Matching Items (24)
Filtering by

Clear all filters

Description
Purinergic receptors sense extracellular nucleotide DAMPs such as ATP and adenosine, which are expressed in high concentrations in the tumor microenvironment (TME). A2AR, an adenosine receptor that is expressed on both T cells and tumor cells, promotes immunosuppression. However, the impact of the TME on changes in purinergic receptor expression

Purinergic receptors sense extracellular nucleotide DAMPs such as ATP and adenosine, which are expressed in high concentrations in the tumor microenvironment (TME). A2AR, an adenosine receptor that is expressed on both T cells and tumor cells, promotes immunosuppression. However, the impact of the TME on changes in purinergic receptor expression on CD8 T cells, as well as the overall dynamic between A2AR expression and tumor control, have not been clearly elucidated. Using in vitro co-culture experiments and in vivo murine tumor models, we found that A2AR is significantly upregulated on effector, tumor-infiltrating CD8 T cells. This upregulation was independent of the hypoxia, which we identified via inhibition of HIF1A. We found that this upregulation was partially dependent on CD8 T cell-tumor contact, but independent of cognate antigen recognition, by using transwell co-cultures, as well as combinations of different transgenic lines of CD8 T cells and tumor cells. We confirmed this observation in vivo using transfer of activated OTI cells into B16.OVA-bearing mice. Ultimately, we observed that the upregulation depended on inhibitory receptors such as Tim3 via the antibody blockade of Tim3. Using CRISPR/Cas9-mediated knockout of A2AR on activated CD8 T cells, we found that tumor-bearing mice receiving A2AR knockout CD8 T cells had increased tumor control. Taken together, these results suggest that inhibitory receptor-dependent, TCR-independent signals in the TME promotes upregulation of A2AR on CD8 T cells, leading to impairment of CD8 T cell-mediated tumor control.
ContributorsZhou, Maggie (Author) / Borges da Silva, Henrique (Thesis director) / Borges Florsheim, Esther (Committee member) / Barrett, The Honors College (Contributor) / School of Life Sciences (Contributor) / Economics Program in CLAS (Contributor)
Created2022-12
161211-Thumbnail Image.png
ContributorsVan Dijk, Sarah (Author) / Holechek, Susan (Thesis director) / Borges da Silvs, Henrique (Committee member) / Barrett, The Honors College (Contributor) / School of Life Sciences (Contributor)
Created2021-12
161212-Thumbnail Image.png
ContributorsVan Dijk, Sarah (Author) / Holechek, Susan (Thesis director) / Borges da Silvs, Henrique (Committee member) / Barrett, The Honors College (Contributor) / School of Life Sciences (Contributor)
Created2021-12
132308-Thumbnail Image.png
Description
T cells, a component of the adaptive immune system, play an instrumental role in directing immune responses and direct cell killing in response to pathogens and cancers. T cells recognize and signal through the T cell receptor, a protein heterodimer on the surface of T cells. The T cell receptor

T cells, a component of the adaptive immune system, play an instrumental role in directing immune responses and direct cell killing in response to pathogens and cancers. T cells recognize and signal through the T cell receptor, a protein heterodimer on the surface of T cells. The T cell receptor is a highly variable structure formed via somatic recombination; the structure recognizes peptides presented on the surface of nucleated cells by major histocompatibility complex proteins in a specific receptor-restricted, peptide-restricted manner. This balance between T cell diversity and T cell specificity stands as a barrier to efficacious development of articificial T cell receptors capable of clearing disease. T cell receptors may be tailored to produce pathogen- or cancer-specific immune responses from autologous T cell populations. This necessitates a pipeline for amplification, cloning, and expression of antigen-specific T cell receptors. This study aims to utilize influenza-specific T cell receptor chains from healthy donor T cells to test a model for T cell receptor cloning and expression. This study utilizes Gateway recombination for high-throughput cloning into mammalian expression vectors. This study has successfully amplified and cloned T cell receptor chains from a population of influenza-specific T cells from donor cell transcripts into mammalian cell expression vectors. Additionally, CD8, a coreceptor for the T cell receptor complex, was successfully cloned and inserted into a vector for expression in mammalian cells. Sanger sequencing has confirmed sequences for influenza-specific T cell receptor chains and the CD8 chain. Future application of this project includes expression in mammalian non-T cells to test for efficacy of expression and, ultimately, expression in cytotoxic cells to create lymphocytes capable of antigen-specific recognition and cytolytic killing of cells of interest.
ContributorsVale, Nolan Richard (Author) / Anderson, Karen (Thesis director) / Blattman, Joseph (Committee member) / Department of Psychology (Contributor) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2019-05
132248-Thumbnail Image.png
Description
The amphibian pathogen Ambystoma Tigrinum Virus (ATV) has been an important topic of study within the amphibian community since its discovery. ATV threatens many salamander populations across the US, including those in east-central and southeast Arizona. These populations remain at risk since there are no treatments available. In this thesis,

The amphibian pathogen Ambystoma Tigrinum Virus (ATV) has been an important topic of study within the amphibian community since its discovery. ATV threatens many salamander populations across the US, including those in east-central and southeast Arizona. These populations remain at risk since there are no treatments available. In this thesis, a novel method of inactivation is tested to produce a vaccine with the aim of safely eliciting an immune response within the salamander host. This novel form of inactivation has been tested on several human pathogens but has yet to be used on amphibian pathogens. It has the potential to revolutionize our traditional approach to inactivating viruses. After laser treatment, viral plaque assays suggested that inactivated ATV ceased to grow completely, pointing to the possibility of creating a vaccine. Animal challenge trials were conducted with 60 juvenile Ambystoma tigrinum, but surprisingly there was no protective effect from viral inactivation. Further study is needed to clarify why in vitro and in vivo tests of viral inactivation produced contradictory results.
ContributorsVazquez, Luis Ernesto (Author) / Collins, James (Thesis director) / Tsen, Kong-Thon (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2019-05
131545-Thumbnail Image.png
Description
Immunology, the study of the immune system and its ability to distinguish self from non-self, is a rapidly advancing sector of molecular biology. Cancer, being host derived, provides a difficult challenge for immune cells to distinguish it from normal tissue. The historic treatment of cancer has had three main methods:

Immunology, the study of the immune system and its ability to distinguish self from non-self, is a rapidly advancing sector of molecular biology. Cancer, being host derived, provides a difficult challenge for immune cells to distinguish it from normal tissue. The historic treatment of cancer has had three main methods: radiation, chemotherapy, and surgery (1). Due to recent advancements in understanding the regulatory role of adaptive immunity against cancer, researchers have been attempting to engineer therapies to enhance patients’ immunities against their cancer. Immunotherapies, both passive and active, demonstrate potential for combating many diseases. Passive immunization provides temporary protection against a pathogen, whereas active immunization teaches the patient’s system to respond to the antigen independently, giving life-long immunity. Passive immunization, generally, is a much more expensive method of providing immunity and is commonly used in emergency situations. Anti-venom, for example, uses antibodies grown in lab to neutralize venom. Examples of active immunization are vaccines, which mimic the wild-type pathogen in a way that elicits an immune response, specifically naïve lymphocyte activation and maturation into memory lymphocytes. In terms of cancer therapy, both passive and active immunization are being tested for efficacy (2).
ContributorsMarquardt, Charles Andrew (Author) / Anderson, Karen S. (Thesis director) / Mason, Hugh S. (Committee member) / Lake, Douglas F. (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2020-05
131368-Thumbnail Image.png
Description
The retinoid-X receptor (RXR) can form heterodimers with both the retinoic-acid
receptor (RAR) and vitamin D receptor (VDR). The RXR/RAR dimer is activated by ligand all
trans retinoic acid (ATRA), which culminates in gut-specific effector T cell migration. Similarly,
the VDR/RXR dimer binds 1,25(OH)2D3 to cause skin-specific effector T cell migration.

The retinoid-X receptor (RXR) can form heterodimers with both the retinoic-acid
receptor (RAR) and vitamin D receptor (VDR). The RXR/RAR dimer is activated by ligand all
trans retinoic acid (ATRA), which culminates in gut-specific effector T cell migration. Similarly,
the VDR/RXR dimer binds 1,25(OH)2D3 to cause skin-specific effector T cell migration.
Targeted migration is a potent addition to current vaccines, as it would induce activated T cell
trafficking to appropriate areas of the immune system and ensure optimal stimulation (40).
ATRA, while in use clinically, is limited by toxicity and chemical instability. Rexinoids
are stable, synthetically developed ligands specific for the RXR. We have previously shown that
select rexinoids can enhance upregulation of gut tropic CCR9 receptors on effector T cells.
However, it is important to establish whether these cells can actually migrate, to show the
potential of rexinoids as vaccine adjuvants that can cause gut specific T cell migration.
Additionally, since the RXR is a major contributor to VDR-mediated transcription and
epidermotropism (15), it is worth investigating whether these compounds can also function as
adjuvants that promote migration by increasing expression of skin tropic CCR10 receptors on T
cells.
Prior experiments have demonstrated that select rexinoids can induce gut tropic migration
of CD8+ T cells in an in vitro assay and are comparable in effectiveness to ATRA (7). The effect
of rexinoids on CD4+ T cells is unknown however, so the aim of this project was to determine if
rexinoids can cause gut tropic migration in CD4+ T cells to a similar extent. A secondary aim
was to investigate whether varying concentrations in 1,25-Dihydroxyvitamin D3 can be linked to
increasing CCR10 upregulation on Jurkat CD4+ T cells, with the future aim to combine 1,25
Dihydroxyvitamin D3 with rexinoids.
These hypotheses were tested using murine splenocytes for the migration experiment, and
human Jurkat CD4+ T cells for the vitamin D experiment. Migration was assessed using a
Transwell chemotaxis assay. Our findings support the potential of rexinoids as compounds
capable of causing gut-tropic migration in murine CD4+ T cells in vitro, like ATRA. We did not
observe conclusive evidence that vitamin D3 causes upregulated CCR10 expression, but this
experiment must be repeated with a human primary T cell line.
ContributorsDebray, Hannah Zara (Co-author) / Debray, Hannah (Co-author) / Blattman, Joseph (Thesis director) / Jurutka, Peter (Committee member) / Manhas, Kavita (Committee member) / Department of Psychology (Contributor) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2020-05
131272-Thumbnail Image.png
Description
Despite a continuously growing body of evidence that they are one of the major causes of pregnancy loss, preterm birth, pregnancy complications, and developmental abnormalities leading to high rates of morbidity and mortality, viruses are often overlooked and underestimated as teratogens. The Zika virus epidemic beginning in Brazil in 2015

Despite a continuously growing body of evidence that they are one of the major causes of pregnancy loss, preterm birth, pregnancy complications, and developmental abnormalities leading to high rates of morbidity and mortality, viruses are often overlooked and underestimated as teratogens. The Zika virus epidemic beginning in Brazil in 2015 brought teratogenic viruses into the spotlight for the public health community and popular media, and its infamy may bring about positive motivation and funding for novel treatments and vaccination strategies against it and a variety of other viruses that can lead to severe congenital disease. Lymphocytic choriomeningitis virus (LCMV) is famous in the biomedical community for its historic and continued utility in mouse models of the human immune system, but it is rarely a source of clinical concern in terms of its teratogenic risk to humans, despite its ability to cause consistently severe ocular and neurological abnormalities in cases of congenital infection. Possibilities for a safe and effective LCMV vaccine remain difficult, as the robust immune response typical to LCMV can be either efficiently protective or lethally pathological based on relatively small changes in the host type, viral strain, viral dose, method of infection/immunization, or molecular characteristics of synthetic vaccination. Introducing the immunologically unique state of pregnancy and fetal development to the mix adds complexity to the process. This thesis consists of a literature review of teratogenic viruses as a whole, of LCMV and its complications during pregnancy, of LCMV immunopathology, and of current understanding of vaccination against LCMV and against other teratogenic viruses, as well as a hypothetical experimental design intended to initially bridge the gaps between LCMV vaccinology and LCMV teratogenicity by bringing a vaccine study of LCMV into the context of viral challenge during pregnancy.
ContributorsHarris, Maryl (Author) / Blattman, Joseph (Thesis director) / Scotch, Matthew (Committee member) / Luna, Evelyn (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2020-05
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
132678-Thumbnail Image.png
Description
Cytokines induced by inflammasome has been used for blood cancer treatments, yet these treatments have been less successful in the solid tumor microenvironment. Here precise-morphology DNA origami structures were implemented to accurately test the effect and mechanism of activation in the NLRP3 inflammasome. THP1 WT cells, a macrophage cell line,

Cytokines induced by inflammasome has been used for blood cancer treatments, yet these treatments have been less successful in the solid tumor microenvironment. Here precise-morphology DNA origami structures were implemented to accurately test the effect and mechanism of activation in the NLRP3 inflammasome. THP1 WT cells, a macrophage cell line, were treated with eleven different DNA origami structures. The inflammasome activation of two cytokines, Interleukin 1 beta (IL-1β) and Interferon beta (IFN-β), was measured using HEK Blue IL-1β cells, HEK Blue IFN-β cells, and enzyme linked immunosorbent assay (ELISA). Differences in activation signaling have the potential to provide the characterization required to address the intrinsic complexity of modulating an immune response. It is hoped that DNA origami will help induce more inflammation for solid tumors. The DNA origami was tested in three different volumes: 1 μL, 5 μL, and 10 μL. Overall, the origami that showed promising results were Mg Square. Tetrahedral and P53 block also showed potential but not as well as Mg square. Further testing of more DNA origami structures and testing them in mice are key to the success of targeted cancer immunotherapies in the neoadjuvant setting.
ContributorsGreenwald, Elinor Vera (Co-author) / Ariola, Amanda (Co-author) / Ning, Bo (Thesis director) / Zhang, Fei (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2019-05