Matching Items (6)
Filtering by

Clear all filters

134306-Thumbnail Image.png
Description
The objective of this research was to create a 3D in vitro model to mimic the native breast tumor microenvironment. Polydimethylsiloxane (PDMS) stamps and micromolding techniques were utilized to develop collagen based 3D tumor model. Geometrical design was optimized for the PDMS stamp to compartmentalize the tumor and stromal region

The objective of this research was to create a 3D in vitro model to mimic the native breast tumor microenvironment. Polydimethylsiloxane (PDMS) stamps and micromolding techniques were utilized to develop collagen based 3D tumor model. Geometrical design was optimized for the PDMS stamp to compartmentalize the tumor and stromal region of the 3D model. Addition of tumor and stromal cells into the platform further demonstrated the successful fabrication of the 3D model which will be used to investigate the role of stromal components on tumor growth and progression. Atomic force microscopy will also be utilized to access stromal remodeling during active invasion.
ContributorsAssefa, Eyerusalem Dibaba (Author) / Nikkhah, Mehdi (Thesis director) / Saini, Harpinder (Committee member) / Harrington Bioengineering Program (Contributor, Contributor) / Barrett, The Honors College (Contributor)
Created2017-05
155825-Thumbnail Image.png
Description
Breast cancer is the second leading cause of disease related death in women, contributing over

40,000 fatalities annually. The severe impact of breast cancer can be attributed to a poor

understanding of the mechanisms underlying cancer metastasis. A primary aspect of cancer

metastasis includes the invasion and intravasation that results in cancer cells

Breast cancer is the second leading cause of disease related death in women, contributing over

40,000 fatalities annually. The severe impact of breast cancer can be attributed to a poor

understanding of the mechanisms underlying cancer metastasis. A primary aspect of cancer

metastasis includes the invasion and intravasation that results in cancer cells disseminating from

the primary tumor and colonizing distant organs. However, the integrated study of invasion and

intravasation has proven to be challenging due to the difficulties in establishing a combined tumor

and vascular microenvironments. Compared to traditional in vitro assays, microfluidic models

enable spatial organization of 3D cell-laden and/or acellular matrices to better mimic human

physiology. Thus, microfluidics can be leveraged to model complex steps of metastasis. The

fundamental aim of this thesis was to develop a three-dimensional microfluidic model to study the

mechanism through which breast cancer cells invade the surrounding stroma and intravasate into

outerlying blood vessels, with a primary focus on evaluating cancer cell motility and vascular

function in response to biochemical cues.

A novel concentric three-layer microfluidic device was developed, which allowed for

simultaneous observation of tumor formation, vascular network maturation, and cancer cell

invasion/intravasation. Initially, MDA-MB-231 disseminated from the primary tumor and invaded

the acellular collagen present in the adjacent second layer. The presence of an endothelial network

in the third layer of the device drastically increased cancer cell invasion. Furthermore, by day 6 of

culture, cancer cells could be visually observed intravasating into the vascular network.

Additionally, the effect of tumor cells on the formation of the surrounding microvascular network

within the vascular layer was evaluated. Results indicated that the presence of the tumor

significantly reduced vessel diameter and increased permeability, which correlates with prior in vivo

data. The novel three-layer platform mimicked the in vivo spatial organization of the tumor and its

surrounding vasculature, which enabled investigations of cell-cell interactions during cancer

invasion and intravasation. This approach will provide insight into the cascade of events leading up

to intravasation, which could provide a basis for developing more effective therapeutics.
ContributorsNagaraju, Supriya (Author) / Nikkhah, Mehdi (Thesis advisor) / Ebrahimkhani, Mohammad (Committee member) / Kiani, Samira (Committee member) / Arizona State University (Publisher)
Created2017
168657-Thumbnail Image.png
Description
The TP53 tumor suppressor gene is the most frequently mutated gene in human cancers. In the highly aggressive triple negative breast cancer (TNBC), TP53 is mutated in 80% of cases. TNBC lacks viable drug targets, resulting in a low prognosis (12.2% 5 year survivability rate). As such, the discovery of

The TP53 tumor suppressor gene is the most frequently mutated gene in human cancers. In the highly aggressive triple negative breast cancer (TNBC), TP53 is mutated in 80% of cases. TNBC lacks viable drug targets, resulting in a low prognosis (12.2% 5 year survivability rate). As such, the discovery of druggable targets in TNBC would be beneficial. Mutated p53 protein typically occurs as a missense mutation and often endows cancer cells with gain of function (GOF) properties by dysregulating metabolic pathways. One of these frequently dysregulated pathways is the Hippo/Yes-associated protein-1 (YAP1)/WW Domain Containing Transcription Regulator 1 (TAZ) tumor suppressor pathway. This study therefore analyzed the involvement of the Hippo/YAP1/TAZ pathway in p53-mediated breast cancer cell invasion. From an RNA-seq screen in MCF10A cell lines harboring different TP53 missense mutations, each with a differing invasive phenotype, components of the Hippo pathway were found to correlate with cell invasion. To this end, the active and inactive forms of YAP1 and TAZ were studied. Phosphorylated (inactive) YAP1 and TAZ are retained in the cytoplasm and eventually degraded. Unphosphorylated (active) YAP1 and TAZ translocate to the nucleus to activate TEAD-family transcription factors, inducing cell survival and proliferation genes leading to increased cell invasion. Using quantitative western blot analysis, it was found that inactive TAZ expression was lower in the most invasive cell lines and higher in the least invasive cell lines (p = 0.003). Moreover, the ratio of inactive TAZ protein to total TAZ protein was also shown to be predominantly lower in the invasive cell lines compared to the non-invasive lines (p = 0.04). Finally, active TAZ expression was primarily higher in p53-mutant invasive cell lines and lower in non-invasive p53 mutant cells. Additionally, although YAP1 and TAZ are thought to be functionally redundant, the pattern seen in TAZ was not seen in the YAP1 protein. Taken together, the results demonstrated here suggest that TAZ holds a more dominant role in governing TNBC cell invasion compared to YAP1 and further highlights TAZ as a potential therapeutic target in TNBC.
ContributorsGrief, Dustin (Author) / LaBaer, Joshua (Thesis advisor) / Anderson, Karen (Committee member) / Nikkhah, Mehdi (Committee member) / Arizona State University (Publisher)
Created2022
Description
Breast cancer cell invasion is a highly orchestrated process driven by a myriad of complex microenvironmental stimuli. These complexities make it difficult to isolate and assess the effects of specific parameters including matrix stiffness and tumor architecture on disease progression. In this regard, morphologically accurate tumor models are becoming instrumental

Breast cancer cell invasion is a highly orchestrated process driven by a myriad of complex microenvironmental stimuli. These complexities make it difficult to isolate and assess the effects of specific parameters including matrix stiffness and tumor architecture on disease progression. In this regard, morphologically accurate tumor models are becoming instrumental to perform fundamental studies on cancer cell invasion within well-controlled conditions. In this study, the use of photocrosslinkable hydrogels and a novel, two-step photolithography technique was explored to microengineer a 3D breast tumor model. The microfabrication process presented herein enabled precise localization of the cells and creation of high stiffness constructs adjacent to a low stiffness matrix. To validate the model, breast cancer cell lines (MDA-MB-231, MCF7) and normal mammary epithelial cells (MCF10A) were embedded separately within the tumor model and cellular proliferation, migration and cytoskeletal organization were assessed. Proliferation of metastatic MDA-MB-231 cells was significantly higher than tumorigenic MCF7 and normal mammary MCF10A cells. MDA-MB-231 exhibited highly migratory behavior and invaded the surrounding matrix, whereas MCF7 or MCF10A cells formed clusters that were confined within the micropatterned circular features. F-actin staining revealed unique 3D protrusions in MDA-MB-231 cells as they migrated throughout the surrounding matrix. Alternatively, there were abundance of 3D clusters formed by MCF7 and MCF10A cells. The results revealed that gelatin methacrylate (GelMA) hydrogel, integrated with the two-step photolithography technique, has great promise in creating 3D tumor models with well-defined features and tunable stiffness for detailed studies on cancer cell invasion and drug responsiveness.
ContributorsSam, Feba Susan (Author) / Nikkhah, Mehdi (Thesis advisor) / Ros, Robert (Committee member) / Smith, Barbara (Committee member) / Arizona State University (Publisher)
Created2015
132188-Thumbnail Image.png
Description
Stromal cells play an important role in facilitating disease progression of ductal carcinoma. Cancer associated fibroblasts (CAFs) are an important component of the extracellular matrix (ECM) which constitutes the microenvironment of breast tumor cells. They are known to participate in chemotherapeutic drug resistance by modulating various biochemical and biophysical factors

Stromal cells play an important role in facilitating disease progression of ductal carcinoma. Cancer associated fibroblasts (CAFs) are an important component of the extracellular matrix (ECM) which constitutes the microenvironment of breast tumor cells. They are known to participate in chemotherapeutic drug resistance by modulating various biochemical and biophysical factors that contribute to increased matrix stiffness and collagen I density of the tumor-adjacent stroma. To address these issues in terms of patient treatment, anti-cancer drug regimes have been assembled to incorporate both chemotherapeutic as well as anti-fibrotic drugs to both target tumor cells while also diminishing the elastic modulus of the microenvironment by targeting CAFs. The quantitative assessment of these drug regimes on tumor progression is missing in terms of CAFs role alone.

A high density 3D tumor model was utilized to recapitulate the tumor microenvironment of ductal carcinoma in vitro. The tumor model consisted of MDA-MB-231 tumors seeded within micromolded collagen wells, chemically immobilized upon a surface treated PDMS substrate. CAFs were seeded within the greater collagen structure from which the microwells were formed. The combinatorial effect of anti-fibrotic drug (Tranilast) and chemotherapy drug (Doxorubicin) were studied within 3D co culture conditions. Specifically, the combinatorial effects of the drugs on tumor cell viability, proliferation, and invasion were examined dynamically upon coculture with CAFs using the microengineered model.

The results of the study showed that the combinatorial effects of Tranilast and Doxorubicin significantly decreased the proliferative ability of tumor cells, in addition to significantly decreasing the ability of tumor cells to remain viable and invade their surrounding stroma, compared to control conditions.
ContributorsSilva, Casey Rudolph (Author) / Nikkhah, Mehdi (Thesis director) / Saini, Harpinder (Committee member) / Harrington Bioengineering Program (Contributor, Contributor) / Barrett, The Honors College (Contributor)
Created2019-05
132583-Thumbnail Image.png
Description
Inhibitor of growth factor 4 (ING4) is a tumor suppressor of which low expression has been associated with poor patient survival and aggressive tumor progression in breast cancer. ING4 is characterized as a transcription regulator of inflammatory genes. Among the ING4-regulated genes is CXCL10, a chemokine secreted by endothelial cells

Inhibitor of growth factor 4 (ING4) is a tumor suppressor of which low expression has been associated with poor patient survival and aggressive tumor progression in breast cancer. ING4 is characterized as a transcription regulator of inflammatory genes. Among the ING4-regulated genes is CXCL10, a chemokine secreted by endothelial cells during normal inflammation response, which induces chemotactic migration of immune cells to the site. High expression of CXCL10 has been implicated in aggressive breast cancer, but the mechanism is not well understood. A potential signaling molecule downstream of Cxcl10 is Janus Kinase 2 (Jak2), a kinase activated in normal immune response. Deregulation of Jak2 is associated with metastasis, immune evasion, and tumor progression in breast cancer. Thus, we hypothesized that the Ing4/Cxcl10/Jak2 axis plays a key role in breast cancer progression. We first investigated whether Cxcl10 affected breast cancer cell migration. We also investigated whether Cxcl10-mediated migration is dependent on ING4 expression levels. We utilized genetically engineered MDAmb231 breast cancer cells with a CRISPR/Cas9 ING4-knockout construct or a viral ING4 overexpression construct. We performed Western blot analysis to confirm Ing4 expression. Cell migration was assessed using Boyden Chamber assay with or without exogenous Cxcl10 treatment. The results showed that in the presence of Cxcl10, ING4-deficient cells had a two-fold increase in migration as compared to the vector controls, suggesting Ing4 inhibits Cxcl10-induced migration. These findings support our hypothesis that ING4-deficient tumor cells have increased migration when Cxcl10 signaling is present in breast cancer. These results implicate Ing4 is a key regulator of a chemokine-induced tumor migration. Our future plan includes evaluation of Jak2 as an intermediate signaling molecule in Cxcl10/Ing4 pathway. Therapeutic implications of these findings are targeting Cxcl10 and/or Jak2 may be effective in treating ING4-deficient aggressive breast cancer.
ContributorsArnold, Emily (Author) / Kim, Suwon (Thesis director) / Blattman, Joseph (Thesis director) / Mason, Hugh (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2019-05