Matching Items (39)
Filtering by

Clear all filters

157161-Thumbnail Image.png
Description
Antibodies are naturally occurring proteins that protect a host during infection through direct neutralization and/or recruitment of the innate immune system. Unfortunately, in some infections, antibodies present unique hurdles that must be overcome for a safer and more efficacious antibody-based therapeutic (e.g., antibody dependent viral enhancement (ADE) and inflammatory pathology).

Antibodies are naturally occurring proteins that protect a host during infection through direct neutralization and/or recruitment of the innate immune system. Unfortunately, in some infections, antibodies present unique hurdles that must be overcome for a safer and more efficacious antibody-based therapeutic (e.g., antibody dependent viral enhancement (ADE) and inflammatory pathology). This dissertation describes the utilization of plant expression systems to produce N-glycan specific antibody-based therapeutics for Dengue Virus (DENV) and Chikungunya Virus (CHIKV). The Fc region of an antibody interacts with Fcγ Receptors (FcγRs) on immune cells and components of the innate immune system. Each class of immune cells has a distinct action of neutralization (e.g., antibody dependent cell-mediated cytotoxicity (ADCC) and antibody dependent cell-mediated phagocytosis (ADCP)). Therefore, structural alteration of the Fc region results in novel immune pathways of protection. One approach is to modulate the N-glycosylation in the Fc region of the antibody. Of scientific significance, is the plant’s capacity to express human antibodies with homogenous plant and humanized N-glycosylation (WT and GnGn, respectively). This allows to study how specific glycovariants interact with other components of the immune system to clear an infection, producing a tailor-made antibody for distinct diseases. In the first section, plant-produced glycovariants were explored for reduced interactions with specific FcγRs for the overall reduction in ADE for DENV infections. The results demonstrate a reduction in ADE of our plant-produced monoclonal antibodies in in vitro experiments, which led to a greater survival in vivo of immunodeficient mice challenged with lethal doses of DENV and a sub-lethal dose of DENV in ADE conditions. In the second section, plant-produced glycovariants were explored for increased interaction with specific FcγRs to improve ADCC in the treatment of the highly inflammatory CHIKV. The results demonstrate an increase ADCC activity in in vitro experiments and a reduction in CHIKV-associated inflammation in in vivo mouse models. Overall, the significance of this dissertation is that it can provide a treatment for DENV and CHIKV; but equally importantly, give insight to the role of N-glycosylation in antibody effector functions, which has a broader implication for therapeutic development for other viral infections.
ContributorsHurtado, Jonathan (Author) / Chen, Qiang (Thesis advisor) / Arntzen, Charles (Committee member) / Borges, Chad (Committee member) / Lake, Douglas (Committee member) / Arizona State University (Publisher)
Created2019
153363-Thumbnail Image.png
Description
Osteosarcoma is the most common bone cancer in children and adolescents. Patients with metastatic osteosarcoma are typically refractory to treatment. Numerous lines of evidence suggest that cytotoxic T-lymphocytes (CTL) limit the development of metastatic osteosarcoma. I have investigated the role of Programmed Death Receptor-1 (PD-1) in limiting the efficacy of

Osteosarcoma is the most common bone cancer in children and adolescents. Patients with metastatic osteosarcoma are typically refractory to treatment. Numerous lines of evidence suggest that cytotoxic T-lymphocytes (CTL) limit the development of metastatic osteosarcoma. I have investigated the role of Programmed Death Receptor-1 (PD-1) in limiting the efficacy of immune mediated control of metastatic osteosarcoma. I show that human metastatic, but not primary, osteosarcoma tumors express the ligand for PD-1 (PD-L1) and that tumor infiltrating CTL express PD-1, suggesting this pathway may limit CTL control of metastatic osteosarcoma in patients. PD-L1 is also expressed on the K7M2 osteosarcoma tumor cell line that establishes metastases in mice, and PD-1 is expressed on tumor infiltrating CTL during disease progression. Blockade of PD-1/PD-L1 interactions dramatically improves the function of osteosarcoma-reactive CTL in vitro and in vivo, and results in decreased tumor burden and increased survival in the K7M2 mouse model of metastatic osteosarcoma. My results suggest that blockade of PD-1/PD-L1 interactions in patients with metastatic osteosarcoma should be pursued as a therapeutic strategy. However, PD-1/PD-L1 blockade treated mice still succumb to disease due to selection of PD-L1 mAb resistant tumor cells via up-regulation of other co-inhibitory T cell receptors. Combinational α-CTLA-4 and α-PD-L1 blockade treated mice were able to completely eradicate metastatic osteosarcoma, and generate immunity to disease. These results suggest that blockade of PD-1/PD-L1 interactions in patients with metastatic osteosarcoma, although improves survival, may lead to tumor resistance, requiring combinational immunotherapies to combat and eradicate disease.
ContributorsLussier, Danielle (Author) / Blattman, Joseph N. (Thesis advisor) / Anderson, Karen (Committee member) / Goldstein, Elliott (Committee member) / Lake, Douglas (Committee member) / Arizona State University (Publisher)
Created2015
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
158492-Thumbnail Image.png
Description
Adoptive transfer of T cells engineered to express synthetic antigen-specific T cell receptors (TCRs) has provocative therapeutic applications for treating cancer. However, expressing these synthetic TCRs in a CD4+ T cell line is a challenge. The CD4+ Jurkat T cell line expresses endogenous TCRs that compete for space, accessory proteins,

Adoptive transfer of T cells engineered to express synthetic antigen-specific T cell receptors (TCRs) has provocative therapeutic applications for treating cancer. However, expressing these synthetic TCRs in a CD4+ T cell line is a challenge. The CD4+ Jurkat T cell line expresses endogenous TCRs that compete for space, accessory proteins, and proliferative signaling, and there is the potential for mixed dimer formation between the α and β chains of the endogenous receptor and that of the synthetic cancer-specific TCRs. To prevent hybridization between the receptors and to ensure the binding affinity measured with flow cytometry analysis is between the tetramer and the TCR construct, a CRISPR-Cas9 gene editing pipeline was developed. The guide RNAs (gRNAs) within the complex were designed to target the constant region of the α and β chains, as they are conserved between TCR clonotypes. To minimize further interference and confer cytotoxic capabilities, gRNAs were designed to target the CD4 coreceptor, and the CD8 coreceptor was delivered in a mammalian expression vector. Further, Golden Gate cloning methods were validated in integrating the gRNAs into a CRISPR-compatible mammalian expression vector. These constructs were transfected via electroporation into CD4+ Jurkat T cells to create a CD8+ knockout TCR Jurkat cell line for broadly applicable uses in T cell immunotherapies.
ContributorsHirneise, Gabrielle Rachel (Author) / Anderson, Karen (Thesis advisor) / Mason, Hugh (Committee member) / Lake, Douglas (Committee member) / Arizona State University (Publisher)
Created2020
158783-Thumbnail Image.png
Description
Skeletal muscle injury, whether acute or chronic, is characterized by influxes of pro- and anti-inflammatory cells that coordinate with muscle to precisely control the reparative process. This intricate coordination is facilitated by a signaling feedback loop between satellite cells and extravasated immune cells. Regulation of the cytokines and chemokines that

Skeletal muscle injury, whether acute or chronic, is characterized by influxes of pro- and anti-inflammatory cells that coordinate with muscle to precisely control the reparative process. This intricate coordination is facilitated by a signaling feedback loop between satellite cells and extravasated immune cells. Regulation of the cytokines and chemokines that mediate healthy repair is critical for the overall success of fiber regeneration and thus provides a prospective direction for the development of therapeutics aimed at fine-tuning the local inflammatory response. This work describes (1) the contribution of non-myogenic cells in skeletal muscle regeneration, (2) the role of the transcription factor Mohawk (Mkx) in regulating inflammation following acute muscle injury and the identification of an overarching requirement for Mkx in the establishment of a pro-inflammatory response, and (3) characterization of eosinophils in acute and chronic muscle damage. Mice deficient for Mkx exhibited delayed muscle regeneration, accompanied by impaired clearance of necrotic fibers and smaller regenerated fibers. This diminished regenerative capacity was associated with a reduction in the recruitment of pro-inflammatory macrophages to the site of damage. In culture, Mkx-/- bone marrow-derived macrophages displayed reduced proliferative capacity but retained the ability to polarize in response to a pro-inflammatory stimulus. The necessity of Mkx in mounting a robust immune response was further confirmed by an immunological challenge in which Mkx-/- mice exhibited increased susceptibility to Salmonella enterica serovar Typhimurium infection. Significant downregulation of key cytokine and chemokine expression was identified throughout the course of muscle repair in Mkx-/- mice and represents one mechanism in which Mkx regulates the establishment of an inflammatory response. Previous research discovered that Mkx is highly expressed in eosinophils, a type of innate immune cell that participates in disease-fighting and inflammation, however the role of eosinophils in muscle repair is not well described. This work outlines the contribution of eosinophils in muscle repair following acute and chronic injury. In healthy mice, eosinophils were found to inhibit efficient muscle repair following acute injury. Utilizing the mdx-/-utrn-/- muscular dystrophy mouse model, eosinophil depletion via administration of anti-IL-5 antibody significantly improved diaphragm fiber diameter and increased the survival rate during the course of treatment.
ContributorsLynch, Cherie Alissa (Author) / Rawls, Alan (Thesis advisor) / Wilson-Rawls, Jeanne (Committee member) / Newbern, Jason (Committee member) / Lake, Douglas (Committee member) / Allen, Ronald (Committee member) / Arizona State University (Publisher)
Created2020
158793-Thumbnail Image.png
Description
The human papillomavirus (HPV) is a double-stranded DNA virus responsible for causing upwards of 80% of head and neck cancers in the oropharyngeal region. Current treatments, including surgery, chemotherapy, and/or radiation, are aggressive and elicit toxic effects. HPV is a pathogen that expresses viral-specific oncogenic proteins that play a role

The human papillomavirus (HPV) is a double-stranded DNA virus responsible for causing upwards of 80% of head and neck cancers in the oropharyngeal region. Current treatments, including surgery, chemotherapy, and/or radiation, are aggressive and elicit toxic effects. HPV is a pathogen that expresses viral-specific oncogenic proteins that play a role in cancer progression. These proteins may serve as potential targets for immunotherapeutic applications. Engineered T cell receptor (TCR) therapy may be an advantageous approach for HPV-associated cancers. In TCR therapy, TCRs are modified to express a receptor that is specific to an immunogenic antigen (part of the virus/cancer capable of eliciting an immune response). Since HPV-associated oropharyngeal cancers typically express unique viral proteins, it is important to identify the TCRs capable of recognizing these proteins. Evidence supports that head and neck cancers typically experience high levels of immune cell infiltration and are subsequently associated with increased survival rates. Most of the immune cell infiltrations in HPV+ HNSCC are CD8+ T lymphocytes, drawing attention to their prospective use in cellular immunotherapies. While TCRs are highly specific, the TCR repertoire is extremely diverse; enabling the immune system to fight off numerous pathogens. In project 1, I review approaches to analyzing TCR diversity and explore the use of DNA origami in retrieving paired TCR sequences from a population. The results determine that DNA origami can be used within a monoclonal population but requires further optimization before being applied in a polyclonal setting. In project 2, I investigate HPV-specific T-cell dysfunction; I detect low frequency HPV-specific CD8+ T cells, determine that they are tumor specific, and show that HPV+HNSCC patients exhibit increased epitope-specific levels of CD8+T cell exhaustion. In project 3, I apply methods to expand and isolate TCRαβ sequences derived from donors stimulated with a previously identified HPV epitope. Single-cell analysis provide ten unique TCRαβ pairs with corresponding CDR3 sequences that may serve as therapeutic candidates. This thesis contributes to fundamental immunology by contributing to the knowledge of T cell dysfunction within HPV+HNSCC and further reveals TCR gene usage within an HPV stimulated population, thus identifying potential TCR pairs for adoptive cell therapies.
ContributorsUlrich, Peaches Rebecca (Author) / Anderson, Karen S (Thesis advisor) / Lake, Douglas (Committee member) / Maley, Carlo (Committee member) / Varsani, Arvind (Committee member) / Arizona State University (Publisher)
Created2020
Description

Memory CD8+ T cells protect against secondary viral infections. They develop and maintain exclusively in circulation (e.g. central memory - Tcm) or are excluded from re-circulation (resident memory - Trm). The extracellular ATP receptor P2RX7 promotes both Tcm and Trm generation. High (P2RX7hi) P2RX7-expressing early effector cells show survival, memory

Memory CD8+ T cells protect against secondary viral infections. They develop and maintain exclusively in circulation (e.g. central memory - Tcm) or are excluded from re-circulation (resident memory - Trm). The extracellular ATP receptor P2RX7 promotes both Tcm and Trm generation. High (P2RX7hi) P2RX7-expressing early effector cells show survival, memory and pluripotency genes. Conversely, many terminal effector (TE) and apoptosis genes are upregulated in low (P2RX7lo) P2RX7-expressing cells. Among these genes is the zinc-finger transcriptional repressor Zeb2, which promotes TE differentiation at the expense of the memory cell pool. Given that Zeb2 was higher in P2RX7lo early effector cells, we postulated that Zeb2 ablation would allow P2RX7-deficient CD8+ T cells to skew towards memory subsets. To test this, we used RNP-based CRISPR-Cas9 to knockout Zeb2 in wild type or P2RX7-deficient P14 cells. At the memory timepoint, Zeb2 ablation led to a rescue of the ability of P2RX7-deficient cells to differentiate into the CD62L+ Tcm and CD69hiCD103hi Trm subsets, as well as increase the population of each. Our data suggest that P2RX7 imprints a pro-memory signature that is, to some extent, dependent on the negative regulation of Zeb2.

ContributorsVan Dijk, Sarah (Author) / Holechek, Susan (Thesis director) / Borges da Silvs, Henrique (Committee member) / Barrett, The Honors College (Contributor) / School of Life Sciences (Contributor) / School of Molecular Sciences (Contributor) / School of International Letters and Cultures (Contributor)
Created2021-12
161538-Thumbnail Image.png
Description
Sepsis is a deadly and debilitating condition resulting from a hyperinflammatory response to infection. Most organ systems are severely impacted, including the neurological complications for survivors of sepsis. Sepsis associated encephalopathy (SAE) is characterized by dysregulated molecular pathways of the immune response impinging upon normal central nervous system (CNS) function

Sepsis is a deadly and debilitating condition resulting from a hyperinflammatory response to infection. Most organ systems are severely impacted, including the neurological complications for survivors of sepsis. Sepsis associated encephalopathy (SAE) is characterized by dysregulated molecular pathways of the immune response impinging upon normal central nervous system (CNS) function and ultimately resulting in lasting cognitive and behavioral impairments. Sepsis predominantly occurs in a few neonates but mostly elderly individuals where they are at high risk of sepsis-induced delirium and other neurological implications that may have overlap with neurodegenerative diseases. This study seeks to identify gene candidates that exhibit altered transcriptional expression in tissues between pigs injected with saline control vs lipopolysaccharide (LPS) to model the early inflammatory aspects of the septic response. Specifically, brain frontal cortex was examined to see which genes and pathways are altered at these early stages and could be targeted for further investigation to alter the cognitive/behavioral decline seen in sepsis survivors. This experiment uses a bulk RNA-seq approach on Yorkshire pigs to identify the variance in gene expression profile. Data analysis showed several gene candidates that were downregulated in the brain in response to LPS that point to early endothelial cell disruption, including OCLN (occludin), SLC19A3 (thiamine transporter), and SLC52A3 (riboflavin transporter). Genes that were upregulated in LPS brain samples implicate endothelial cell dysfunction as well as immune/inflammatory alterations, possibly due to alterations in microglia, the primary immune cell of the brain. Several studies are now underway to understand the cellular origin of these transcriptional changes, as well as analyzing the molecular signatures altered in response to sepsis in whole blood and kidney using bulk RNAseq. In conclusion, specific gene candidates were identified as early changes in the septic brain that could be targets to prevent long-term cognitive and behavioral changes in future studies, establishing a baseline panel to interrogate in animal models with the goal of advancing treatments for human patients who experience sepsis.
ContributorsNeill, Ryan (Author) / Fryer, John D (Thesis advisor) / Hogue, Ian (Thesis advisor) / Lake, Douglas (Committee member) / Arizona State University (Publisher)
Created2021
Description
Purinergic receptors play an important role in the response to infectious diseases by sensing damage-associated molecular patterns (DAMPs). The P2X family of purinergic receptors is known to recognize extracellular ATP (eATP) at different affinities. One of these receptors, P2X3, shows high affinity for eATP, but its role in modulating

Purinergic receptors play an important role in the response to infectious diseases by sensing damage-associated molecular patterns (DAMPs). The P2X family of purinergic receptors is known to recognize extracellular ATP (eATP) at different affinities. One of these receptors, P2X3, shows high affinity for eATP, but its role in modulating responses to infectious diseases has not been studied. Using the pulmonary infection model with influenza virus PR8 strain on wild type (WT) and P2RX3-deficient (P2RX3-KO) mice, we aimed to discover the role of P2RX3 in influenza infection in the lungs. We found that there was not a significant difference in the severity of disease in WT and P2RX3-KO mice during the acute phase, but there was more fibrotic tissue visible in P2RX3-KO mice lungs on day 40 post infection (p.i.) using Masson’s trichrome staining. To further investigate these differences, we analyzed myeloid cell populations and flu-specific lymphocytes in the infected lungs. We found that there was a significant decrease in the number of antigen-specific CD4+ T cells in the lungs of P2RX3-KO mice after 7 days p.i. After performing t-SNE (t-distributed stochastic neighbor embedding) analysis on CD4+ T cells of P2RX3-KO and WT mice, we discovered that P2RX3-KO mice had a population of cells which was not present in the WT mice. This population showed high expression of most proteins such as T-bet and BCL6, which is not characteristic of the typical Th1 population induced by influenza virus. Using in vitro activation and differentiation of Th1 CD4+ T cells from WT and P2RX3-KO mice, we found that P2RX3-KO CD4+ T cells had greater expression of markers related to Tfh (T follicular helper cells), such as ICOS and CXCR5, and overall hyperactivation, demonstrating irregular Th1 differentiation. Taken together, these results suggest that P2RX3 may be linked to the maintenance of “healthy” CD4+ T cells and may be important in preventing fibrosis in influenza infection.
ContributorsWhite, Emily (Author) / Florsheim, Esther (Thesis director) / Borges da Silva, Henrique (Committee member) / Barrett, The Honors College (Contributor) / Department of Psychology (Contributor) / School of Life Sciences (Contributor)
Created2024-05