Matching Items (60)
168781-Thumbnail Image.png
Description
Human papillomavirus (HPV) infection has a large burden on society. It is a causal agent of 99.7% of all cervical cancer cases. The prevalence of HPV infection worldwide is high, but the burden of HPV infections lies on less developed regions. Cervical cancer is not associated with immediate symptoms, screening

Human papillomavirus (HPV) infection has a large burden on society. It is a causal agent of 99.7% of all cervical cancer cases. The prevalence of HPV infection worldwide is high, but the burden of HPV infections lies on less developed regions. Cervical cancer is not associated with immediate symptoms, screening methods are needed to detect HPV disease presence before lesions progress to cervical cancer. Protein biomarkers are a growing area of diagnostic medicine and facilitate the detection of disease at an early and treatable stage. Technologies for healthcare diagnostics often require laboratory space or expensive instrumentation, which are not feasible for point of care applications. In order for clinical diagnostics to advance in developing countries, low cost, rapid, portable, and easy to use point of care diagnostic tests are needed. The project adapts the Enzyme Linked Immunosorbent Assays (ELISA) and Nucleic Acid-Programmable Protein Array (NAPPA) to a proof of concept assay for use in magnetic bead based microfluidics. The biomarker used for analyte detection was E7, as a strong correlation has been found between presence of E7 antibodies and development of advanced cervical cancer. It is demonstrated that magnetic microfluidic assay design for rapid detection of antibodies is amenable to fluorescence detection in point of care settings. The data demonstrates that the microfluidic assay is rapid, low-cost, specific, and relevant to serology detection. The assay detects antibody responses to analytes with the point of care reader system and is realized in an on chip capacity. With the integration of anti-GST capture antibodies conjugated to the magnetic beads in the microfluidic system, many analytes can be detected without large changes to the existing assay structure, which gives the ability to adapt the system to analytes of interest rapidly.
ContributorsSnow, Kylie (Author) / Anderson, Karen (Thesis advisor) / Blain Christen, Jennifer (Committee member) / Lake, Douglas (Committee member) / Arizona State University (Publisher)
Created2022
190819-Thumbnail Image.png
Description
Human Papillomavirus (HPV) is the most commonly transmitted STI and isresponsible for an estimated 5% of cancer cases worldwide. HPV infection is implicated in 70% of cervical cancer incidence and is also responsible for a variety of oropharyngeal and anogenital cancers. While vaccination has greatly reduced the cervical cancer burden in developed countries,

Human Papillomavirus (HPV) is the most commonly transmitted STI and isresponsible for an estimated 5% of cancer cases worldwide. HPV infection is implicated in 70% of cervical cancer incidence and is also responsible for a variety of oropharyngeal and anogenital cancers. While vaccination has greatly reduced the cervical cancer burden in developed countries, HPV infection remains high in developing countries due to high cost and poor access to healthcare. Several studies have highlighted the presence of anti-HPV antibodies following infection and their potential use as biomarkers for developing novel screening methods. Progression from initial infection to cancer is slow, thus presenting an opportunity for effective screening programs. Biomarker screening is an important area of cancer detection and Lateral Flow Assays (LFA) are a low cost, easy to use alternative to other screening methods that require extensive training and laboratory space. Therefore, this project proposes as a hypothesis that the development of an LFA screening for HPV specific IgG can provide clinically relevant data for the early detection of cervical dysplasia. This project adapts an LFA in a multiplexed format for fluorescence-based serologic detection of HPV specific IgG in patient plasma.
ContributorsJohns, William (Author) / Anderson, Karen (Thesis advisor) / Lake, Douglas (Committee member) / Hogue, Brenda (Committee member) / Arizona State University (Publisher)
Created2023
190893-Thumbnail Image.png
Description
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative pathogen of Coronavirus Disease 2019 (COVID-19). Successful vaccination aims to elicit neutralizing antibodies (NAbs) which inhibit viral infection. Traditional NAb quantification methods (neutralization assays) are labor-intensive and expensive, with limited practicality for routine use (e.g. monitoring vaccination response). Thus, a rapid

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative pathogen of Coronavirus Disease 2019 (COVID-19). Successful vaccination aims to elicit neutralizing antibodies (NAbs) which inhibit viral infection. Traditional NAb quantification methods (neutralization assays) are labor-intensive and expensive, with limited practicality for routine use (e.g. monitoring vaccination response). Thus, a rapid (10-minute) lateral flow assay (LFA) for quantification of SARS-CoV-2 NAbs was developed. Using the NAb LFA, an 18-month longitudinal study assessing monthly NAb titers was conducted in a cohort of over 500 COVID-19 mRNA vaccine recipients. Three NAb response groups were identified: vaccine strong responders (VSRs), moderate responders (VMRs), and poor responders (VPRs). VSRs generated high and durable NAb titers. VMRs initially generated high NAb titers but showed more rapid waning with time post-vaccination. Finally, VPRs rarely generated NAb titers ≥1:160, even after 3rd dose. Although strong humoral responses correlate with vaccine effectiveness, viral-specific CD4+ and CD8+ T cells are critical for long-term protection. Discordant phenotypes of viral-specific CD8+ and CD4+CXCR5+ T follicular helper (cTfh) cells have recently been associated with differential NAb responses. The second portion of this dissertation was to investigate whether/how SARS-CoV-2 T cell responses differ in individuals with impaired NAb titers following mRNA vaccination. Thus, phenotypic and functional characterization of T cell activation across NAb response groups was conducted. It was hypothesized that VPRs would exhibit discordant SARS-CoV-2 T cell activation and altered cTfh phenotypes. Peripheral blood mononuclear cells were isolated from VPRs, VMRs, VSRs, naturally infected, and normal donors. SARS-CoV-2 responsive T cells were characterized using in vitro activation induced marker assays, multicolor flow cytometry, and multiplex cytokine analysis. Further, CXCR5+ cTfh were examined for chemokine receptor expression (CCR6 and CXCR3). Results demonstrated that despite differential NAb responses, activation of SARS-CoV-2 responsive CD4+ and CD8+ T cells was comparable across NAb groups. However, double-positive CD4+CD8+, CD8low, and activated CD4+CXCR5+CCR6-CXCR3+ (Tfh1-like) T cells were expanded in VPRs compared to VMR and VSRs. Interestingly, a unique population of CD8+CXCR5+ T cells was also expanded in VPRs. These novel findings may aid in identification of individuals with impaired or altered immune responses to COVID-19 mRNA vaccination.
ContributorsRoeder, Alexa Jordan (Author) / Lake, Douglas (Thesis advisor) / McFadden, Grant (Committee member) / Borges Florsheim, Esther (Committee member) / Chang, Yung (Committee member) / Rahman, Masmudur (Committee member) / Arizona State University (Publisher)
Created2023
190960-Thumbnail Image.png
Description
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic, declared in March 2020 resulted in an unprecedented scientific effort that led to the deployment in less than a year of several vaccines to prevent severe disease, hospitalizations, and death from coronavirus disease 2019 (COVID-19). Most vaccine models focus on the

The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic, declared in March 2020 resulted in an unprecedented scientific effort that led to the deployment in less than a year of several vaccines to prevent severe disease, hospitalizations, and death from coronavirus disease 2019 (COVID-19). Most vaccine models focus on the production of neutralizing antibodies against the spike (S) to prevent infection. As the virus evolves, new variants emerge that evade neutralizing antibodies produced by natural infection and vaccination, while memory T cell responses are long-lasting and resilient to most of the changes found in variants of concern (VOC). Several lines of evidence support the study of T cell-mediated immunity in SARS-CoV-2 infections. First, T cell reactivity against SARS-CoV-2 is found in both (cluster of differentiation) CD4+ and CD8+ T cell compartments in asymptomatic, mild, and severe recovered COVID-19 patients. Second, an early and stronger CD8+ T cell response correlates with less severe COVID-19 disease [1-4]. Third, both CD4+ and CD8+ T cells that are reactive to SARS-CoV-2 viral antigens are found in healthy unexposed individuals suggesting that cross-reactive and conserved epitopes may be protective against infection. The current study is focused on the T cell-mediated response, with special attention to conserved, non-spike-cross-reactive epitopes that may be protective against SARS-CoV-2. The first chapter reviews the importance of epitope prediction in understanding the T cell-mediated responses to a pathogen. The second chapter centers on the validation of SARS-CoV-2 CD8+ T cell predicted peptides to find conserved, immunodominant, and immunoprevalent epitopes that can be incorporated into the next generation of vaccines against severe COVID-19 disease. The third chapter explores pre-existing immunity to SARS-CoV-2 in a pre-pandemic cohort and finds two highly immunogenic epitopes that are conserved among human common cold coronaviruses (HCoVs). To end, the fourth chapter explores the concept of T cell receptor (TCR) cross-reactivity by isolating SARS-CoV-2-reactive TCRs to elucidate the mechanisms of cross-reactivity to SARS-CoV-2 and other human coronaviruses (HCoVs).
ContributorsCarmona, Jacqueline (Author) / Anderson, Karen S (Thesis advisor) / Lake, Douglas (Thesis advisor) / Maley, Carlo (Committee member) / Mangone, Marco (Committee member) / LaBaer, Joshua (Committee member) / Arizona State University (Publisher)
Created2023
128421-Thumbnail Image.png
Description

Clear cell renal cell carcinomas (ccRCCs) harbor frequent mutations in epigenetic modifiers including SETD2, the H3K36me3 writer. We profiled DNA methylation (5mC) across the genome in cell line-based models of SETD2 inactivation and SETD2 mutant primary tumors because 5mC has been linked to H3K36me3 and is therapeutically targetable. SETD2 depleted

Clear cell renal cell carcinomas (ccRCCs) harbor frequent mutations in epigenetic modifiers including SETD2, the H3K36me3 writer. We profiled DNA methylation (5mC) across the genome in cell line-based models of SETD2 inactivation and SETD2 mutant primary tumors because 5mC has been linked to H3K36me3 and is therapeutically targetable. SETD2 depleted cell line models (long-term and acute) exhibited a DNA hypermethylation phenotype coinciding with ectopic gains in H3K36me3 centered across intergenic regions adjacent to low expressing genes, which became upregulated upon dysregulation of the epigenome. Poised enhancers of developmental genes were prominent hypermethylation targets. SETD2 mutant primary ccRCCs, papillary renal cell carcinomas, and lung adenocarcinomas all demonstrated a DNA hypermethylation phenotype that segregated tumors by SETD2 genotype and advanced grade. These findings collectively demonstrate that SETD2 mutations drive tumorigenesis by coordinated disruption of the epigenome and transcriptome,and they have important implications for future therapeutic strategies targeting chromatin regulator mutant tumors.

ContributorsTiedmann, Rochelle L. (Author) / Hlady, Ryan A. (Author) / Hanavan, Paul (Author) / Lake, Douglas (Author) / Tibes, Raoul (Author) / Lee, Jeong-Heon (Author) / Choi, Jeong-Hyeon (Author) / Ho, Thai H. (Author) / Robertson, Keith D. (Author) / College of Liberal Arts and Sciences (Contributor)
Created2015-12-05
128420-Thumbnail Image.png
Description

Quiescin sulfhydryl oxidase 1 (QSOX1) is a highly conserved disulfide bond-generating enzyme that is overexpressed in diverse tumor types. Its enzymatic activity promotes the growth and invasion of tumor cells and alters extracellular matrix composition. In a nude mouse-human tumor xenograft model, tumors containing shRNA for QSOX1 grew significantly more

Quiescin sulfhydryl oxidase 1 (QSOX1) is a highly conserved disulfide bond-generating enzyme that is overexpressed in diverse tumor types. Its enzymatic activity promotes the growth and invasion of tumor cells and alters extracellular matrix composition. In a nude mouse-human tumor xenograft model, tumors containing shRNA for QSOX1 grew significantly more slowly than controls, suggesting that QSOX1 supports a proliferative phenotype in vivo. High throughput screening experiments identified ebselen as an in vitro inhibitor of QSOX1 enzymatic activity. Ebselen treatment of pancreatic and renal cancer cell lines stalled tumor growth and inhibited invasion through Matrigel in vitro. Daily oral treatment with ebselen resulted in a 58% reduction in tumor growth in mice bearing human pancreatic tumor xenografts compared to controls. Mass spectrometric analysis of ebselen-treated QSOX1 mechanistically revealed that C165 and C237 of QSOX1 covalently bound to ebselen. This report details the anti-neoplastic properties of ebselen in pancreatic and renal cancer cell lines. The results here offer a “proof-of-principle” that enzymatic inhibition of QSOX1 may have clinical relevancy.

ContributorsHanavan, Paul (Author) / Borges, Chad (Author) / Katchman, Benjamin (Author) / Faigel, Douglas O. (Author) / Ho, Thai H. (Author) / Ma, Chen-Ting (Author) / Sergienko, Eduard A. (Author) / Meurice, Nathalie (Author) / Petit, Joachim L. (Author) / Lake, Douglas (Author) / College of Liberal Arts and Sciences (Contributor)
Created2015-06-01
129040-Thumbnail Image.png
Description

Introduction: Quiescin sulfhydryl oxidase 1 (QSOX1) oxidizes sulfhydryl groups to form disulfide bonds in proteins. Tumor specific expression of QSOX1 has been reported for numerous tumor types. In this study, we investigate QSOX1 as a marker of breast tumor progression and evaluate the role of QSOX1 as it relates to breast

Introduction: Quiescin sulfhydryl oxidase 1 (QSOX1) oxidizes sulfhydryl groups to form disulfide bonds in proteins. Tumor specific expression of QSOX1 has been reported for numerous tumor types. In this study, we investigate QSOX1 as a marker of breast tumor progression and evaluate the role of QSOX1 as it relates to breast tumor growth and metastasis.

Methods: Correlation of QSOX1 expression with breast tumor grade, subtype and estrogen receptor (ER) status was gathered through informatic analysis using the "Gene expression based Outcome for Breast cancer Online" (GOBO) web-based tool. Expression of QSOX1 protein in breast tumors tissue microarray (TMA) and in a panel of breast cancer cell lines was used to confirm our informatics analysis. To investigate malignant cell mechanisms for which QSOX1 might play a key role, we suppressed QSOX1 protein expression using short hairpin (sh) RNA in ER+ Luminal A-like MCF7, ER+ Luminal B-like BT474 and ER- Basal-like BT549 breast cancer cell lines.

Results: GOBO analysis revealed high levels of QSOX1 RNA expression in ER+ subtypes of breast cancer. In addition, Kaplan Meyer analyses revealed QSOX1 RNA as a highly significant predictive marker for both relapse and poor overall survival in Luminal B tumors. We confirmed this finding by evaluation of QSOX1 protein expression in breast tumors and in a panel of breast cancer cell lines. Expression of QSOX1 in breast tumors correlates with increasing tumor grade and high Ki-67 expression. Suppression of QSOX1 protein slowed cell proliferation as well as dramatic inhibition of MCF7, BT474 and BT549 breast tumor cells from invading through Matrigel™ in a modified Boyden chamber assay. Inhibition of invasion could be rescued by the exogenous addition of recombinant QSOX1. Gelatin zymography indicated that QSOX1 plays an important role in the function of MMP-9, a key mediator of breast cancer invasive behavior.

Conclusions: Taken together, our results suggest that QSOX1 is a novel biomarker for risk of relapse and poor survival in Luminal B breast cancer, and has a pro-proliferative and pro-invasive role in malignant progression partly mediated through a decrease in MMP-9 functional activity.

ContributorsKatchman, Benjamin (Author) / Ocal, I. Tolgay (Author) / Cunliffe, Heather E. (Author) / Chang, Yu-Hui (Author) / Hostetter, Galen (Author) / Watanabe, April (Author) / LoBello, Janine (Author) / Lake, Douglas (Author) / College of Liberal Arts and Sciences (Contributor)
Created2013-03-28
128865-Thumbnail Image.png
Description

Pancreatic adenocarcinoma (PAC) is among the most lethal malignancies. While research has implicated multiple genes in disease pathogenesis, identification of therapeutic leads has been difficult and the majority of currently available therapies provide only marginal benefit. To address this issue, our goal was to genomically characterize individual PAC patients to

Pancreatic adenocarcinoma (PAC) is among the most lethal malignancies. While research has implicated multiple genes in disease pathogenesis, identification of therapeutic leads has been difficult and the majority of currently available therapies provide only marginal benefit. To address this issue, our goal was to genomically characterize individual PAC patients to understand the range of aberrations that are occurring in each tumor. Because our understanding of PAC tumorigenesis is limited, evaluation of separate cases may reveal aberrations, that are less common but may provide relevant information on the disease, or that may represent viable therapeutic targets for the patient. We used next generation sequencing to assess global somatic events across 3 PAC patients to characterize each patient and to identify potential targets. This study is the first to report whole genome sequencing (WGS) findings in paired tumor/normal samples collected from 3 separate PAC patients. We generated on average 132 billion mappable bases across all patients using WGS, and identified 142 somatic coding events including point mutations, insertion/deletions, and chromosomal copy number variants. We did not identify any significant somatic translocation events. We also performed RNA sequencing on 2 of these patients' tumors for which tumor RNA was available to evaluate expression changes that may be associated with somatic events, and generated over 100 million mapped reads for each patient. We further performed pathway analysis of all sequencing data to identify processes that may be the most heavily impacted from somatic and expression alterations. As expected, the KRAS signaling pathway was the most heavily impacted pathway (P<0.05), along with tumor-stroma interactions and tumor suppressive pathways. While sequencing of more patients is needed, the high resolution genomic and transcriptomic information we have acquired here provides valuable information on the molecular composition of PAC and helps to establish a foundation for improved therapeutic selection.

ContributorsLiang, Winnie S. (Author) / Craig, David W. (Author) / Carpten, John (Author) / Borad, Mitesh J. (Author) / Demeure, Michael J. (Author) / Weiss, Glen J. (Author) / Izatt, Tyler (Author) / Sinari, Shripad (Author) / Christoforides, Alexis (Author) / Aldrich, Jessica (Author) / Kurdoglu, Ahmet (Author) / Barrett, Michael (Author) / Phillips, Lori (Author) / Benson, Hollie (Author) / Tembe, Waibhav (Author) / Braggio, Esteban (Author) / Kiefer, Jeffrey A. (Author) / Legendre, Christophe (Author) / Posner, Richard (Author) / Hostetter, Galen H. (Author) / Baker, Angela (Author) / Egan, Jan B. (Author) / Han, Haiyong (Author) / Lake, Douglas (Author) / Stites, Edward C. (Author) / Ramanathan, Ramesh K. (Author) / Fonseca, Rafael (Author) / Stewart, A. Keith (Author) / Von Hoff, Daniel (Author) / College of Liberal Arts and Sciences (Contributor)
Created2012-10-10
128791-Thumbnail Image.png
Description

Our previous studies show reduced abundance of the β-subunit of mitochondrial H+-ATP synthase (β-F1-ATPase) in skeletal muscle of obese individuals. The β-F1-ATPase forms the catalytic core of the ATP synthase, and it is critical for ATP production in muscle. The mechanism(s) impairing β-F1-ATPase metabolism in obesity, however, are not completely

Our previous studies show reduced abundance of the β-subunit of mitochondrial H+-ATP synthase (β-F1-ATPase) in skeletal muscle of obese individuals. The β-F1-ATPase forms the catalytic core of the ATP synthase, and it is critical for ATP production in muscle. The mechanism(s) impairing β-F1-ATPase metabolism in obesity, however, are not completely understood. First, we studied total muscle protein synthesis and the translation efficiency of β-F1-ATPase in obese (BMI, 36±1 kg/m2) and lean (BMI, 22±1 kg/m2) subjects. Both total protein synthesis (0.044±0.006 vs 0.066±0.006%·h-1) and translation efficiency of β-F1-ATPase (0.0031±0.0007 vs 0.0073±0.0004) were lower in muscle from the obese subjects when compared to the lean controls (P<0.05). We then evaluated these same responses in a primary cell culture model, and tested the specific hypothesis that circulating non-esterified fatty acids (NEFA) in obesity play a role in the responses observed in humans. The findings on total protein synthesis and translation efficiency of β-F1-ATPase in primary myotubes cultured from a lean subject, and after exposure to NEFA extracted from serum of an obese subject, were similar to those obtained in humans. Among candidate microRNAs (i.e., non-coding RNAs regulating gene expression), we identified miR-127-5p in preventing the production of β-F1-ATPase. Muscle expression of miR-127-5p negatively correlated with β-F1-ATPase protein translation efficiency in humans (r = – 0.6744; P<0.01), and could be modeled in vitro by prolonged exposure of primary myotubes derived from the lean subject to NEFA extracted from the obese subject. On the other hand, locked nucleic acid inhibitor synthesized to target miR-127-5p significantly increased β-F1-ATPase translation efficiency in myotubes (0.6±0.1 vs 1.3±0.3, in control vs exposure to 50 nM inhibitor; P<0.05). Our experiments implicate circulating NEFA in obesity in suppressing muscle protein metabolism, and establish impaired β-F1-ATPase translation as an important consequence of obesity.

ContributorsTran, Lee (Author) / Hanavan, Paul (Author) / Campbell, Latoya (Author) / De Filippis, Elena (Author) / Lake, Douglas (Author) / Coletta, Dawn (Author) / Roust, Lori R. (Author) / Mandarino, Lawrence (Author) / Carroll, Chad C. (Author) / Katsanos, Christos (Author) / College of Health Solutions (Contributor)
Created2016-08-17
129084-Thumbnail Image.png
Description

Background: Recent advances in the treatment of cancer have focused on targeting genomic aberrations with selective therapeutic agents. In rare tumors, where large-scalec linical trials are daunting, this targeted genomic approach offers a new perspective and hope for improved treatments. Cancers of the ampulla of Vater are rare tumors that comprise

Background: Recent advances in the treatment of cancer have focused on targeting genomic aberrations with selective therapeutic agents. In rare tumors, where large-scalec linical trials are daunting, this targeted genomic approach offers a new perspective and hope for improved treatments. Cancers of the ampulla of Vater are rare tumors that comprise only about 0.2% of gastrointestinal cancers. Consequently, they are often treated as either distal common bile duct or pancreatic cancers.

Methods: We analyzed DNA from a resected cancer of the ampulla of Vater and whole blood DNAfrom a 63 year-old man who underwent a pancreaticoduodenectomy by whole genomesequencing, achieving 37× and 40× coverage, respectively. We determined somatic mutations and structural alterations.

Results: We identified relevant aberrations, including deleterious mutations of KRAS and SMAD4 as well as a homozygous focal deletion of the PTEN tumor suppressor gene. These findings suggest that these tumors have a distinct oncogenesis from either common bile duct cancer or pancreatic cancer. Furthermore, this combination of genomic aberrations suggests a therapeutic context for dual mTOR/PI3K inhibition.

Conclusions: Whole genome sequencing can elucidate an oncogenic context and expose potential therapeutic vulnerabilities in rare cancers.

ContributorsDemeure, Michael J. (Author) / Craig, David W. (Author) / Sinari, Shripad (Author) / Moses, Tracy M. (Author) / Christoforides, Alexis (Author) / Dinh, Jennifer (Author) / Izatt, Tyler (Author) / Aldrich, Jessica (Author) / Decker, Ardis (Author) / Baker, Angela (Author) / Cherni, Irene (Author) / Watanabe, April (Author) / Koep, Lawrence (Author) / Lake, Douglas (Author) / Hostetter, Galen (Author) / Trent, Jeffrey M. (Author) / Von Hoff, Daniel D. (Author) / Carpten, John D. (Author) / College of Liberal Arts and Sciences (Contributor)
Created2012-07-04