Characterizing The Effects of The Mohawk Gene on The Immune System and Response to LCMV Infection

165142-Thumbnail Image.png
Description

The Mohawk (mkx) gene functions as a transcriptional repressor for tendon morphogenesis during embryonic development. Previous research showed that mkx KO mice overexpressed the osteogenic gene Runx2. Runx2 plays a role in recognition and long-term immune memory. A study showed

The Mohawk (mkx) gene functions as a transcriptional repressor for tendon morphogenesis during embryonic development. Previous research showed that mkx KO mice overexpressed the osteogenic gene Runx2. Runx2 plays a role in recognition and long-term immune memory. A study showed Runx2 KO mice had a significantly lower number of CD8 T cells specific to lymphocytic choriomeningitis virus (LCMV) and CD8 memory precursor T cells. To determine the direct effects of Mohawk expression on the immune system, development, acute response, and immune memory of innate, B and T cells were compared between WT and mkx KO mice after LCMV infection. Paired t-test analyses were performed between KO and WT data. We first found significantly higher numbers of granulocytes and dendritic cells in the periphery but lower numbers of B cells in the bone marrow and T cells in the thymus of KO mice. When analyzing immune response, we observed a significantly high number of activated CD8 T cells that proliferated in the KO mice in response to the infection. Next, we found no difference in cytokine production for TNF and IFNγ which shows Mohawk does not impair acute immune response. Finally, we found no significant difference between WT and KO mice in the CD8 T cells' ability to make an immune memory. In the present study, we found that, fewer immune cells continued their maturation. However, Mohawk expression did not impact their acute response or ability to become memory cells once the T cells matured and became activated. Rather, T cells specific for LCMV were present in higher numbers in mkx KO mice. Further research will study the impact Mohawk has on both B and T cell memory.

Date Created
2022-05
Agent

Initial experiment of Adaptive Therapy to control Breast Cancer

148450-Thumbnail Image.png
Description

Adaptive therapy utilizes competitive interactions between resistant and sensitive cells by keeping some sensitive cells to control tumor burden with the aim of increasing overall survival and time to progression. The use of adaptive therapy to treat breast cancer, ovarian

Adaptive therapy utilizes competitive interactions between resistant and sensitive cells by keeping some sensitive cells to control tumor burden with the aim of increasing overall survival and time to progression. The use of adaptive therapy to treat breast cancer, ovarian cancer, and pancreatic cancer in preclinical models has shown significant results in controlling tumor growth. The purpose of this thesis is to draft a protocol to study adaptive therapy in a preclinical model of breast cancer on MCF7, estrogen receptor-positive, cells that have evolved resistance to fulvestrant and palbociclib (MCF7 R). In this study, we used two protocols: drug dose adjustment and intermittent therapy. The MCF7 R cell lines were injected into the mammary fat pads of 11-month-old NOD/SCID gamma (NSG) mice (18 mice) which were then treated with gemcitabine.<br/>The results of this experiment did not provide complete information because of the short-term treatments. In addition, we saw an increase in the tumor size of a few of the treated mice, which could be due to the metabolism of the drug at that age, or because of the difference in injection times. Therefore, these adaptive therapy protocols on hormone-refractory breast cancer cell lines will be repeated on young, 6-week old mice by injecting the cell lines at the same time for all mice, which helps the results to be more consistent and accurate.

Date Created
2021-05
Agent

Armed Oncolytic Myxoma Viruses that Eliminate Acute Myeloid Leukemia and Multiple Myeloma Cells

158412-Thumbnail Image.png
Description
Novel biological strategies for cancer therapy have recently been able to generate improved anti-tumor effects in the clinic. Of these new advancements, oncolytic virotherapy is a promising strategy through a dual mechanism of oncolysis and stimulation of tumor immunogenicity against

Novel biological strategies for cancer therapy have recently been able to generate improved anti-tumor effects in the clinic. Of these new advancements, oncolytic virotherapy is a promising strategy through a dual mechanism of oncolysis and stimulation of tumor immunogenicity against the target cancer cells. Myxoma virus (MYXV) is an oncolytic poxvirus that has a natural tropism for Leporids, being nonpathogenic in humans and all other known vertebrates. MYXV is able to infect cancer cells due to mutations and defects in many innate signaling pathways, such as those involved in anti-viral responses. While MYXV alone infects and kills many classes of human cancer cells, recombinant techniques allow for the implementation of therapeutic transgenes, which have the potential of ‘arming’ the virus to enhance its potential as an oncolytic virus. The implementation of certain transgenes allows improved cancer cell killing and/or promotion of more robust anti-tumor immune responses. To investigate the potential of immune-inducing transgenes in MYXV, in vitro screening experiments were performed with several single transgene-armed recombinant MYXVs. As recent studies have shown the ability of MYXV to uniquely target malignant human hematopoietic stem cells, the potential of oncolytic MYXV armed with individual immune-enhancing transgenes was investigated through in vitro killing analysis using human acute myeloid leukemia (AML) and multiple myeloma (MM) cell lines. Additionally, in vitro experiments were performed using primary bone marrow (BM) cells obtained from human patients diagnosed with MM. Furthermore, the action of an engineered bispecific killer engager (huBIKE) was investigated through co-culture studies between the CD138 surface marker of target MM cells and the CD16 surface marker of primary effector peripheral blood mononuclear cells (PBMCs), particularly NK cells and neutrophils. In this study, several of the test armed MYXV-infected human AML and MM cell lines resulted in increased cell death compared to unarmed MYXV-infected cells. Additionally, increased killing of CD138+ MM cells from primary human BM samples was observed following infection with huBIKE-armed MYXV relative to infection with unarmed MYXV. Furthermore, analysis of co-culture studies performed suggests enhanced killing of target MM cells via engagement of NK cells with U266 MM cells by huBIKE.
Date Created
2020
Agent

Necroptosis: Role in Poxvirus Pathogenesis and Oncolytic Virotherapy

158400-Thumbnail Image.png
Description
Necroptosis is a pro-inflammatory mechanism of programmed cell death. It has been implicated in many diseases such as inflammatory diseases, neurodegenerative diseases, cancer and during viral infections. The focus of this research work was to establish the relationship between poxvirus

Necroptosis is a pro-inflammatory mechanism of programmed cell death. It has been implicated in many diseases such as inflammatory diseases, neurodegenerative diseases, cancer and during viral infections. The focus of this research work was to establish the relationship between poxvirus pathogenesis and necroptosis, and the translation implications of necroptosis in oncolytic virotherapy. Vaccinia virus (VACV) is the currently used vaccine for smallpox and it has also been developed as a vaccine vector for several pathogens. E3L is one of the key innate immune evasion genes of VACV and it encodes E3 protein composed of dsRNA binding domain in the C-terminus and Z-NA-binding domain (Z-NA BD) in the N terminus. Both domains are necessary for type 1 interferon resistance and pathogenesis. Recently, it has been shown that in in vitro, the N-terminus of E3 is necessary to inhibit necroptosis occurring through the host-encoded cellular proteins RIP3 and Z-NA-binding protein DAI interaction leading to phosphorylation of MLKL, the key executioner step in the pathway. The research work presented here clearly demonstrates that in a mouse model, the N-terminus of VACV E3 is necessary to inhibit necroptosis during pathogenesis in mice. Another poxvirus belonging to the same family as VACV is monkeypox virus (MPXV) and is an emerging human pathogen. MPXV contains a natural truncation in the N-terminus of its E3 homologue, F3. The results indicate that during MPXV infection in mice, pathogenesis was higher only in DAI knockout mice and not in MLKL knockout mice, suggesting that DAI is possibly activating other proteins not leading to necroptosis. The characterization of VACV as an oncolytic virus was carried out with a focus on future clinical trials. In this study, a pan screening was conducted in various cancer cell lines as many cancers downregulate necroptotic proteins. The results reveal that the N-terminal deletion mutant of VACV selectively replicates in cancer cell lines with a deficient necroptotic pathway and thus, can be used as a potential treatment against specific tumors and evidently, provides abundant scope for future studies.
Date Created
2020
Agent

Effects of LCMV Infection on Murine Fetal Development in Immunized Mothers

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

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.
Date Created
2020-05
Agent

Effect of Rexinoids on Inducing Effector T Cell Chemotaxis

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

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.
Date Created
2020-05
Agent

Water as a physiological currency: hydration state impacts immune function, metabolic substrates, and reproductive investment

157036-Thumbnail Image.png
Description
Environmental changes are occurring at an unprecedented rate, and these changes will undoubtedly lead to alterations in resource availability for many organisms. To effectively predict the implications of such changes, it is critical to better understand how organisms have adapted

Environmental changes are occurring at an unprecedented rate, and these changes will undoubtedly lead to alterations in resource availability for many organisms. To effectively predict the implications of such changes, it is critical to better understand how organisms have adapted to coping with seasonally limited resources. The vast majority of previous work has focused on energy balance as the driver of changes in organismal physiology. While energy is clearly a vital currency, other resources can also be limited and impact physiological functions. Water is essential for life as it is the main constituent of cells, tissues, and organs. Yet, water has received little consideration for its role as a currency that impacts physiological functions. Given the importance of water to most major physiological systems, I investigated how water limitations interact with immune function, metabolism, and reproductive investment, an almost entirely unexplored area. Using multiple species and life stages, I demonstrated that dehydrated animals typically have enhanced innate immunity, regardless of whether the dehydration is a result of seasonal water constraints, water deprivation in the lab, or high physiological demand for water. My work contributed greatly to the understanding of immune function dynamics and lays a foundation for the study of hydration immunology as a component of the burgeoning field of ecoimmunology. While a large portion of my dissertation focused on the interaction between water balance and immune function, there are many other physiological processes that may be impacted by water restrictions. Accordingly, I recently expanded the understanding of how reproductive females can alter metabolic substrates to reallocate internal water during times of water scarcity, an important development in our knowledge of reproductive investments. Overall, by thoroughly evaluating implications and responses to water limitations, my dissertation, when combined previous acquired knowledge on food limitation, will enable scientists to better predict the impacts of future climate change, where, in many regions, rainfall events are forecasted to be less reliable, resulting in more frequent drought.
Date Created
2019
Agent

Cloning and Expression of Antigen-Specific T-Cell Receptors

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

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.
Date Created
2019-05
Agent

Armed Oncolytic Myxoma Viruses to Eliminate Acute Myeloid Leukemia and Multiple Myeloma Cells

132423-Thumbnail Image.png
Description
Novel biological strategies for cancer therapy have recently been able to generate antitumor effects in the clinic. Of these new advancements, oncolytic virotherapy seems to be a promising strategy through a dual mechanism of oncolysis and immunogenicity of the host

Novel biological strategies for cancer therapy have recently been able to generate antitumor effects in the clinic. Of these new advancements, oncolytic virotherapy seems to be a promising strategy through a dual mechanism of oncolysis and immunogenicity of the host to the target cells. Myxoma virus (MYXV) is an oncolytic poxvirus that has a natural tropism for European rabbits, being nonpathogenic in humans and all other known vertebrates. MYXV is able to infect cancer cells which, due to mutations, have defects in many signaling pathways, notably pathways involved in antiviral responses. While MYXV alone elicits lysis of cancer cells, recombinant techniques allow for the implementation of transgenes, which have the potential of ‘arming’ the virus to enhance its potential as an oncolytic virus. The implementation of certain transgenes allow for the promotion of robust anti-tumor immune responses. To investigate the potential of immune-inducing transgenes in MYXV, in vitro experiments were performed with several armed recombinant MYXVs as well as unarmed wild-type and rabbit-attenuated MYXV. As recent studies have shown the ability of MYXV to uniquely target malignant human hematopoietic stem cells, the potential of oncolytic MYXV armed with immune-inducing transgenes was investigated through in vitro killing analysis using human acute myeloid leukemia (AML) and multiple myeloma (MM) cell lines. Furthermore, in vitro experiments were also performed using primary bone marrow (BM) cells obtained from human patients diagnosed with MM. In this study, armed MYXV-infected human AML and MM cells resulted in increased cell death relative to unarmed MYXV-infected cells, suggesting enhanced killing via induced mechanisms of cell death from the immune-inducing transgenes. Furthermore, increased killing of primary BM cells with multiple myeloma was seen in armed MYXV-infected primary cells relative to unarmed MYXV-infected primary cells.
Date Created
2019-05
Agent

CXCL10-Induced Migration of Triple-Negative Breast Cancer Cells

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

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.
Date Created
2019-05
Agent