• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 47
  • 21
  • 16
  • 6
  • 3
  • 2
  • 2
  • Tagged with
  • 115
  • 115
  • 114
  • 109
  • 23
  • 22
  • 22
  • 18
  • 18
  • 16
  • 16
  • 15
  • 15
  • 14
  • 12
  • 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.
11

Study of RPC32α, subunit of the RNA polymerase III, in a tumor model / Etude de la régulation de RPC32alpha, sous-unité de l'ARN polymérase III, dans des modèles tumoraux

Bretting, Wiebke 11 December 2017 (has links)
Les ARN polymérases sont des acteurs indispensables de la transcription. Chez les eucaryotes il existe trois ARN polymérases (I, II et III). La ARN polymérase III (Pol III) possède 17 sous-unités, dont une qui existe sous deux formes: RPC32α et RPC32β. Seulement une des deux formes peut être intégrée dans la Pol III, créant ainsi deux polymérases différentes Pol IIIα et Pol IIIβ. Alors que RPC32β est présent dans les cellules somatiques, RPC32α est exprimé surtout dans des cellules souches et des cellules tumorales. Aujourd’hui rien n’est connu sur leurs rôles respectifs. Le cancer du sein est un problème majeur de santé publique car c’est le cancer féminin le plus fréquent. Plusieurs types de cancer du sein sont identifiés selon la présence ou absence de certains récepteurs hormonaux. Des cancers qui testent négative pour le récepteur d’oestrogène et de progestérone et qui ne surexpriment pas le récepteur pour les facteurs de croissance épidermiques humains 2 (HER2) sont appelés triple-négative. Ils ont un pronostique peu favorable, due à l’agressivité de ce type de cancer et un manque de thérapie cibles. Pour étudier le rôle de RPC32α il fallait identifier un model tumorale. En collaboration avec Jean-Paul Feugeas (INSERM UMR 1098) une étude transcriptomique a été fait sur 2627 échantillons cliniques de tissus de sein. L’étude montre que RPC32α est surexprimé dans les cancers triple-négative, alors que son homologue RPC32β est surexprimé dans les tissues normaux. Une analyse sur six lignées de cancer du sein et une ligné non-tumorale ont pu confirmer les résultats de l’analyse transcriptomique. Le modèle de cancer du sein a donc été validé. Une caractérisation des différentes lignées de cancer du sein a démontré que d’autres sousunités de la Pol III n’étaient pas surexprimées dans les cancers triple-négative. La surexpression de RPC32α n’était donc pas une conséquence d’une hyperactivité de la Pol III. Une analyse des transcrits synthétisé par la Pol III a montré que en générale les transcrits de la Pol III étaient plus fortement exprimé dans les cancers triple-négative que dans d’autres cancers. Afin d’étudier l’implication de RPC32α dans les phénomènes de tumorisation, plusieurs lignées cellulaires dépourvues de RPC32alpha ont été créé utilisant la technique CRISPRCAS9. L’absence de RPC32α n’a pas induit une augmentation de transcription ni de l’ARN de 4 RPC32α, ni de celle de RPC32β. Il n’existe donc pas de boucle de rétroaction pour RPC32α et les deux homologues ne sont pas co-régulés. Plusieurs, mais pas tous les transcrits synthétisé par la Pol III ont une expression fortement baissé dans les lignées mutants. Le fait que pas tous les transcrits ne soit affectés par la perte de RPC32α, indique qu’il existe une spécificité de transcription pour Pol IIIα et Pol IIIβ. Les cellules des linges mutants ne présentaient pas de phénotype différent des cellules mères et la croissance était la même dans toutes les lignées. Par contre les tests de croissance en agar-mou ont révélé que les lignées mutants formaient 85% de moins de colonies, indiquant que RPC32α est nécessaire pour la croissance tumorigénique in vitro. Pour tester l’effet de la perte de RPC32α sur la croissance tumorigénique in vivo, des cellules mutants et des cellules mères ont été injecté dans des souris. Les souris greffées avec des cellules mutantes montrent un départ de tumorisation retardé. Au bout de six semaines elles avaient de tumeurs deux fois plus petit que les souris avec des cellules mères. Après ablation de la tumeur primaire, les souris ont été surveillées pour l’apparition de métastases. Quatre semaines plus tard les souris greffées avec des cellules mutantes avaient 100 fois moins de métastases que les souris contrôles. Ces résultats montrent que RPC32α est nécessaire pour la tumorisation in vitro et in vivo. La protéine semble surtout jouer un rôle dans la formation des métastases, qui sont un des problèmes majeurs dans le traitement des cancers. / The RNA polymerases are key players of transcription. Eukaryotes have three RNA polymerases (I, II and III). The RNA polymerase III (Pol III) has 17 subunits, one of which exists in two alternative forms: RPC32α and RPC32β. Only one of the two forms can be integrated into the enzymes, thus generating either Pol IIIα or Pol IIIβ. While RPC32β is found in all somatic cells, RPC32α is expressed in stem cells and tumor cells. To date nothing is known of their respective roles. Breast cancer is one of the major public health problems, as it is the most common cancer in women. Several types of breast cancers are distinguished, according to the presence or absence of hormonal receptors. Cancers that test negative for estrogen receptors, progesterone receptors and that do not overexpress the human epidermal growth factor receptor 2, are called triple-negative breast cancers. They tend to have a poor prognosis, due to the aggressive nature of the cancer and the lack of targeted therapies. To study the role of RPC32α, a tumor model needed to be identified. In collaboration with Jean-Paul Feugeas (INSERM UMR 1098) a transcriptomic study was performed on 2627 clinical breast tissue samples. The study showed that RPC32α was overexpressed in triplenegative breast cancer, whereas RPC32β was overexpressed in normal tissue. A study on six breast cancer cell lines and one non-tumorigenic line confirmed the results of the transcriptomic study. The breast cancer model was thus validated. A characterization of different breast cancer cell lines showed that other Pol III subunits were not overexpressed in triple-negative breast cancer. The overexpression of RPC32α was therefore not a mere consequence of a Pol III hyperactivity. An analysis of the transcripts synthesized by Pol III showed that overall the Pol III transcript levels were elevated in triplenegative breast cancer compared to other breast cancer subtypes. In order to study the role of RPC32α in tumorigenesis, several RPC32α knock-out cell lines were created using CRISPR-Cas9. The loss of RPC32α did not induce an increase in transcription of the RNAs of RPC32α or RPC32β. This shows that no feed-back loop exists for RPC32α and that the two homologues are not co-regulated. Various Pol III transcripts showed decreased expression levels in the knock-out cell lines. Yet not all transcripts were reduced in the absence of RPC32α. This indicates that some sort of transcription specificity must exist for Pol IIIα and Pol IIIβ. The knock-out cell lines did not show any alterations in their phenotype or growth rates. However, in soft agar assays the knock-out cell lines produced 85% less colonies than the mother cell line. This proves that RPC32α is necessary for tumorigenic growth in vitro. To find out if RPC32α was also necessary for tumorigenic growth in vivo, knock-out and wild type cells were injected into mice. The mice grafted with knock-out cells showed a slowed onset of tumor growth. After six weeks, the mice injected with knock-out cells had tumors half the size of the mice injected with wild type cells. The primary tumor was ablated and mice were tracked for metastasis. Four weeks later, mice injected with RPC32α knock-out cells had 100 times less metastasis than the control group. These results show that RPC32α is necessary for tumorigenic growth in vitro and in vivo. The protein seems also to be implicated in the formation of metastasis, which are one of the greatest problems in cancer treatment today.
12

Cytotoxic Activity of Sphingosine-1-Phosphate against Human Triple-negative/ Basal-like Breast Cancer

2016 January 1900 (has links)
Breast cancer is one of the most common malignancy diagnosed in women and is the primary cause of cancer-related deaths in women worldwide. It is a heterogeneous group of diseases that have a different response, prognosis, and clinical outcomes. Estrogen, progesterone and HER2 negative breast cancer, known as triple negative breast cancer (TNBC), does not respond to hormonal therapy. Basal-like breast cancer (BLBC) has shorter overall survival rate among other subtypes. Tumors sharing both TNBC and BLBC are considered less responsive to currently available treatment. Chemoresistance to treatment has been a challenge in cancer biology and force investigation toward developing new targeted therapies, which selectively target specific subtypes. Sphingolipid metabolites have an important physiological role in determining cell fate. Sphingolipid metabolites, ceramide, sphingosine, and sphingosine-1-phosphate (S1P), are implicated in cancer. S1P exerts its functions via extracellular and intracellular targets. S1P synthesized inside the cell is exported outside and binds to G-protein coupled receptors, the sphingosine-1-phosphate receptors 1-5 (S1PR1-5). Although the intracellular function is not well defined, its suggested intracellular S1P promotes cell apoptosis. The S1P pathway has received great attention recently due its function in cell survival and death. This effect was reported to be concentration dependent. In this research, I focused on S1P effect on nine TNBC/BLBC cell lines. I examined the in-vitro effects of S1P on apoptosis, proliferation, and cytotoxicity in triple negative/ basal-like breast cancer cell lines. Moreover, I studied the co-administration of S1P with currently used chemotherapeutic agents in these cell lines. Data show that S1P can selectively induce cell death in TNBC/BLBC cell lines at a specific concentration. In this research, I found that the mechanism of cell death following treatment with different S1P concentrations was mainly due to apoptosis. Results show that S1P leads to cell shrinkage, rounding and detachment in the nine TNBC/BLBC cell lines. S1P combination with doxorubicin and docetaxel at different concentrations shows no beneficial effect of the combination compared to the chemotherapeuitc agent alone. In some cell lines, the combination showed a protective effect. Further studies are required to determine the mechanism by which S1P induces cell apoptosis, inhibits cell growth, and demonstrates lack of responsiveness in combination studies.
13

Regulation of Phenotypic Plasticity in Triple-Negative Breast Cancer

D'Amato, Nicholas January 2011 (has links)
<p>Breast cancers with a basal-like gene signature are primarily triple-negative, are frequently metastatic, and carry the worst prognosis. Basal-like breast cancers are frequently enriched for markers of breast cancer stem cells as well as markers of epithelial-mesenchymal transition (EMT). While EMT is generally thought to be important in the process of metastasis, direct in vivo evidence of EMT in human disease remains rare. Here we report a novel model of human triple-negative breast cancer, the DKAT cell line, which was isolated from an aggressive, treatment-resistant triple-negative breast cancer that demonstrated morphological and biochemical evidence of epithelial-mesenchymal plasticity in the patient.</p><p>In culture, the DKAT cell line exhibits a basal epithelial phenotype under normal culture conditions in serum-free MEGM, and can undergo a reversible EMT in response to serum-containing media, a unique property among the breast cancer cell lines we tested. This EMT is marked by increased expression of the transcription factor Zeb1, and Zeb1 is required for the enhanced migratory ability of DKAT cells in the mesenchymal state. Additionally, we find that expression of the cytokine IL-6 is dramatically increased in mesenchymal DAKT cells, and blocking IL-6 signaling reduces expression of Zeb1. DKAT cells also express progenitor-cell markers, and single DKAT cells are able to generate tumorspheres containing both epithelial and mesenchymal cell types. In vivo, as few as ten DKAT cells are capable of forming xenograft tumors which display a range of epithelial and mesenchymal phenotypes. Finally, we also show evidence of vimentin expression in mammary epithelial cell clusters from asymptomatic women at high risk for breast cancer, suggesting that changes characteristic of epithelial-mesenchymal plasticity may be inherent to some breast cancers from their earliest stages.</p><p>Our results provide evidence that the aggressive behavior of a subset of triple-negative breast cancers is driven by inherent epithelial-mesenchymal plasticity. The novel finding that IL-6 regulates Zeb1 expression adds further rationale for the development of anti-IL-6 therapeutics, which will have the potential to target pathways at the intersection of metastasis and tumor recurrence. The DKAT cell line represents a novel model for further study of the molecular mechanisms that regulate plasticity in highly aggressive triple-negative breast cancers. An improved understanding of the pathways that are critical for this plasticity may lead to improved treatment options for highly aggressive and deadly breast cancers.</p> / Dissertation
14

Molecular underpinnings of tumor suppression of colon and triple-negative breast cancers

Wong, Chen Khuan 21 February 2019 (has links)
Colon and breast cancers are amongst the leading causes of cancer deaths in the United States, mostly attributed to metastasis and resistance to therapy. Hence, there is a critical need to identify novel biomarkers for effective prognosis and to design targeted therapies to combat the metastatic diseases. Loss of heterozygosity (LOH) at chromosome 18q and inactivation of the target gene, SMAD4, corresponds to resistance to the common chemotherapeutic agent, 5-fluorouracil (5-FU), in colon cancer. Our examination of the therapeutic resistance phenomenon in SMAD4-negative colon cancer cells with the three common agents revealed significant resistance to both 5-FU and irinotecan but not to oxaliplatin. We also followed up with the earlier findings from our group, which suggested that SMAD4 might interact with metastasis-promoting factors to suppress metastatic progression and render sensitivity to chemotherapy. Co-immunoprecipitation and mass spectrometry analysis revealed that SMAD4 interacts with and inhibits RICTOR, a component of mTORC2 that activates oncogenic AKT via phosphorylation at Serine 473. Overexpression of SMAD4, depletion of RICTOR, or inhibition of AKT signaling restores sensitivity to irinotecan in SMAD4-negative colon cancer cells in vitro. Furthermore, as expected pharmacological inhibition of AKT sensitizes these cells to irinotecan in vivo. Interestingly, high RICTOR/AKT expression correlates with worse survival in colon cancer patients, suggesting them as novel prognostic biomarkers and therapeutic targets. On the other hand, triple-negative breast cancer (TNBC) is the most aggressive form of breast cancer due to lack of effective targeted therapies. Using miRNA expression profiling of a model for epithelial-mesenchymal transition in TNBC, we found suppression of miR-4417 during the progression from non-malignant to malignant stage. Furthermore, localization of miR-4417 to chromosome 1p36, a region corresponding to high frequency of LOH in multiple cancers and low-level expression in TNBC patients associated with poor overall survival is consistent with its likely role as a tumor suppressor. Interestingly, we found that overexpression of miR-4417 is sufficient to inhibit migration and tumorigenecity of TNBC cells in vitro. Overall, our findings suggest miR-4417 exerts a tumor-suppressive effect and could serve as a novel prognostic biomarker and therapeutic tool against TNBC. / 2021-02-20T00:00:00Z
15

Ciblage de la protéase cathepsine D par des anticorps monoclonaux humains pour la thérapie des cancers du sein triple-négatifs / Targeting the protease cathepsin D with monoclonal human antibodies for triple-negative breast cancer therapy

Mansouri, Hanane 26 October 2018 (has links)
Les cancers du sein triple-négatif (TNBC) (RE-, RP-, HER2) représentent 15% des cas de cancer du sein. Les patientes atteintes de TNBC sont traitées uniquement par chimiothérapie. A l’heure actuelle, il n’existe aucune thérapie ciblée efficace. Malgré une chimiosensibilité initiale, les rechutes sont fréquentes. Ainsi de nouveaux traitements sont nécessaires pour soigner ces patientes. Dans le cancer du sein, l’aspartyl protéase cathepsine D (cath-D), un marqueur de mauvais pronostic, est surexprimée par les cellules cancéreuses et est hyper-secrétée dans le microenvironnement tumoral. La cath-D stimule la prolifération des cellules cancéreuses, la croissance invasive des fibroblastes, la croissance tumorale, l’angiogenèse tumorale et la formation des métastases. Différentes études ont mis en exergue le rôle oncogénique de la cath-D extracellulaire dans le cancer du sein, suggérant qu’elle serait une cible thérapeutique d’intérêt. Afin de neutraliser sélectivement la forme sécrétée de la cath-D, le laboratoire a généré des anticorps humains IgG1 dirigés contre la cath-D par un crible de phage display (International patent N° PCT/EP2016/061454).Les principaux objectifs de ma thèse ont été i) de valider la cath-D comme une cible extracellulaire d’intérêt pour les patientes atteintes de TNBC, ii) d’évaluer les effets thérapeutiques et iii) de caractériser les mécanismes d’action des anticorps humains anti-cath-D.Nous avons montré que des niveaux élevés d'ARNm de CTSD sont corrélés à une survie sans récidive plus courte. Par analyse protéomique et étude immunohistochimique anti-cath-D réalisée sur Tissue Micro-Array, nous avons observé que la cath-D extracellulaire est détectée dans le microenvironnement tumoral des TNBC contrairement au tissu mammaire normal. Nos résultats mettent ainsi en exergue que la cath-D serait un biomarqueur tumoral extracellulaire, suggérant que les patientes atteintes de TBNC pourraient bénéficier d’une thérapie par des anticorps anti-cath-D. Par des analyses de SPECT-CT (Single Photon Emission Computed Tomography) et de biodistribution, nous avons validé que les anticorps humains anti-cath-D, F1 et E2, s’accumulent dans les xénogreffes de la lignée TNBC MDA-MB-231 chez la souris athymique. Les anticorps F1 et E2 inhibent la croissance tumorale des xénogreffes MDA-MB-231 et améliorent la survie des souris athymiques. Notre meilleur anticorps anti-cath-D, F1, inhibe également la croissance tumorale de deux lignées de TNBC PDX (patient-tumor derived xenografts). Au niveau mécanistique, l’anticorps F1 module le microenvironnement immunitaire dans les tumeurs issues des xénogreffes MDA-MB-231. L’ensemble de nos résultats suggèrent qu’une immunothérapie avec des anticorps humains anti-cath-D pourrait être une nouvelle approche thérapeutique pour les patientes atteintes de TNBC. / Triple-negative breast cancer (TNBC) accounts for 15-20% of all breast cancer cases, and chemotherapy is the only available treatment. Thus identification of new therapeutic targets is required to improve TNBC outcome. In breast cancer, the aspartic protease cathepsin D (cath-D) is a marker of poor prognosis associated with metastatic risk. This protease is overexpressed by breast cancer cells and is abnormally hypersecreted into the tumor microenvironment. Cath-D affects both cancer and stromal cells in the breast tumor microenvironment by increasing the proliferation of breast cancer cells, fibroblast invasive outgrowth, tumor growth and angiogenesis, and metastasis formation. Many studies indicated that extracellular cath-D displays oncogenic activities, suggesting it could represent a novel therapeutic target in TNBC. In order to block its oncogenic actions, the laboratory generated human IgG1 antibodies against extracellular cath-D by phage display (International patent N° PCT/EP2016/061454). The aims of my PhD project was i) to validate the potential value of cath-D as a tumor-specific extracellular target in TNBC, ii) to evaluate the therapeutic activity, and iii) to characterize the mechanisms of action of these anti-cath-D human antibodies.We showed that elevated CTSD mRNA levels correlated with shorter recurrence-free survival. Using proteomics analysis and anti-cath-D immunohistochemistry performed on Tissue Micro-Array, we observed that extracellular cath-D was detected in the tumor microenvironment of TNBC, but not in matched normal breast stroma samples. Our results thus indicate that cath-D is a tumor cell-associated extracellular biomarker and strongly suggest that it could be a good candidate for antibody-based therapy in TNBC. We found that anti-cath-D human antibodies, F1 and E2, accumulated in TNBC MDA-MB-231 tumor xenografts in athymic mice by SPECT-CT (Single Photon Emission Computed Tomography) and biodistribution analysis. F1 and E2 antibodies inhibited tumor growth of MDA-MB-231 tumor xenografts and improved mice survival without apparent toxicity. F1, the best antibody candidate, inhibited tumor growth of two TNBC patient-derived xenografts (PDX). Mechanistically, F1 treatment modulates immune tumor microenvironment in the MDA-MB-231 tumor cell xenograft model. Together, our results indicate that antibody-based targeting of cath-D may have therapeutic efficacy for TNBC treatment.
16

Treatment pattern, risk for hospitalization and mortality in elderly patients with triple-negative breast cancer

Nyström, Pontus January 2019 (has links)
Introduction Triple-negative breast cancer (TNBC) has limited treatment options, as chemotherapy is the only systemic therapy. This poses a challenge in the elderly population due to the limited research done, but also the increased risk for adverse events. Aim To describe the treatment pattern, identify the risk of hospitalization and potential risk factors for hospitalization within 1 year from diagnosis, and investigate the causes of death in elderly with TNBC and possible predictors for mortality.MethodsWe performed a registry-based cohort study using the BCBaSe database which links cases of breast cancer from 3 Swedish healthcare regions with socioeconomic factors, hospitalizations and causes of death. Women ≥ 70 years old with non-metastatic TNBC, between 1/1 2007 and 31/12 2012 were chosen (n = 413). Logistic regression and Cox proportional hazards regression analyses were used. Results Age, stage and comorbidities influenced administration of chemotherapy. The risk of hospitalization overall was increased in the group receiving chemotherapy (OR 2.35, 95% CI 1.30 – 4.26) mainly due to toxicities. Chemotherapy use was not associated with either breast cancer-specific (HR 0.95, 95% CI 0.51 – 1.79) or overall survival (HR 0.72, 95% CI 0.44 – 1.18). Stage at diagnosis and comorbidities were associated with both breast cancer-specific mortality and overall mortality, whereas age was only associated with overall mortality. Conclusions In elderly TNBC patients, chemotherapy use was associated with increased risk for hospitalization within 1 year from diagnosis without improving breast cancer-specific or overall survival. No benefit regarding mortality was seen in those who were administered chemotherapy.
17

Differential regulation of the EMT axis by MEK1/2 and MEK5 in triple-negative breast cancer

January 2016 (has links)
acase@tulane.edu / Triple-negative breast cancer (TNBC) presents a clinical challenge due to the aggressive nature of the disease and a lack of targeted therapies. Constitutive activation of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway has been linked to chemoresistance and metastatic progression through distinct mechanisms, including activation of epithelial-to-mesenchymal transition (EMT) whereby cells adopt a motile and invasive phenotype through loss of epithelial markers, namely Cadherin 1/E-Cadherin (CDH1), and acquisition of mesenchymal markers, such as vimentin (VIM) and Cadherin 2/N-Cadherin (CDH2). While MAPK/ERK1/2 kinase inhibitors (MEKi) have shown promise as antitumor agents in the preclinical setting, application has had limited success clinically. Activation of compensatory signaling, potentially contributing to the emergence of drug resistance, has shifted the therapeutic strategy to combine MEK1/2 inhibitors with agents targeting oncoproteins (RAF) or parallel growth pathways (PI3K). Conventional MAPK family members have been well-characterized in modulation of cellular processes involved in tumor initiation and progression, yet the role of MEK5-ERK5 in cancer biology is not completely understood. Recent studies have highlighted the importance of the MEK5 pathway in metastatic progression of various cancer types, including those of the prostate, colon, bone and breast. Furthermore, elevated levels of ERK5 expression and activity observed in breast carcinomas are linked to worse prognosis in TNBC patients. The purpose of this work is to explore MEK5 regulation of the EMT axis and to evaluate a novel pan-MEK inhibitor on clinically aggressive TNBC cells. Our results show a distinction between the MEK1/2 and MEK5 cascades in maintenance of the mesenchymal phenotype, suggesting that the MEK5 pathway may be necessary and sufficient in EMT regulation while MEK1/2 signaling further sustains the mesenchymal state of TNBC cells. Furthermore, additive effects on MET induction are evident through the inhibition of both MEK1/2 and MEK5. Taken together, these data demonstrate the need for a better understanding of the individual roles of MEK1/2 and MEK5 signaling in breast cancer and provide rationale for combined targeting of these pathways to circumvent compensatory signaling and subsequent therapeutic resistance. / 1 / Van Hoang
18

Epigenetic regulation in triple-negative breast cancer: tools to identify novel microRNA pathways

January 2014 (has links)
Triple-negative breast cancer (TNBC) accounts for 15% of all diagnosed breast cancers nationally and affects African-American women 3 times more likely than any other ethnic group. Locally, African-American women in the New Orleans area see higher incidence of TNBC cases versus African-American women from the rest of the state of Louisiana, which represents an area of heightened public health interest for the metropolitan area. TNBC is a highly metastatic disease, and targeted therapies such as tamoxifen and herceptin are ineffective due to the lack of estrogen receptor (ER) and HER2/neu target expression in TNBC tumors. Chemotherapy remains the only effective drug therapy in TNBC cases. Evaluating new classes of drugs for clinical use against TNBC as well as furthering our understanding of underlying regulatory mechanisms in TNBC is a priority. Pan-deacetylase inhibitors (DACi), like panobinostat, have shown promise in clinical trials as therapies in other cancers. Pre-clinical data of panobinostat treatment in TNBC cell lines published by this lab has been positive so far, exhibiting a reduction in TNBC metastatic potential. DACi can alter multiple signaling pathways and are known to restore dysregulated microRNA (miRNA) expression patterns (miRnome) in cancers. MiRNA are a relatively new class of non-protein coding regulatory biomolecules that exhibit a variety of cancer-related properties, many still unknown in TNBC. Pan-DACi treatment combined with miRnome analysis in TNBC cell lines can be used to identify previously unknown miRNA cancer-related properties in TNBC. The specific aim of this project consists of using DACi treatment on TNBC cell lines in conjunction with miRnome analysis to identify previously undescribed anti-cancer miRNAs and elucidate their cancer-related properties in TNBC while uncovering affected cancer pathways, detecting miRNA targets, and revealing affected downstream components. Initial miRNA expression analysis of MDA-MB-231 TNBC cells treated with panobinostat or trichostatin A versus controls produced a list of potential anti-cancer miRNA candidates for further study. Among them, investigations into miR-203 and miR-335 produced unclear results as these were theorized to have anti-metastatic properties in TNBC yet enhanced cancer properties in our models and assays. Overexpression of mir-215 (a tumor suppressor in other cancers) unexpectedly enhanced tumor growth five fold in SCID mice xenografted with lentivirally-transduced MDA-MB-231 breast cancer cells stably overexpressing miR-215 (231/215+). Further qPCR analysis of 231/215+ cells uncovered upregulation of the breast cancer-associated lncRNA, HOTAIR; the breast cancer-associated miRNA, miR-196a; as well as the entire HOXC cluster in which they reside. This represents a previously unidentified regulatory mechanism of the HOXC cluster in humans. Additionally, miR-200b overexpression in MDA-MB-231 cells induced a change in cell morphology to an epithelial-like phenotype, reduced migration by 50%, and re-expressed the epithelial marker CDH1. This demonstrates a partial reversal of epithelial-mesenchymal transition (EMT), which indicates a reduction in metastatic potential by miR-200b overexpression in MDA-MB-231 cells. Additionally, these cells exhibited increased estrogen receptor alpha and related signaling pathways while also being susceptible to reduced proliferation with the anti-estrogen drug fulvestrant at high doses. Using pan-DACi treatment of TNBC cells to analyze changes in the miRnome for unknown cancer-related miRNA candidates suitable for further investigation in TNBC, we identified miR-215 overexpression in MDA-MB-231 cells as an oncogenic event that enhances tumor growth, cell proliferation, and HOXC cluster transcription while miR-200b is an anti-metastatic miRNA that partially reverses EMT and reduces fulvestrant resistance through re-expression of estrogen receptor signaling. / acase@tulane.edu
19

Effects of PTEN Loss and Activated KRAS Overexpression on Viscoelasticity, Adhesion, and Mechanosensitivity of Breast Epithelial Cells

Linthicum, Will H. 14 June 2019 (has links)
Therapeutics targeting the PI3K (phosphatidylinositol 3-kinase) and the Ras/MAPK (mitogen-activated protein kinases) pathways have potential as non-toxic treatments for triple-negative breast cancer due to their frequent over-activation in several forms of cancer. Interestingly, the PI3K and Ras/MAPK pathways have been shown to incite cancer dormancy behavior individually and tumorigenic behavior in unison when induced in healthy breast epithelial cells (MCF-10A) in vivo. Tumorigenesis and metastasis are heavily reliant on the specific mechanical and adhesive properties of cells, including decreased stiffness, increased mechanosensitivity, and decreased adhesion. However, the describe cellular behaviors are poorly understood for dormant cancer phenotypes. Understanding the mechanical and adhesive behaviors of MCF-10A cells as a function of PI3K and/or Ras/MAPK pathway over-activation further explores the cross-talk enabling unique dormant and tumorigenic characteristics. Cellular viscoelasticity and adhesion were measured for MCF-10A cells with PTEN (phosphatase and tensin homolog) knockout and activated KRAS (Kristen rat sarcoma viral oncogene homolog) overexpression to activate the PI3K and Ras/MAPK pathways respectively with atomic force microscopy. PTEN knockout alone has no observable influence on cell adhesion but resulted in softer cells with less organized cytoskeleton. Activated KRAS overexpression increased cell stiffness and cell adhesion regardless of PTEN expression level. Moreover, the overexpression of activated KRAS enhanced the sensitivity of cells to the substrate stiffness. The findings suggest that the cancer-associated pathways PI3K and Ras/MAPK regulate cell adhesion and mechanics to promote tumor formation and metastasis. More importantly, the results that signify mutations of different molecular pathways associated with cancer dormancy regulate cell mechanics differently suggests that cell stiffness is a biomarker that detects and differentiates different types of dormant cancers.
20

Nuclear Basic Fibroblast Growth Factor Regulation of Triple-­Negative Breast Cancer Dormancy/Recurrence

Li, Shenduo January 2014 (has links)
<p>Chemotherapy remains the only available treatment for triple-negative (TN) breast cancer. Although some TN breast cancers respond initially to neoadjuvant chemotherapy, the majority of patients die within three years of treatment due to recurrent tumor growth. Developing ex vivo models for TN breast cancer recurrence and defining responsible molecules will be crucial to developing effective combination therapies for TN breast cancer patients. We have developed an in vitro model of TN breast cancer dormancy/recurrence. Short-term exposure of tumor cells to chemotherapy at clinically relevant doses enriches for a dormant tumor cell population. Several days after removing chemotherapy, dormant tumor cells regain proliferative ability and establish colonies, resembling tumor recurrence. Tumor cells from "recurrent" colonies exhibit increased chemotherapy resistance, resembling therapy resistance of recurrent tumors in patients. Furthermore, we identify a novel signaling axis [nuclear bFGF/DNA-dependent protein kinase (DNA-PK)] supported by chemotherapy-enriched dormant TN breast cancer cells. This signaling axis drives accelerated DNA repair in chemo-residual TN breast cancer cells. Targeting this axis with either with a bFGF shRNA or DNA-PK small molecule inhibitor blocks recurrent colony formation. Using the Oncomine gene expression database, we found that bFGF expression in tumor samples from TN breast cancer patients predicts five year tumor recurrence following neoadjuvant chemotherapy treatment. Finally, we demonstrate that recurrent tumor cells exhibit increased invasiveness, reflecting the aggressive behavior of recurrent tumors in patients. Collectively, these studies identify a novel signaling axis in TN breast cancer that likely contributes to tumor recurrence and provide molecular targets for developing future therapeutics against TN breast cancer.</p> / Dissertation

Page generated in 0.0518 seconds