161 |
Multiscale Modeling and Image Analysis of Epithelial Tissuesand Cancer DynamicsHirway, Shreyas U. 30 September 2022 (has links)
No description available.
|
162 |
The Requirement of Matrix Metalloproteinase 2 and 9 in Transforming Growth Factor Beta Induced Epithelial to Mesenchymal Transition of Lens Epithelial CellsPino, Giuseppe 04 1900 (has links)
<p><strong> </strong>Fibrotic cataracts such as anterior subcapsular cataract (ASC) are induced by transforming growth factor beta (TGFβ). The mechanism which governs TGFβ-mediated ASC has not been elucidated. What is known is that TGFβ initiates the conversion of lens epithelial cells (LECs) to myofibroblast-like cells, through a process known as epithelial to mesenchymal transition (EMT). TGFβ-induced EMT leading to ASC has been associated with the upregulation of two matrix metalloproteinases (MMPs), MMP2 and MMP9. However, roles for either of these MMPs have yet to be established in ASC. To determine the involvement of MMP2 and MMP9 I used synthetic inhibitors in conjunction with an established <em>ex vivo </em>rat lens model initiated by TGFβ. The results demonstrated that co-culturing rat lenses with TGFβ and the matrix metalloproteinase inhibitor (MMPI), GM6001 or an MMPI specific for MMP2/9 suppressed ASC. Additionally, studies conducted on the conditioned media from these treatments revealed that TGFβ induces the cleavage of E-cadherin ectodomain which is suppressed by coculturing with either MMPI. To further delineate a role for MMP9 <em>in vivo</em>, ASC formation was examined in two models of lens specific TGFβ overexpression in the absence of functional MMP9. Adenoviral delivery of TGFβ to the anterior chamber of the eye in the absence of functional MMP9 resulted in complete suppression of ASC. Similarly, lens specific TGFβ overexpression in the absence of MMP9 suppressed ASC in 75% of mouse lenses. Additional studies determined that connective tissue growth factor is able to mediate ASC, albeit to a lesser degree than TGFβ.</p> / Doctor of Philosophy (PhD)
|
163 |
Internally Translated Cx43 Isoform GJA1-20k Affects Epithelial to Mesenchymal Transition and Metastatic Cancer Cell BehaviorYoung, Kenneth Lee, II 08 August 2024 (has links)
Epithelial-mesenchymal transition (EMT) is a trans-differentiation program essential for development and wound healing that is pathologically activated during cancer progression. During this process, cells undergo complex changes at the transcriptional and translational levels leading to dissolution of cell-cell junctions, loss of apical-basal polarity, and cytoskeleton reorganization. Transforming Growth Factor-β (TGF-β) is well-established in driving cancer progression through EMT induction. Remodeling of cellular junctions, including gap junctions, is critical to acquiring migratory and invasive characteristics during EMT. The gene GJA1 encodes for Connexin43 (Cx43), the most ubiquitously expressed gap junction protein where altered regulation of Cx43 is associated with cancer progression. Intriguingly, Cx43 mRNA undergoes alternative ‘internal’ translation initiation, generating N-terminally truncated isoforms, including GJA1-20k, which regulates Cx43 gap junction formation. We have previously demonstrated GJA1-20k expression is inhibited during TGF-β-induced EMT, limiting gap junction formation; however, the relationship between GJA1-20k modulation of gap junction localization and cellular invasion and migration remains unknown. Given the role GJA1-20k has in regulating gap junctions, we hypothesize that suppression of GJA1-20k expression promotes metastatic trait acquisition through limiting gap junction formation. Utilizing lentivirally transduced stable mouse mammary gland epithelial (NMuMG) and triple-negative human breast epithelial (MDA-MB-231) cells expressing GJA1-20k, or Lac Z as control, we tested effects on TGF-β-induced EMT induction and metastatic trait induction. Boyden chambers, would/scratch assays were employed to analyze cell invasion and migration respectively. We found GJA1-20k overexpression during EMT results in decreased cell invasion and migration to LacZ controls. Future directions include evaluation of GJA1-20k restoration in a metastatic breast cancer model in vivo. Investigating the underlying role of GJA1-20k in EMT-induced cell junction remodeling could be promising as a potential pharmacological target process independent of transcriptional or post-translational pathways. Ultimately, by adding novel information in the expanding and compelling field of translational control, this work could aid in developing the future of precision medicine as new therapeutic solutions to treat cancer will require limiting cancer cell’s ability to metastasize. / R01 HL132236 JWS
R41 CA250874 SL
R01HL132236 Diversity Supplement KLY II
23PRE1025483 AHA Predoctoral Fellowship KLY II / Doctor of Philosophy / Every organ system relies upon cell-to-cell communication to properly function and is the basis of multi-cellular life. Gap junctions are nanoscale conduits allowing the passage of small signaling molecules and ions between adjacent cells, similar to telephone tubes. Gap junctions are formed from proteins called connexins. Interestingly, it is well known that shorter pieces of connexin proteins can regulate the formation of gap junctions and are uniquely created by a process called alternative ‘internal’ translation. Changes in the amounts of short-length and full-length connexin proteins are often found in cancer cells. Cancer is the uncontrolled growth of abnormal cells. Commonly, the morphology of cancer cells, and the way they communicate with neighboring cells, is altered. Cancer progression is aided by changes in cell signaling molecules, including TGF-β which can drive cancer cells to leave primary tumor sites and grow elsewhere in the body. This is important for the cancer cells to continue dividing and eventually metastasizing (invading other organ systems). Treating cancer once it has spread to other regions of the body is difficult and is the main cause of cancer deaths worldwide. Using TGF-β to model metastatic changes in mouse and human cell lines, we studied how short-length connexin protein affects metastatic cancer cell behavior. With this information we will be able to guide the development of druggable alternative ‘internal’ translation targets, by restoring the proper communication between neighboring cells and therefore preventing spread of cancer cells.
|
164 |
Implications physiopathologiques de la Nestine lors du remodelage pulmonaire et cardiaque à la suite de l’infarctus du myocarde, du diabète et de l’hypertension pulmonaireChabot, Andréanne 07 1900 (has links)
Il est reconnu que la protéine filamenteuse intermédiaire Nestine est exprimée lors du processus de cicatrisation et du remodelage fibrotique. De plus, nous avons identifié l’expression de la Nestine au sein de deux populations distinctes qui sont directement impliquées dans les réponses de fibroses réparative et réactive. Ainsi, une population de cellules souches neurales progénitrices résidentes du coeur de rat adulte exprime la Nestine et a été identifiée à titre de substrat de l’angiogenèse et de la neurogenèse cardiaque. Également, la Nestine est exprimée par les myofibroblastes cicatriciels cardiaques et il a été établi que la protéine filamenteuse intermédiaire joue un rôle dans la prolifération de ces cellules. Ainsi, l’objectif général de cette thèse était de mieux comprendre les évènements cellulaires impliqués dans la réponse neurogénique des cellules souches neurales progénitrices résidentes cardiaques Nestine(+) (CSNPRCN(+)) lors de la fibrose réparative cardiaque et d’explorer si l’apparition de fibroblastes Nestine(+) est associée avec la réponse de fibrose réactive secondaire du remodelage pulmonaire. Une première publication nous a permis d’établir qu’il existe une régulation à la hausse de l’expression de la GAP43 (growth associated protein 43) et que cet événement transitoire précède l’acquisition d’un phénotype neuronal par les CSNPRCN(+) lors du processus de cicatrisation cardiaque chez le rat ayant subi un infarctus du myocarde. De plus, la surimposition de la condition diabétique de type 1, via l’injection unique de Streptozotocine chez le rat, abolit la réponse neurogénique des CSNPRCN(+), qui est normalement induite à la suite de l’ischémie cardiaque ou de l’administration de 6-hydroxydopamine. Le second article a démontré que le développement aigu de la fibrose pulmonaire secondaire de l’infarctus du myocarde chez le rat est associé avec une augmentation de l’expression protéique de la Nestine et de l’apparition de myofibroblastes pulmonaires Nestine(+). Également, le traitement de fibroblastes pulmonaires avec des facteurs de croissances peptidiques pro-fibrotiques a augmenté l’expression de la Nestine par ces cellules. Enfin, le développement initial de la condition diabétique de type 1 chez le rat est associé avec une absence de fibrose réactive pulmonaire et à une réduction significative des niveaux protéiques et d’ARN messager de la Nestine pulmonaire. Finalement, la troisième étude représentait quant à elle un prolongement de la deuxième étude et a alors examiné le remodelage pulmonaire chronique chez un modèle établi d’hypertension pulmonaire. Ainsi, les poumons de rats adultes mâles soumis à l’hypoxie hypobarique durant 3 semaines présentent un remodelage vasculaire, une fibrose réactive et une augmentation des niveaux d’ARN messager et de la protéine Nestine. De plus, nos résultats ont démontré que la Nestine, plutôt que l’alpha-actine du muscle lisse, est un marqueur plus approprié des diverses populations de fibroblastes pulmonaires activés. Également, nos données suggèrent que les fibroblastes pulmonaires activés proviendraient en partie de fibroblastes résidents, ainsi que des processus de transition épithélio-mésenchymateuse et de transition endothélio-mésenchymateuse. Collectivement, ces études ont démontré que des populations distinctes de cellules Nestine(+) jouent un rôle majeur dans la fibrose réparative cardiaque et la fibrose réactive pulmonaire. / It is well established that the intermediate filamentous protein Nestin is expressed during wound healing and fibrotic remodeling. Furthermore, we have identified Nestin expression in two distinct populations directly implicated in reparative and reactive fibrosis. The adult rodent heart contains a resident population of neural progenitor/stem cells that express Nestin and identified as a cellular substrate of cardiac angiogenesis and neurogenesis. Moreover, Nestin is also expressed in cardiac scar myofibroblasts and the intermediate filament protein plays a direct role in proliferation. Thus, the general aim of the present thesis was to better understand the cellular events implicated in the neurogenic response of neural progenitor/stem cells during cardiac reparative fibrosis and to explore whether the appearance of Nestin(+)-fibroblasts was associated with reactive fibrotic response secondary to pulmonary remodeling. The first study revealed that the transient upregulation of growth associated protein 43 (GAP43) represents a transition event during the acquisition of a neuronal-like phenotype by cardiac resident neural progenitor/stem cells in the scar of the infarcted rat heart. Furthermore, the superimposition of a type 1 diabetic environment, via the single injection of streptozotocin in rats, abrogated the neurogenic response of cardiac resident neural progenitor/stem cells to ischemia and 6-hydroxydopamine, respectively. The second study has demonstrated that the development of acute pulmonary fibrosis secondary to myocardial infarction of the adult rat heart was associated with the increased expression of Nestin protein levels and appearance of Nestin(+)-myofibroblasts. Furthermore, the treatment of pulmonary fibroblasts with putative pro-fibrotic peptide growth factors increased Nestin protein levels. Lastly, in the lungs of type 1 diabetic rats, the absence of a reactive fibrotic response was associated with a significant downregulation of Nestin protein/mRNA levels. Finally, the third study represented an extension of the second study and examined chronic lung remodeling in an established model of pulmonary hypertension. The lungs of adult male rats subjected to 3 weeks of hypobaric hypoxia were associated with vascular remodeling, reactive fibrosis and increased Nestin protein and mRNA levels. Moreover, Nestin, rather than smooth muscle α-actin expression was identified as a more relevant marker of activated pulmonary fibroblasts. Furthermore, the appearance of activated pulmonary fibroblasts may be derived in part from resident fibroblasts and secondary to endothelial-mesenchymal transition and epithelial-mesenchymal transition. Collectively, these studies have demonstrated that distinct populations of Nestin-expressing cells play a seminal role in cardiac reparative fibrosis and pulmonary reactive fibrosis.
|
165 |
Rôle de la GTPase ARF1 dans la migration et l’invasion des cellules du cancer du seinSchlienger, Sabrina 03 1900 (has links)
La capacité des cellules à être invasives et métastasiques est une caractéristique fondamentale de la malignité tumorale. Nous avons récemment montré que le facteur d’ADP-ribosylation 1 (ARF1) est surexprimé dans les lignées cellulaires hautement invasives du cancer du sein et que la stimulation du récepteur au facteur de croissance épidermique (EGFR) peut activer cette isoforme pour contrôler la migration ainsi que la prolifération. Cependant, le rôle de cette GTPase dans la régulation du processus d’invasion cellulaire et les mécanismes moléculaires associés demeure inconnu. Nous avions comme objectifs dans cette thèse, de définir les voies de signalisation sous le contrôle d’ARF1 dans les cellules de cancer du sein et démontrer que l’expression et l’activation de cette GTPase est associée à un phénotype hautement invasif.
Nos études démontrent que la modulation de l'expression et l'activité d’ARF1 affecte la capacité des cellules MDA-MB-231 (pour M. D. Anderson-metastatic breast-231), une ligne hautement invasive, à dégrader la matrice extracellulaire via l'activité de la métalloprotéinase MMP-9. ARF1 contrôle les deux principales structures impliquées dans l'invasion, en jouant sur la maturation d’invadopodes ainsi que la relâche de microvésicules membranaires. D’un point de vue mécanistique, l'axe de signalisation ARF1, RhoA-RhoC et la chaine légère de la myosine (MLC) explique ces phénomènes.
De plus, nous démontrons que l'un des mécanismes par lequel ARF1 régule la migration est en contrôlant l'assemblage des points adhésions focaux et ce, dans plusieurs types de cellules cancéreuses du sein. ARF1, en étant un membre du complexe d’adhésion, réglemente le recrutement et l’activité de protéines clés à la β1-intégrine tels que la paxilline, la talin et la kinase d’adhésion focale (FAK).
Pour finir, nous rapportons que ARF1 et ARF6 ont un rôle majeur dans la transition épithélio-mésenchymateuse. ARF1 est retrouvé fortement exprimé dans les tissus de sous-types les plus agressifs et les plus avancés de cancer du sein. Dans un modèle murin, la modulation à la baisse de l’expression d’ARF1 dans les cellules MDA-MB-231 corrèle avec la diminution de croissance des tumeurs primaires et l’installation des métastases pulmonaires. De plus, nous rapportons que la surexpression des ARF dans des cellules non invasives, les MCF7 (pour Michigan Cancer Foundation-7), permet la nidification de métastases. En effet, dans les MCF7, ARF1 contrôle l’adhésion intercellulaire via la β-caténine et l’E-cadhérine, promeut l’activation de l’oncogène Ras (pour Rat Sarcoma/ Rat Fibrosarcoma virus) et l’expression de plusieurs inducteurs de transition épithélio-mésenchymateuse comme snail et slug. De plus, ARF1 contrôle l’invasion, la prolifération cellulaire et même la résistance à certains agents chimio-thérapeutiques.
Globalement, nos études identifient ARF1 comme un interrupteur moléculaire de la progression tumorale et suggèrent que la limitation de son expression/activité pourrait améliorer le devenir des patients atteints du cancer du sein. / Invasive and metastatic chapacities are fundamental features for tumor malignancy. We have recently shown that the ADP-ribosylation factor 1 (ARF1) is over-expressed in highly invasive breast cancer cell lines and stimulation of the epidermal growth factor receptor (EGFR) may activate this isoform to regulate migration and proliferation. However, the role of this GTPase in regulating cell invasion process and related molecular mechanisms remain unknown. In this thesis, we had as objectives, to define the signaling pathways under the control ARF1 in breast cancer cells and show that the expression and activation of the GTPase is associated with highly invasive phenotype.
Our studies show that the modulation of the expression and activity of ARF1 affect the ability of MDA-MB-231 cells (M. D. Anderson-metastatic breast-231), a highly invasive line, to degrade the extracellular matrix via the activity of the metalloproteinase MMP-9. ARF1 controls the two main structures involved in the invasion, playing on invadopodia maturation and shedding of membrane microvesicles. The molecular mechanisms involve the regulation of RhoA and RhoC activity by ARF1 and the following downtream events associated with and the myosin light chain (MLC) phosphorylation.
Furthermore, we demonstrate that ARF1 also regulates migration by controlling the assembly of focal adhesion complexes in many types of breast cancer cells. ARF1, also prensent in adhesion complexes, regulates the recruitment and activity of key proteins such as paxillin, talin and focal adhesion kinase (FAK) to β1 integrin.
Finally, we report that ARF1 and ARF6 play a major role in the epithelial-mesenchymal transition (EMT). ARF1 is found highly expressed in tumor tissue of the most aggressive and advanced subtypes of breast cancer. Lowered expression of ARF1 in vivo in the MDA-MB-231 cells impars tumor growth in primary tumors and inhibits lung metastasis. We report that upregulation of the ARF in non-invasive cells, MCF7 (Michigan Cancer Foundation-7) induce metastasis nidification. Indeed, we show in MCF7 that ARF1 controls intercellular adhesion via the β-catenin and E-cadherin, promotes Ras (Rat Sarcoma/ Rat Fibrosarcoma virus) oncogene activation, and conrols expression of several epithelial-mesenchymal transition markers such as snail and slug. Moreover, we demonstrate that ARF1 controls invasion, proliferation and even resistance to certain chemo-therapeutic agents, in MCF7 cells.
Overall, our studies identify ARF1, as a molecular switch of tumor progression and suggest that limiting its expression / activity could improve the outcome of breast cancer patients.
|
166 |
Odpověď metastatických buněčných linií karcinomu prostaty na genotoxický stres / Odpověď metastatických buněčných linií karcinomu prostaty na genotoxický stresImrichová, Terezie January 2013 (has links)
Prostate cancer is the fourth most frequent cause of cancer-related deaths in men worldwide. One of current successful approaches to treat prostate cancer is radical prostatectomy followed by radiotherapy. However, this treatment is not 100% successful, as 53% patients develop secondary tumors. Our hypothesis is, that ionizing radiation itself contributes to the development of metastases by inducing changes in cell phenotype, particularly in terms of epithelial-to-mesenchymal transition and stemness. To test this hypothesis, we irradiated the cells of metastatic prostate cancer cell line DU145 by fractionated radiation 2 x 10 Gy and we compared the expression of selected epithelial, mesenchymal and stem-cell markers prior to and after irradiation. Besides we focused on a subpopulation of so called floating cells which arise during irradiation. These cells can survive the radiation treatment and after some time they are able to reattach and give rise to readherent population. We wanted to asses what is the cell cycle profile of these cells and whether and how fast they proliferate. In this thesis we have shown that radiation causes only minor changes in epithelial/mesenchymal and stem-like character of adherent fraction of the DU145 cell line. However, we have also described that small population of...
|
167 |
O envolvimento da remodelação da cromatina no controle do comportamento agressivo dos carcinomas epidermoides de cabeça e pescoço / The involvement of chromatin remodeling in the control of head and neck squamous cell carcinoma behaviorGiudice, Fernanda Salgueiredo 10 December 2012 (has links)
Modificações nas histonas são conhecidas por regular a estrutura conformacional da cromatina e a expressão gênica em células adultas e células-tronco pluripotentes. Tem sido postulado que a acetilação e deacetilação das histonas podem influenciar a expressão de genes envolvidos na iniciação, progressão e metástase tumoral, além de contribuir para o desenvolvimento de resistência à quimioterapia. Assim, buscou-se avaliar a influência das modificações nas histonas sobre a biologia do carcinoma epidermoide de cabeça e pescoço (CECP) e sua respectiva subpopulação de células semelhantes às células-tronco (CSC). Inicialmente, foi checado os níveis de acetilação da histona H3 (membro das histonas nucleares associado à compactação da cromatina) em um painel representativo de linhagens celulares de CECP. Posteriormente, para estudar a influência do estroma tumoral no padrão de acetilação da histona H3, o microambiente do tumor foi mimetizado através da utilização de meio condicionado derivado do cultivo de fibroblastos e cultura primária de células endoteliais humanas. Além disso, validamos esses resultados in vitro por meio de amostras humanas de CECP. Finalmente, a acetilação e deacetilação da cromatina foi induzida, respectivamente, pela administração dos inibidores das enzimas histona deacetilase tricostatina A (TSA) e histona acetiltransferase curcumina, em linhagens celulares de CECP. Foi feita a análise da formação de esferas (ensaio funcional de células-tronco), juntamente com a verificação dos níveis de ALDH, marcador de células-tronco (citometria de fluxo - FACS), além da determinação do índice de proliferação tumoral (Ki-67) e realização dos ensaios de invasão e migração celular. Linhagens celulares de CECP apresentaram níveis baixos de acetilação da histona H3 e demonstraram capacidade de retenção de uma subpopulação de CSC. Apenas o meio condicionado de células endoteliais humanas foi capaz de alterar a conformação da cromatina, uma vez que induziu o aumento da acetilação da histona H3. Interessantemente, foi também notado um concomitante aumento da agressividade de linhagens celulares de CECP (aumento dos níveis de BMI-1 e vimentina). Esses resultados foram confirmados em amostras humanas de CECP que mostraram, apenas no fronte de invasão, células com cromatina acetilada. Curiosamente, essas mesmas células também expressaram vimentina. Os tratamentos com TSA e curcumina resultaram na diminuição significativa da subpopulação de CSC, interrompendo a formação de esferas e reduzindo os níveis de ALDH. Além disso, o tratamento com curcumina mostrou resultados muito interessantes, uma vez que gerou uma redução evidente da invasão celular e impactou por completo o potencial de migração tumoral, sendo nesse sentido mais eficiente que a cisplatina, droga antineoplásica bem estabelecida. Por outro lado, o tratamento com TSA induziu a transição epitélio-mesenquimal nas linhagens celulares de CECP, detectada pelo aumento da expressão de vimentina e indução de um fenótipo fusiforme, juntamente com o aumento da invasão tumoral e os níveis de BMI-1. Portanto, a organização da cromatina está envolvida na modulação da presença de CSC e os altos níveis de acetilação das histonas intensificam o comportamento agressivo de células de CECP. / Histone modifications are known to regulate chromatin conformation structure and gene expression in adult cells and pluripotent stem cells. It has been postulated that histone acetylation and deacetylation could influence the expression of genes involved in cancer initiation, progression, metastasis, and development of resistance to chemotherapies. Here, we sought to evaluate the influence of histone modifications over the biology of head and neck squamous cell carcinoma (HNSCC) and its stem cell-like subpopulation (CSC). Initially, we checked the status of histone H3 acetylation (a member of the core histones associated to chromatin compaction) in a representative set of HNSCC cell lines. Subsequently, to analyze the influence of tumor stroma over the histone H3 acetylation, we mimicked the tumor microenvironment by using conditioned medium from fibroblasts and primary human endothelial cells. Further we validated these in vitro findings through human samples of HNSCC. Finally, we induced chromatin acetylation and deacetylation by the administration of the histone deacetylase inhibitor trichostatin A (TSA) and histone acetyltransferase inhibitor curcumin, respectively, in HNSCC cell lines. The analysis of spheres formation (stem cell functional assay), along with the levels of stem cells marker ALDH (showed by flow cytometry - FACS), tumor proliferation index (Ki-67), invasion and migration cellular potencial were verified. HNSCC cell lines showed lower levels of histone H3 acetylation and ability to retain a subpopulation of CSC. Only conditioned media from human endothelial cells was able to alter the conformation of chromatin, since it induced the increase of histone H3 acetylation. Interestingly, it was also noted a concomitant augment of HNSCC cell lines aggressiveness (enhanced BMI-1 and vimentin levels). These findings were confirmed in human samples of HNSCC that showed, only at the invasive front, cells with acetylated chromatin. Curiously, these same cells also expressed vimentin. TSA and curcumin treatments resulted in significant decrease of the CSC subpopulation by disrupting the spheres and reducing the levels of ALDH. Also, curcumin treatment showed exciting results since it caused an evident reduction of cellular invasion and it impacted the tumoral migration potential, being more efficient than cisplantin, a well-established antineoplastic drug. However, TSA induced epithelial to mesenchymal transition in HNSCC cell lines detected by the upregulation of vimentin and the induction of a fusiform phenotype along with augmented tumor invasion and the levels of BMI-1. Chromatin organization is involved in the modulation of CSC where high levels of histone acetylation intensify the aggressive behavior of HNSCC cells.
|
168 |
Role Of Tumor Microenvironment in Breast Cancer MetastasisAparna B. Shinde (5930267) 10 June 2019 (has links)
<p>Metastasis of primary
mammary tumors to vital secondary organs is the primary cause of breast cancer-associated
death, with no effective treatment. Metastasis is a highly selective process
that requires cancer cells to overcome multiple barriers to escape the primary
tumor, survive in circulation, and eventually colonize distant secondary
organs. One of the important aspects of metastatic cancers is the ability to
undergo epithelial-mesenchymal transition (EMT) and the reverse process
mesenchymal-epithelial transition (MET) process. Constant interconversion of
tumor cells between these phenotypes creates epithelial-mesenchymal heterogeneity
(EMH) and interaction between these tumor cell types and the stromal cell
compartment is clearly important to metastasis. In healthy tissues, stromal
cells maintain the composition and structure of the tissue through the production
of extracellular matrix (ECM) proteins and paracrine signaling with epithelial
cells. However, little is known about how EMH
promotes changes in the ECM to promote breast cancer progression and
metastasis. Cancer cells also secret exosomes, nano-size extracellular
vesicles, to establish intercellular communication with distant organs in order
to induce metastasis. These exosomes contain a plethora of different proteins
including extracellular matrix proteins and matrix crosslinking enzymes.
Fibronectin, an important ECM protein, plays an active role in tumor
progression and is often crosslinked by tissue transglutaminase 2 (TGM2) to
promote fibrosis in cancer. Both FN and TGM2 exist in exosomes and are
expressed by heterogenous breast tumors. Although FN and TGM2 have been
reported to play essential roles in cancer, their involvement in metastasis
remains unclear. This work utilizes a variety of approaches to investigate the
role of tumor heterogeneity and ECM proteins in promoting breast cancer
metastasis. In this dissertation, we establish that mesenchymal cells
expressing intracellular FN are held in a stable non-metastatic mesenchymal
phenotype and produce cellular fibrils containing functionalized FN capable of
supporting the growth of metastatic competent epithelial cells. We introduce a
novel 3D culture system consisting of a tessellated scaffold which is capable
of recapitulating cellular and matrix phenotypes <i>in vivo. </i>Further, we
also demonstrate breast tumor cells secrete exosomes containing TGM2
crosslinked FN fibrils to promote premetastatic niche formation and induction
of metastasis.<i> </i>Using genetic approaches, we establish TGM2 is essential
and sufficient to drive metastasis. Finally, we demonstrate pharmacological
inhibition of TGM2 offers a potential therapeutic strategy to treat metastatic
breast cancer. Altogether, our research provides insights into the mechanism
through which TGM2 promotes metastatic breast cancer. This work will help in
developing new drugs to target TGM2 aimed at reducing breast cancer metastasis.<br></p>
|
169 |
Implication du microenvironnement sur la survenue de la maladie métastatique et l’apparition d’une maladie résiduelle dans les adénocarcinomes sein séreux / Implication of Microenvironment on the Onset of Metastasis & Initiation of Residual Disease in Breast AdenocarcinomaGhiabi, Pegah 07 October 2013 (has links)
Le cancer du sein est le cancer le plus fréquemment diagnostiqué et la deuxième cause de décès par cancer dans les pays développé. Récemment le rôle du microenvironnement a été mis en évidence dans l’oncogenèse et la progression tumorale. Plusieurs études ont montré que le microenvironnement tumoral est un élément dynamique en constant dialogue avec les cellules tumorales. Parmi les éléments du microenvironnement les cellules endothéliales jouent un rôle particulier. Effectivement ces cellules constituent la paroi des vaisseaux et permettent l’acheminement des nutriments et de l’oxygène vers la tumeur. Ainsi ces dernières années des thérapies visant a détruire les vaisseaux sanguins ont vu le jour mais n’ont pas permis d’atteindre les progrès thérapeutiques escomptés. Notre groupe ainsi que d’autres ont mis en évidence des rôles des cellules endothéliales indépendant de la perfusion tumorale. Dans ce travail de thèse nous avons caractérisé l’interaction entre cellules endothéliales et tumorales et mis en évidence le rôle pro-tumoral de la niche vasculaire. Nous avons tout d’abord pu montrer en utilisant une stratégie de souris transgénique où nous annulons l’expression de Jag1 des cellules endothéliales la réduction drastique de l’occurrence des métastases. Nous avons pu montrer que cela n’est pas dépendant de la perfusion tumorale mais dépend de la modification de nombreux gènes pro-métastatiques dont Id1 dans les cellules tumorales par les cellules endothéliales. Parallèlement nous avons montré que le dialogue entre cellules tumorales et endothéliales induit une transition mésenchymateuse des cellules endothéliales avec pour conséquence une augmentation de leur survie ainsi que de leur migration et de leur capacité angiocrine.Ainsi nous avons montré comment le dialogue entre cellules tumorales et endothéliales induit une modification du phénotype tumoral et endothéliale et le rôle de la voie Notch dans ce dialogue. Notre travail suggère la possibilité de moduler l’agressivité tumorale en interrompant le dialogue entre cellules endothéliales et tumorales. / Breast cancer is a heterogeneous disease, which is characterized by distinct morphological features and clinical behaviors and is the most commonly diagnosed cancer among women in the United States and worldwide and the second cause of cancer-related mortality in women. Several years of investigation have demonstrated that tumor initiation, progression and metastasis are closely regulated by the adjacent non-neoplastic tissues that are collectively referred to as tumor microenvironment (stroma). The components of tumor stroma such as mesenchymal stem cells have been shown to enhance cancer stem cell population in breast tumor. Also, the endothelial cells (ECs) conventional role in tumor angiogenesis is crucial in determining the tumor fate as microscopic and asymptomatic versus aggressive. This outstanding characteristic of ECs has set them as promising targets in cancer therapy. However, failure of anti-angiogenic therapies despite vessel disruption suggests the blood flow-independent ability of ECs to facilitate tumor growth. In this study, we show that ECs promoted breast cancer cell self-renewal, stemness, migratory characteristic and lung metastasis through Jagged1/Notch dependent Id1 modulation. ECs with Jag1 knock down (ECsJag1-) failed to sustain breast cancer cell proliferation and stemness in vitro and during xenografted tumor growth. Furthermore, we established a breast tumor mouse model with EC specific Jag1 mutation, by crossing the MMTV-PyMT mice with Cdh5-Cre+/-Jag1loxP/loxP mice. It demonstrated significant decrease in primary tumor growth and dramatic reduction in lung metastasis in Cdh5-Cre+/-Jag1loxP/loxPPyMT+ mice. Transcriptome sequencing analysis of the sorted primary tumor cells identified Notch downstream targets, specifically, Id1, which was reported to be essential for lung metastasis of breast tumors. Additionally, we were interested in determining the mechanisms that derive the activation of ECs toward supporting tumor growth and expansion. Previous studies have shown that ECs show tremendous degree of plasticity when placed under different conditions. Here, we showed that ECs show EndMT phenotypes upon having contact with tumor cells. Interestingly, the EndMT transforms the ECs into activated entities with increased proliferation, migration and angiogenesis properties. Our results demonstrated that the EndMT was reversible and dependent on EC-tumor cell contacts. Moreover, we were able to show that the tumor-induced EndMT in ECs is synergistically regulated by TGFβ and notch signaling pathways. Overall, our findings implicate the significance of endothelial-tumor cells perfusion-independent interaction in cancer progression, stemness, and metastasis. Besides, this study might have determined novel targets in combating cancer in a more effective way.
|
170 |
Décryptage des changements épigénétiques impliqués dans la transition épithélio-mésenchymateuse et le cancer / Deciphering the Epigenetic Changes Involved in Epithelial-Mesenchymal Transition and CancerMalouf, Gabriel 15 July 2014 (has links)
La transition épithélio-Mésenchymateuse (TEM) est un processus de plasticité cellulaire qui existe dans le développement embryonnaire et qui permet la formation des tissus et organes. Dans la cancérogénèse, ce processus est réactivé par des facteurs de transcription dont l’action implique très probablement un remodelage de la chromatine. La cartographie exacte de ces changements épigénétiques est peu connue à l’échelle du génome entier, même si il y a eu quelques études antérieures explorant les changements de quelques loci de façon bien ciblée. Ce mémoire traite du remodelage épigénétique médié par le facteur de transcription Twist1 dans un modèle de lignée mammaire immortalisée. L’architecture de ce remodelage a été cartographiée grâce à l’utilisation des techniques de haut-Débit pour analyser la méthylation de l’ADN (DREAM) et les modifications des histones (ChIPseq). Nos résultats montrent un changement majeur du méthylome pendant la TEM avec une hyperméthylation focale et une hypométhylation globale des corps des gènes prédominant au niveau des « domaines partiellement méthylés »; ces domaines sont déjà connus dans le développement pour gagner de façon concomitante à leur hypométhylation des marques d’histone répressives. Nous avons aussi observé un remodelage des domaines de l’histone répressive H3K27me3 avec une réduction de leur taille, et surtout le quasi doublement du nombre de gènes bivalents qui accompagne la transition. Le couplage de la méthylation de l’ADN avec le profil des microRNA nous a permis d’identifier le miR-203 comme l’unique microRNA régulé par méthylation de l’ADN durant la TEM; nous avons aussi montré que l’extinction épigénétique du miR-203 est requise pour la TEM et l’acquistion des propriétés de cellules souches. Enfin, nous avons réalisé une caractérisation génétique et/ou épigénétique de deux cancers rares, les carcinomes fibrolamellaires du foie et les carcinomes du rein à translocation. Pour les carcinomes fibrolamellaires du foie, nous avons décrit la nature endocrine de cette tumeur et établi une signature épigénétique basée sur la méthylation de l’ADN pouvant servir à différencier les formes histologiques appelées « pures » des formes « mixtes ». Pour les cancers du rein à translocation, nous avons montré les bases génétiques et épigénétiques de la différence entre les formes pédiatriques et adultes, avec la découverte fréquente du gain du bras chromosomique 17q dans les formes adultes. Nous avons aussi identifié une mutation récurrente dans le gène qui remodèle la chromatine INO80D appartenant à la famille INO80. En conclusion, ce travail explore le rôle de l’étude de l’épigénome pour comprendre la reprogrammation pendant les processus physiologiques comme la TEM d’une part et le cancer d’autre part. / The epithelial-Mesenchymal transition (EMT) is a process of cellular plasticity that exists in embryonic development and which allows the formation of tissues and organs. In carcinogenesis, the process is reactivated by transcription factors whose action probably involves chromatin remodeling. The exact mapping of these epigenetic changes is poorly understood genome-Wide, although there have been some previous studies exploring changes in so few well-Targeted loci. This thesis deals with the epigenetic remodeling mediated by the transcription factor Twist1 in a model of human mammary immortalized cell line. The architecture of this remodeling has been mapped through the use of high-Throughput techniques to analyze DNA methylation (DREAM) and histone modifications (ChIPseq). Our results suggest a major change in the EMT methylome with focal hypermethylation and gene body hypomethylation predominantly within "partially methylated domains"; these areas are already known in development to gain repressive histone marks concomitantly with DNA hypomethylation. We also observed landscape remodeling of repressive histone mark H3K27me3 with a reduction in domains size, and especially the almost doubling of the number of bivalent genes. The coupling of DNA methylation with the profile of microRNA has allowed us to identify miR-203 as single microRNA regulated by DNA methylation during EMT; we have also shown that epigenetic suppression of miR-203 is both required for EMT and acquisition of stem cell properties. Finally, we performed a genetic and/or epigenetic characterization of two rare cancers, named fibrolamellar hepatocellular carcinomas and translocation renal cell carcinomas. In fibrolamellar hepatocellular carcinoma, we described the endocrine nature of this tumor and established a signature based on DNA methylation which can be used to distinguish histological forms called "pure" from "mixed" fibrolamellar hepatocellular carcinomas. Regarding translocation renal cell carcinomas, we established the genetic and epigenetic basis of differences between pediatric and adult forms, characterized by frequent gain of 17q gain chromosomal arm in adults. We also identified recurrent mutations in the chromatin remodeling gene INO80D which belongs to INO80 family. In conclusion, this work explores the impact of analyzing the epigenome to understand reprogramming during physiological processes such as EMT and cancer.
|
Page generated in 0.1167 seconds