Matching Items (5)
Filtering by

Clear all filters

153543-Thumbnail Image.png
Description
The majority of non-small cell lung cancer (NSCLC) patients (70%) are diagnosed with adenocarcinoma versus other histological subtypes. These patients often present with advanced, metastatic disease and frequently relapse after treatment. The tumor suppressor, Liver Kinase B1, is frequently inactivated in adenocarcinomas and loss of function is associated with

The majority of non-small cell lung cancer (NSCLC) patients (70%) are diagnosed with adenocarcinoma versus other histological subtypes. These patients often present with advanced, metastatic disease and frequently relapse after treatment. The tumor suppressor, Liver Kinase B1, is frequently inactivated in adenocarcinomas and loss of function is associated with a highly aggressive, metastatic tumor (1). Identification of the mechanisms deregulated with LKB1 inactivation could yield targeted therapeutic options for adenocarcinoma patients. Re-purposing the immune system to support tumor growth and aid in metastasis has been shown to be a feature in cancer progression (2). Tumor associated macrophages (TAMs) differentiate from monocytes, which are recruited to the tumor microenvironment via secretion of chemotaxic factors by cancer cells. We find that NSCLC cells deficient in LKB1 display increased secretion of C-C motif ligand 2 (CCL2), a chemokine involved in monocyte recruitment. To elucidate the molecular pathway regulating CCL2 up-regulation, we investigated inhibitors of substrates downstream of LKB1 signaling in A549, H23, H2030 and H838 cell lines. Noticeably, BAY-11-7082 (NF-κB inhibitor) reduced CCL2 secretion by an average 92%. We further demonstrate that a CCR2 antagonist and neutralizing CCL2 antibody substantially reduce monocyte migration to NSCLC (H23) cell line conditioned media. Using an in vivo model of NSCLC, we find that LKB1 deleted tumors demonstrate a discernible increase in CCL2 levels compared to normal lung. Moreover, tumors display an increase in the M2:M1 macrophage ratio and increase in tumor associated neutrophil (TAN) infiltrate compared to normal lung. This M2 shift was significantly reduced in mice treated with anti-CCL2 or a CCR2 antagonist and the TAN infiltrate was significantly reduced with the CCR2 antagonist. These data suggest that deregulation of the CCL2/CCR2 signaling axis could play a role in cancer progression in LKB1 deficient tumors.
ContributorsFriel, Jacqueline (Author) / Inge, Landon (Thesis advisor) / Lake, Douglas (Thesis advisor) / Blattman, Joseph (Committee member) / Arizona State University (Publisher)
Created2015
Description
Extracellular vesicles (EVs) represent a heterogeneous population of small vesicles, consisting of a phospholipidic bilayer surrounding a soluble interior cargo. These vesicles play an important role in cellular communication by virtue of their protein, RNA, and lipid content, which can be transferred among cells. Peripheral blood is a rich source

Extracellular vesicles (EVs) represent a heterogeneous population of small vesicles, consisting of a phospholipidic bilayer surrounding a soluble interior cargo. These vesicles play an important role in cellular communication by virtue of their protein, RNA, and lipid content, which can be transferred among cells. Peripheral blood is a rich source of circulating EVs. An analysis of EVs in peripheral blood could provide access to unparalleled amounts of biomarkers of great diagnostic, prognostic as well as therapeutic value. In the current study, a plasma EV enrichment method based on pluronic co-polymer was first established and characterized. Plasma EVs from breast cancer patients were then enriched, profiled and compared to non-cancer controls. Proteins signatures that contributed to the prediction of cancer samples from non-cancer controls were created by a random-forest based cross-validation approach. We found that a large portion of these signatures were related to breast cancer aggression. To verify such findings, KIAA0100, one of the features identified, was chosen for in vitro molecular and cellular studies in the breast cancer cell line MDA-MB-231. We found that KIAA0100 regulates cancer cell aggression in MDA-MB-231 in an anchorage-independent manner and is particularly associated with anoikis resistance through its interaction with HSPA1A. Lastly, plasma EVs contain not only individual proteins, but also numerous molecular complexes. In order to measure millions of proteins, isoforms, and complexes simultaneously, Adaptive Dynamic Artificial Poly-ligand Targeting (ADAPT) platform was applied. ADAPT employs an enriched library of single-stranded oligodeoxynucleotides to profile complex biological samples, thus achieving a deep coverage of system-wide, native biomolecules. Profiling of EVs from breast cancer patients was able to obtain a prediction AUC performance of 0.73 when compared biopsy-positive cancer patient to healthy controls and 0.64 compared to biopsy-negative controls and such performance was not associated with the physical breast condition indicated by BIRAD scores. Taken together, current research demonstrated the potential of profiling plasma EVs in searching for therapeutic targets as well as diagnostic signatures.
ContributorsZhong, Zhenyu (Author) / Spetzler, David (Thesis advisor) / Yan, Hao (Thesis advisor) / Lake, Douglas (Committee member) / Mangone, Marco (Committee member) / Arizona State University (Publisher)
Created2018
157235-Thumbnail Image.png
Description
Quiescin sulfhydryl oxidase 1 (QSOX1) is an enzyme that catalyzes disulfide bond formation by oxidizing two free sulfhydryl groups. QSOX1 consists of a thioredoxin (Trx) and an ERV (essential for respiration and viability)/ALR (augmenter of liver regeneration) domain which each contain CxxC motifs that work to bind to substrates and

Quiescin sulfhydryl oxidase 1 (QSOX1) is an enzyme that catalyzes disulfide bond formation by oxidizing two free sulfhydryl groups. QSOX1 consists of a thioredoxin (Trx) and an ERV (essential for respiration and viability)/ALR (augmenter of liver regeneration) domain which each contain CxxC motifs that work to bind to substrates and shuttle electrons to a flavin adenine dinucleotide (FAD) cofactor that accepts the electrons and reduces molecular oxygen to hydrogen peroxide. Investigation of the role of QSOX1 in cancer progression started when it was found at higher abundance in pancreatic ductal adenocarcinoma (PDA) patient plasma compared to healthy normal donor plasma. Increased expression in QSOX1 has been further identified in breast, lung, kidney, prostate, and other cancers. QSOX1 expression is associated with cell proliferation and invasion in vitro and tumor growth in vivo. Additionally, the enzymatic activity of QSOX1 in the extracellular matrix (ECM) is important for cell invasion in vitro. Small molecule inhibitors of QSOX1 have been shown to have antitumorigenic properties in vitro and in vivo. It was hypothesized that monoclonal antibodies (mAbs) against QSOX1 would inhibit cell invasion in vitro. In this work, mice were immunized with eukaryotic-derived rQSOX1 for generation of hybridomas. Hundreds of hybridoma clones were screened by enzyme-linked immunosorbent assay (ELISA) and a fluorescent QSOX1 activity assay. Multiple rounds of subcloning and screening identified 2F1.14 and 3A10.6 as mAbs of interest. The genes for the variable regions of the antibodies were rescued and sequenced. The sequences were aligned with the variable region sequences of another published αQSOX1 mAb scFv492.1. 2F1.14 inhibits the enzymatic activity of QSOX1 by binding to the active site of QSOX1, which was determined by epitope mapping against mutants of QSOX1 that contained mutations in the active site. 3A10.6 did not appear to inhibit the function of QSOX1 in the activity assay; however, it, along with 2F1.14, suppressed tumor invasion in a 3D invasion model. These findings support the developing idea that QSOX1 is a viable target for cancer treatment because targeted inhibition of QSOX1 extracellularly reduced invasive activity. The mAbs and rQSOX1 variants produced here can serve as tools in furthering the characterization of QSOX1 and its role in cancer.
ContributorsKoelbel, Calvin John (Author) / Lake, Douglas (Thesis advisor) / Chen, Qiang "Shawn" (Committee member) / Ho, Thai (Committee member) / Arizona State University (Publisher)
Created2019
136922-Thumbnail Image.png
Description
A high voltage plasma arc can be created and sustained in air by subjecting the gases to an electric field with high voltage potential, causing ionization. The internal energy of the ionized gases can be transferred to corresponding pressure waves when the matter involved switches between the gaseous and plasma

A high voltage plasma arc can be created and sustained in air by subjecting the gases to an electric field with high voltage potential, causing ionization. The internal energy of the ionized gases can be transferred to corresponding pressure waves when the matter involved switches between the gaseous and plasma states. By pulse-width modulating a transformer driving signal, the transfer of internal electrical energy to resonating pressure waves may be controlled. Audio wave input to the driver signal can then be modulated into the carrier wave and be used to determine the width of each pulse in the plasma, thus reconstructing the audio signal as pressure, or sound waves, as the plasma arc switches on and off. The result will be the audio waveform resonating out of the plasma arc as audible sound, and thus creating a plasma loudspeaker. Theory of operation was tested through construction of a plasma arc speaker, and resultant audio playback was analyzed. This analysis confirmed accurate reproduction of audio signal in audible sound.
ContributorsBoehringer, Brian Thomas (Author) / Roedel, Ronald (Thesis director) / Huffman, James (Committee member) / Barrett, The Honors College (Contributor) / Electrical Engineering Program (Contributor)
Created2014-05
154259-Thumbnail Image.png
Description
Quiescin sulfhydryl oxidase 1 (QSOX1) is a highly conserved disulfide bond-generating enzyme that represents the ancient fusion of two major thiol-disulfide oxidoreductase gene families: thioredoxin and ERV. QSOX1 was first linked with cancer after being identified as overexpressed in pancreatic ductal adenocarcinoma (but not in adjacent normal ductal epithelia, infiltrating

Quiescin sulfhydryl oxidase 1 (QSOX1) is a highly conserved disulfide bond-generating enzyme that represents the ancient fusion of two major thiol-disulfide oxidoreductase gene families: thioredoxin and ERV. QSOX1 was first linked with cancer after being identified as overexpressed in pancreatic ductal adenocarcinoma (but not in adjacent normal ductal epithelia, infiltrating lymphocytes, or chronic pancreatitis). QSOX1 overexpression has been confirmed in a number of other histological tumor types, such as breast, lung, kidney, prostate, and others. Expression of QSOX1 supports a proliferative and invasive phenotype in tumor cells, and its enzymatic activity is critical for promoting an invasive phenotype. An in vivo tumor growth study utilizing the pancreatic tumor cell line MIAPaCa-2 containing a QSOX1-silencing shRNA construct revealed that QSOX1 expression supports a proliferative phenotype. These preliminary studies suggest that suppressing the enzymatic activity of QSOX1 could represent a novel therapeutic strategy to inhibit proliferation and invasion of malignant neoplasms.

The goal of this research was to identify and characterize biologically active small molecule inhibitors for QSOX1. Chemical inhibition of QSOX1 enzymatic activity was hypothesized to reduce growth and invasion of tumor cells. Recombinant QSOX1 was screened against libraries of small molecules using an enzymatic activity assay to identify potential QSOX1 inhibitors. Two lead QSOX1 inhibitors were confirmed, 2-phenyl-1, 2-benzisoselenazol-3-one (ebselen), and 3-methoxy-n-[4-(1 pyrrolidinyl)phenyl]benzamide. The biological activity of these compounds is consistent with QSOX1 knockdown in tumor cell lines, reducing growth and invasion in vitro. Treatment of tumor cells with these compounds also resulted in specific ECM defects, a phenotype associated with QSOX1 knockdown. Additionally, these compounds were shown to be active in pancreatic and renal cancer xenografts, reducing tumor growth with daily treatment. For ebselen, the molecular mechanism of inhibition was determined using a combination of biochemical and mass spectrometric techniques. The results obtained in these studies provide proof-of-principle that targeting QSOX1 enzymatic activity with chemical compounds represents a novel potential therapeutic avenue worthy of further investigation in cancer. Additionally, the utility of these small molecules as chemical probes will yield future insight into the general biology of QSOX1, including the identification of novel substrates of QSOX1.
ContributorsHanavan, Paul D (Author) / Lake, Douglas (Thesis advisor) / LaBaer, Joshua (Committee member) / Mangone, Marco (Committee member) / Borges, Chad (Committee member) / Arizona State University (Publisher)
Created2015