for Cell-Specific inactivation of Signal Transduction

Usoskin, Dmitry January 2004 (has links)
Members of the Transforming Growth factor-β (TGF-β) superfamily and its subclass Bone Morphogenetic Proteins (BMP) play important roles for nervous system development. In order to study the BMP role for catecholaminergic neurons in vivo, we generated three knock-in mice, expressing the transgenes specifically in the targeting cells. Two genetic modifications result in expression of dominant negative (dn) BMP receptors (BMPRII and ALK2). The tissue-specific expression was achieved by the transgene insertion into 3’- untranslated region of the endogenous gene for tyrosine hydroxylase (TH), the first enzyme in catecholamine biosynthesis. An Internal Ribosome Entry site (IRES) preceded inserted cDNAs, allowing for functional bicistronic mRNA production. While almost no defects in Th-IRES-dnALK2, the Th-IRES-dnBMPRII mouse demonstrated declined levels of catecholamines, including dopamine in the striatum. Losses of midbrain dopaminergic neurons (MDN) might cause the effect. Additionally, intermediate lines of these mice, preserving a neo-cassette, oriented opposite to the locus transcription, demonstrate dramatic decrease of catecholamine level, hence, represent models for rare catecholamine-deficiency diseases, including L-DOPA-responsive dystonia. The third mouse, expressing in the same way Cre-recombinase (Th-IRES-Cre), represents a tool for catecholaminergic cell-limited deletion of any gene, which has to be flanked by loxP sites. Besides TH-positive areas, unexpected sites of Cre-recombination were identified, indicating regions of transient TH expression. Surprising recombination in oocytes opens a possibility to use our mouse as a general Cre-deletor. Using TH-IRES-Cre mouse we generated tissue-specific knockout mice for two BMP signal transducers: Smad1 and Smad4 (also crucial for TGF-β). While no phenotype in Smad1 knockout, TH-IRES-Cre/Smad4 mouse revealed several defects including decreased level of striatal dopamine. These results demonstrate a positive role of BMPs for MDN fate in vivo. Generated mice represent a tool-box for comprehensive study of the BMP function in catecholaminergic neurons. This study is of potential interest for understanding some aspects of Parkinson’s disease.
92

Novel Regulators of the TGF-β Signaling Pathway

Kowanetz, Marcin January 2005 (has links)
The transforming growth factor-β (TGF-β) superfamily consists of related multifunctional cytokines, which include TGF-βs, activins, and bone morphogenetic proteins (BMPs) and coordinate several biological responses in diverse cell types. The biological activity of TGF-β members is executed by transmembrane serine/threonine kinase receptors and intracellular Smad proteins. The effects of TGF-β on the epithelium are of high interest. Carcinomas (tumors of epithelial origin) are the most common type of human cancer and frequently exhibit aberrant responses to TGF-β. Therefore, TGF-β can be defined as tumor suppressor as it inhibits growth of normal epithelial cells. However, TGF-β also induces an epithelial-mesenchymal transition (EMT), a key component of metastasis, and thus promotes cancer spread. The scope of this thesis is the mechanism of TGF-β signaling in epithelial cells. We established that only TGF-β, but not BMP pathways can elicit EMT. Moreover, we found that Smad signaling is critical for regulation of EMT. In a transcriptomic analysis, we identified a large group of novel genes, whose regulation is pivotal for TGF-β-induced EMT and metastasis. We focused on two of such genes, Id2 and Id3. Interestingly, we found that TGF-β-induced repression of Ids is necessary for inducing EMT and potent cell cycle arrest. BMP increases expression of Ids and therefore it cannot induce the same biological responses as TGF-β. Hence, knock-down of endogenous Id2 and Id3 proteins sensitized epithelial cell to BMP-7. We proposed a model, in which Id2 and Id3 are important components controlling concerted regulation of cell proliferation and EMT downstream of TGF-β pathways. Furthermore, we identified a serine/threonine kinase, SNF1LK, whose mRNA is rapidly induced by TGF-β in epithelial cells. We found that SNF1LK is a negative regulator of the TGF-β pathway and it promotes TGF-β receptor turnover. Subsequently, we demonstrated that SNF1LK together with Smad7 and Smurf2 targets TGF-β receptor for ubiquitin-dependent degradation. Furthermore, SNF1LK interacts with proteasomes, suggesting that SNF1LK serves as bridge between ubiquitinated receptors and proteasomes, helping proteasomes to recognize the ubiquitinated cargo destined for degradation. We therefore established a novel negative feedback regulatory mechanism of TGF-β signaling.
93

Epigenetic Regulation of Replication Timing and Signal Transduction

Bergström, Rosita January 2008 (has links)
Upon fertilization the paternal and maternal genomes unite, giving rise to the embryo, with its unique genetic code. All cells in the human body are derived from the fertilized ovum: hence they all contain (with a few exceptions) the same genetic composition. However, by selective processes, genes are turned on and off in an adaptable, and cell type-specific, manner. The aim of this thesis is to investigate how signals coming from outside the cell and epigenetic factors residing in the cell nucleus, cooperate to control gene expression. The transforming growth factor-β (TGF-β) superfamily consists of around 30 cytokines, which are essential for accurate gene regulation during embryonic development and adult life. Among these are the ligands TGF-β1 and bone morphogenetic (BMP) -7, which interact with diverse plasma membrane receptors, but signal via partly the same Smad proteins. Smad4 is essential to achieve TGF-β-dependent responses. We observed that by regulating transcription factors such as Id2 and Id3 in a specific manner, TGF-β1 and BMP-7 achieve distinct physiological responses. Moreover, we demonstrate that CTCF, an insulator protein regulating higher order chromatin conformation, is able to direct transcription by recruiting RNA polymerase II to its target sites. This is the first mechanistic explanation of how an insulator protein can direct transcription, and reveals a link between epigenetic modifications and classical regulators of transcription. We also detected that DNA loci occupied by CTCF replicate late. The timing of replication is a crucial determinant of gene activity. Genes replicating early tend to be active, whereas genes replicating late often are silenced. Thus, CTCF can regulate transcription at several levels. Finally, we detected a substantial cross-talk between CTCF and TGF-β signaling. This is the first time that a direct interplay between a signal transduction pathway and the chromatin insulator CTCF is demonstrated.
94

Regulation of TGF-β Signaling by Post-Translational Modifications

Lönn, Peter January 2010 (has links)
Transforming growth factor-β (TGF-β) signaling is initiated when the ligand binds to type II and type I serine/threonine kinase receptors at the cell surface. Activated TGF-β type I receptors phosphorylate R-Smads which relocate, together with co-Smads, to the cell nucleus and regulate transcription. Enhancement or repression of Smad-specific gene targets leads to intracellular protein compositions which organize functional complexes and thus govern cellular processes such as proliferation, migration and differentiation. TGF-β/Smad signaling relays are regulated by various post-translational modifications. From receptors to gene promoters, intricate interplays between phosphorylation, acetylation, ubiquitination and numerous other modifications, control Smad signaling initiation and duration. However, many steps in the cascade, including receptor internalization, Smad nuclear shuttling and transcriptional termination, still remain elusive. The open gaps in our understanding of these mechanisms most likely involve additional post-translational regulations. Thus, the aim of the present investigation was to identify novel modulators of TGF-β/Smad signaling. In the first part of this thesis, we show the importance of ADP-ribosylation in Smad-mediated transcription. We identified poly(ADP-ribose) polymerase 1 (PARP-1) as a Smad interacting protein. Our work revealed that PARP-1 forms direct interactions with Smad3/4, and PARylates residues in their MH1 domains. This modification restricts Smads from binding to DNA and attenuates Smad-activated transcription. PARylation is reversed by the glycohydrolase PARG. We provide evidence that PARG can de-ADP-ribosylate Smads, which enhances Smad-promoted gene regulation. In the second part, we examine a Smad-dependent gene target of TGF-β signaling, salt inducible kinase 1 (SIK). After induction, SIK cooperates with Smad7 and Smurf2 to downregulate the TGF-β type I receptor. The mechanism relies on both the kinase and UBA domain of SIK as well as the E3-ligase activity of Smurf2. In summary, we have unveiled two enzyme-dependent TGF-β/Smad modulatory mechanisms; SIK promoted receptor turnover and PARP-1/PARG-regulated Smad signaling.
95

Role of stroma and Wound Healing in carcinoma response to ionizing radiation

Arshad, Adnan 03 July 2014 (has links) (PDF)
Wound healing and carcinogenesis are defined as complex, adaptive processes which are controlled by intricate communications between the host and the tissue microenvironment. A number of phenotypic similarities are shared by wounds and cancers in cellular signaling and gene expression. Radiotherapy is the second most effective modality of cancer treatment after surgery and can be used, either alone or in combination with chemotherapy. Recent findings suggest that radiotherapy apart from tumor cell death also rapidly and persistently modifies the tissue microenvironment. These modifications affect cell phenotype, tissue metabolism, bidirectional exchanges and signaling events between cells. The complex interactions between stromal cells and cancer cells are of immense interest and in The First Part of My Thesis, I tried to explore the crosstalk between stromal and carcinoma cells in response to radiotherapy by genetic modulation of the stroma and irradiation. We found that fibroblasts, irrespective of their RhoB status, do not modulate intrinsic radiosensitivity of TC-1 but produce diffusible factors able to modify tumor cell fate. Then we found that Wt and RhoB deficient fibroblasts stimulated TC-1 migration through distinct mechanisms respectively, TGF-β1 and MMP-mediated. We also found that co-irradiation of fibroblasts and TC-1 abrogated the pro-migratory phenotype by repression of TGF-β and MMP secretion. This result is highly relevant to the clinical situation and suggests that conversely to, the current view; irradiated stroma would not enhance carcinoma migration and could be manipulated to promote anti-tumor immune response. Secondly, our in vivo experiments, tends to confirm the in vitro data showing that irradiated tumor bed does not stimulate tumor growth and escape. Our results also challenges the view that irradiated stroma would promote migration of carcinoma cells as we show that independently from their genotype co-irradiation of fibroblasts and carcinoma cells repressed carcinoma cell migration and confirmations studies are currently performed in vivo. The Third Part of My Project, was dedicated to investigate the effect on CTC release after radiotherapy. Consistently with the results reported after surgery , the number of CTC increases in the blood stream after radiotherapy probably due to radiation-induced vascular injury induced or/and by EMT induction in tumor cells but these cells seemed to be entrapped into the cardiac cavity. The significance of these CTC to metastatic development is still under investigation but there is evidence for a metastasis-promoting effect of RT from animal studies.Thus the microenvironment can exert antagonist stimulatory or inhibitory effects on malignant cells.
96

Inibição da metástase via transição epitélio-mesenquimal por shRNA, metformina e Y27632 em neoplasia mamária / Inhibition of metastasis via epithelial-mesenchymal transition by shRNA, metformin and Y27632 in breast cancer

Silva, Camila Leonel da [UNESP] 23 April 2016 (has links)
Submitted by CAMILA LEONEL DA SILVA null (camilaleonels_1@hotmail.com) on 2016-05-02T19:33:26Z No. of bitstreams: 1 TESE - Camila Leonel da Silva.pdf: 12839463 bytes, checksum: a432431aa8b8009a0183a4c0896c3e34 (MD5) / Approved for entry into archive by Felipe Augusto Arakaki (arakaki@reitoria.unesp.br) on 2016-05-04T19:19:33Z (GMT) No. of bitstreams: 1 silva_cl_dr_sjrp.pdf: 12839463 bytes, checksum: a432431aa8b8009a0183a4c0896c3e34 (MD5) / Made available in DSpace on 2016-05-04T19:19:33Z (GMT). No. of bitstreams: 1 silva_cl_dr_sjrp.pdf: 12839463 bytes, checksum: a432431aa8b8009a0183a4c0896c3e34 (MD5) Previous issue date: 2016-04-23 / Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) / A transição epitélio-mesenquimal (EMT) é o processo pelo qual as células cancerosas a partir de tumores primários passam por uma conversão fenotípica para invadir e migrar, gerar metástases em tecidos ou órgãos distantes. Este processo pode ser induzido por fatores de crescimento, tais como Fator de Crescimento Transformante beta (TGF-β) e sua alta expressão tem sido implicada na angiogênese tumoral, na migração e invasão celular em muitos tipos de tumores. A expressão de ROCK-1 está associada com a malignidade dos tumores, enquanto a inibição desta molécula resulta em uma supressão significativa de metástases tumorais. A metformina, um fármaco utilizado no tratamento da diabetes, demonstrou inicialmente inibir a EMT e impedir o fenótipo mesenquimal pela repressão transcricional de pontos chave da regulação da EMT (ZEB1, TWIST1, SNAIL2, TGF-β) em células de câncer de mama. Os objetivos foram avaliar a expressão gênica e proteica de marcadores relacionados a metástase, em um estudo in vitro e in vivo, em linhagens de câncer de mama, após o tratamento com metformina, além do silenciamento gênico do TGF-β1 para inibição da transição epitélio-mesenquimal. Foi realizado a transfecção da linhagem celular metastática de tumor mamário canino CF41 de forma estável após a construção de um pequeno RNA de interferência para desenvolver derivados clonais que expressam níveis reduzidos de TGF-β1 (células TGF-β1sh). Este foi subsequentemente combinado com o tratamento com metformina, para analisar os efeitos sobre a migração de células, assim como a expressão dos marcadores de EMT E-caderina e N-caderina, quantificados através de imunofluorescência e do qRT-PCR. As linhagens mamárias humanas MCF-7 (não-metastática) e MDA-MB-231 (metastática) foram tratadas com metformina e inibidor Y27632, após a indução da EMT por TGF-β1 para examinar os efeitos sobre a migração destas células, bem como a expressão proteica dos marcadores ROCK-1, vimentina, E-caderina, CD44 e CD24 por imunocitoquímica. Em um estudo in vivo, as células não modificadas CF41 ou que expressam TGF-β1 shRNA foram injetadas na região inguinal de camundongos fêmea nude atímicos tratados com metformina. Os camundongos foram eutanasiados após o tratamento e os pulmões foram recolhidos para avaliação do número de metástases. As regiões metastáticas foram subsequentemente avaliadas pela expressão de N-caderina, E-caderina, vimentina e claudina-7 através da imuno-histoquímica. Foi possível avaliar que a taxa de migração e invasão foi menor em células TGF-β1sh, em comparação com as células parentais CF41 e esta inibição foi significativa quando combinado com o tratamento com metformina. As análises in vitro demonstraram que o tratamento com metformina reduziu a expressão de N-caderina e aumentou a expressão de E-caderina nas células CF41 e TGF-β1sh. Os resultados demonstram também que após a indução do TGF-β1 nas linhagens MCF-7 e MDA-MB-231 houve menor expressão das proteínas ROCK-1, vimentina, CD44 e CD24 em ambas as linhagens após tratamento com metformina e Y27632. Nas células MDA-MB-231 a expressão de E-caderina foi maior em todos os grupos de tratamento. O tratamento da linhagems MDA-MB-231 com metformina e Y27632 reduziu significativamente a invasão destas células. O estudo in vivo demonstrou que o tratamento com metformina reduziu o número de metástases pulmonares em animais portadores de tumores induzidos com as células TGF-β1sh. Houve diminuição da expressão de marcadores mesenquimais N-caderina e vimentina, e aumento da expressão de marcadores epiteliais E-caderina e claudina-7 nas metástases pulmonares. Assim, concluimos que este estudo confirma os benefícios do silenciamento do TGF-β1, além do tratamento com metformina e Y27632 como potenciais agentes terapêuticos em tumores de mama, bloqueando o processo de EMT e seu potencial metastático. / Epithelial mesenchymal transition (EMT) is the process by which cancer cells from primary tumors pass through a phenotypic conversion to invade and migrate, generating metastases in organs or tissues distant. This process can be induced by growth factors such as transforming growth factor beta (TGF-β) and its overexpression has been implicated in tumor angiogenesis, cell migration and invasion in many cancers. ROCK-1 expression is associated with the malignant character of tumors, while inhibiting this molecule results in a significant suppression of tumor metastasis. Metformin, a drug use for the treatment of diabetes, was previously shown to inhibit EMT by suppressing expression of key transcription factors in breast cancer cells. The aims were to evaluate the gene expression and protein expression of related markers metastasis, in a study in vitro and in vivo in breast cancer cell lines after treatment with metformin in addition to the gene silencing of TGF-β1 for inhibiting epithelial-mesenquimal transition. These aims were contemplated performing transfected of canine metastatic mammary tumor cell line CF41 with small interfering RNA constructs to develop clonal derivatives expressing reduced levels of TGF-β1 (TGF-β1sh cells). This was subsequently combined with metformin treatment, to look at effects on cell migration, as well as the expression of the EMT markers E-cadherin and N-cadherin, which were quantified by immunofluorescence and qRT-PCR. MCF-7 and MDA-MB-231 cell lines were treated with metformin and Y27632, after induction of EMT by TGF-β1, to examine the effects on cell migration as well as the protein expression of the ROCK-1 markers, vimentin, E-cadherin, CD44 and CD24 by immunocitochemistry. In an in vivo study, unmodified or TGF-β1 shRNA-expressing CF41 cells were injected in the inguinal region of nude athymic female mice that were treated with metformin. Mice were sacrificed after treatment and the lungs were collected to assess the number of metastases. Metastatic nodules were subsequently assessed for, N-cadherin, E-cadherin, vimentin and claudin-7 expression via immunohistochemistry. With the obtained results it was possible to assess the migration and invasion rate was lower in TGF-β1sh cells as compared to parental CF41 cells and this inhibition was significant when combined with metformin treatment. In vitro analyses demonstrated that metformin treatment reduced n-cadherin expression and increased E-cadherin expression in both CF41 and TGF-β1sh cells. After TGF-β1 induction in MDA-MB231 and MCF-7 cell lines, there was a lower protein expression of ROCK-1, vimentin, CD44 and CD24 in both cell lines after treatment with metformin and Y27632. In MDA-MB-231 cells, E-cadherin expression was increased in all treatment groups. Treatment of MDA-MB-231 cell line with metformin and Y27632 significantly reduced the invasion of these cells. In vivo studies demonstrated that metformin treatment reduced the number of lung metastases in animals bearing TGF-β1sh tumors. This paralleled a decreased expression of mesenchymal markers N-cadherin and vimentin, and increased expression of epithelial markers E-cadherin and claudin-7 in lung metastases.This study confirms the benefits of TGF-β1 silencing in addition to metformin and Y27632 as potential therapeutic agents in mammary tumors, by blocking EMT process and metastatic potential. / FAPESP: 2012/09778-1
97

Regulation of cell polarity and invasion by TGF-β and BMP signaling

Shahidi Dadras, Mahsa January 2017 (has links)
Transforming growth factor β (TGF-β) and bone morphogenetic protein (BMP) signaling pathways are involved in many physiological processes during embryonic and adult life. TGF-β promotes epithelial to mesenchymal transition (EMT). We identified a gene target of TGF-β signaling, encoding the salt-inducible kinase 1 (SIK1). A potential substrate of this kinase, the polarity protein Par3, is an established regulator of tight junction assembly. SIK1 associates with Par3, can potentially phosphorylate Par3 and leads to its degradation, contributing to tight junction disassembly. Glioblastoma multiforme (GBM) is a common malignancy in the central nervous system, characterized by high heterogeneity, invasiveness, and resistance to therapy. One of the causes of heterogeneity and therapy-resistance is the existence of glioblastoma stem cells (GSCs). TGF-β signaling promotes self-renewal while BMP signaling induces differentiation of GSCs. Snail is a potent inducer of the EMT in carcinomas. However, in the context of GBM, Snail induces BMP signaling and represses TGF-β signaling through interaction with SMADs, the signaling mediators of TGF-β and BMP. In conclusion, Snail differentially regulates the activity of the opposing BMP and TGF-β pathways, thus promoting an astrocytic fate switch and repressing stemness in GSCs. Although profound changes in cell polarity is a hallmark of invasive malignancies, little is known about the role of the polarity machinery in tumor suppression. Patient transcriptomic data suggested low Par3 expression, correlating with poor survival of the GBM patients. Par3 silencing decreased the GSC self-renewal capacity and enhanced their invasiveness. Transcriptomic analysis indicates that loss of Par3 leads to downregulation of genes encoding mitochondrial enzymes that generate ATP. These results support a novel role of Par3 in GBM, beyond its contribution to junctional contacts between cells. Another regulator of TGF-β and BMP signaling is the liver kinase B1 (LKB1). According to GBM patient mRNA analysis, high levels of LKB1 correlate with poor prognosis. Silencing of LKB1 in GSCs impairs invasion and self-renewal capacity due to downregulation of genes involved in these processes. Moreover, loss of LKB1 induces mitochondrial dysfunction, leading to decreased ATP levels. Collectively, this thesis has delivered a group of novel regulatory pathways that control critical aspects of cancer cell polarity, invasion and stemness.
98

A PILOT STUDY EXPLORING THE ROLE OF IRAP IN SENESCENT CELLS

Tawfik, Dalya January 2020 (has links)
Insulin regulated aminopeptidase (IRAP) was first identified in fat and muscle cells where it is believed to regulate GLUT4 translocation. It has since been found to be behind a variety of functions, many not yet fully understood. Preliminary research from Monash University suggested that IRAP may play a role in cellular senescence. Senescence is a term that describes arrested cell division and is a tumor repressive mechanism. Senescent cells have been shown to secrete, among other things, the growth hormone TGFβ1, which in turn plays an important role in the cell differentiation of fibroblasts to myofibroblasts. The potential link between IRAP and senescence was the basis of this work. Senescent fibroblasts from three different passages (n=3) in the BJ3 cell-line were cultured and treated with different IRAP inhibitors; ANG-4, AL06 and HFI-419 which were all compared to an untreated control group. They were marked with a β-galactosidase stain, a senescent cellmarker, and imaged. The study demonstrated that the IRAP inhibitors led to a certain decrease in % of senescent cells compared to the control groups. However, this reduction was not considered statistically significant. Similarly, inhibition of the enzyme did not indicate any influence over the differentiation of the cells. The lack of effect could be due to chance based on the low number of sample size, or the condition of the cells used in the trial as they were partially immortalized BJ fibroblasts well beyond the passage of their intended use. In order to further demonstrate an association between IRAP and senescence, further trials are required. / Insulin reglerad aminopeptidas (IRAP) introducerades till en början som ett markörprotein. Man har sedan dess funnit att den står bakom en rad olika funktioner, många ännu inte  fullt klarlagda. Preliminär forskning från laboratoriet i Monash University tydde på att IRAP kan ha en koppling till senescerande fibroblaster. Senescence är en term som beskriver upphörd celldelning och är en tumörrepressiv mekanism. Senescerande celler har påvisats utsekrera bland annat tillväxthormonet TGFβ1, som i sin tur spelar en viktig roll i celldifferentieringenav fibroblaster till myofibroblaster. Den potentiella kopplingen mellan IRAP och senescence låg som grund till detta arbete. Senescerande fibroblaster från tre olika kulturer (n=3) i BJ3-cellinjen odlades och behandlades med olika IRAP-inhibitorer; ANG-4, AL06 och HFI-419 som alla jämfördes med en kontrollgrupp. Därefter markerades de med en β-galaktosidas-markör, en markör för senescerande celler, och mikroskoperades. Studien påvisade att IRAP-inhibitorerna ledde till en viss procentuell minskning av senescerande celler jämfört med kontrollgrupperna. Dock bedömdes inte denna minskning som statistiskt signifikant i studien. Likväl fann man ingen procentuell minskning av differentierade fibroblaster. Hypotetiskt sett skulle man vilja se att reduktionen av senescerande celler motsvarade en nedreglering av TGFβ1-proteiner. Eftersom närvaron av TGFβ1 tros spela en ledande roll i celldifferentiering till myofibroblastfenotypen, bör den procentuella mängden differentierade cellerna minska med inhibitorbehandlingarna. Den bristande påverkan av enzyminhibitionen kan bero på en rad olika faktorer. Cellerna som användes under försökets gång var väl bortom deras brukliga användningscykel. För att vidare påvisa ett potentiellt samband mellan IRAP och senescence behöver vidare försök utföras.
99

The role of AmotL2 in the regulation of mesenchymal transitioning of endothelial cells

Monteiro, Anita-Ann January 2023 (has links)
Background During development, endothelial cells acquire mesenchymal-like properties to migrate and facilitate normal vascular formation. This process of transformation is known as endothelial to mesenchymal transition (EndMT) and has also been implicated in diseases like vascular pathologies contributing to endothelial inflammation, atherosclerosis and tumour angiogenesis. The Angiomotin family of scaffold proteins play a role in transducing mechanical force at cell junctions. Of this family, Angiomotin-Like 2 (AmotL2) localises to endothelial cell junctions and was recently found to play a role in regulating endothelial cell mechanosensing and inflammation. Methods/Materials Primary human endothelial cell lines (HUVEC) were cultured and manipulated in vitro to investigate the role of AmotL2 in EndMT. Lentiviral short hairpin RNA interference was employed in AmotL2-loss-of-function studies, (produced using HEK - Human Embryonic Kidney - cells) to generate knockdown(kd) cells. Western blotting (WB) was used to assess AmotL2 depletion and changes in protein expression of key EndMT markers. qPCR was performed to look at the same at a transcriptional level. Immunofluorescent staining and confocal imaging were performed to validate WB and qPCR results as well as to study protein localisation. Results AmotL2 was found to regulate Snail1 and N-cadherin at both protein and mRNA levels. Morphological findings displayed the AmotL2kd cells to be elongated, deviating from the regular cobblestone morphology observed in control cells. An increase in scaffold protein levels was observed in the AmotL2 kd samples. Similar results were seen in qPCR data where increased mRNA expression was observed in the AmotL2 kd samples for the same targets. On analysis of IF image data, more nuclear staining was observed in the kd samples. qPCR analysis done on samples treated with TGF-β, exhibited an increase in mRNA expression of targets involved in the EndMT pathway in the treatment samples against the controls. Conclusion The results suggest that AmotL2 plays a role in EndMT by affecting the transcription factors and proteins involved in the pathway, which leads to changing morphology and behaviour of the cells. Looking into more targets involved in EndMT may give us a better understanding of how this process leads to diseases like atherosclerosis and tumour angiogenesis.
100

Étude des conséquences fonctionnelles de la mutation SGO1 K23E sur la voie de signalisation TGF-β

Gosset, Natacha 06 1900 (has links)
No description available.

Page generated in 0.0311 seconds